1
|
Ma MY, Deng G, Zhu WZ, Sun M, Jiang LY, Li WH, Liu YB, Guo L, Song BL, Zhao X. Defects in CYB5A and CYB5B impact sterol-C4 oxidation in cholesterol biosynthesis and demonstrate regulatory roles of dimethyl sterols. Cell Rep 2024; 43:114912. [PMID: 39489939 DOI: 10.1016/j.celrep.2024.114912] [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: 06/07/2024] [Revised: 09/13/2024] [Accepted: 10/09/2024] [Indexed: 11/05/2024] Open
Abstract
Cytochrome b5 (CYB5) is a hemoprotein crucial for electron transfer to oxygenases. Although microsomal CYB5A is required for sterol C4-demethylation in vitro, cholesterol biosynthesis remains intact in Cyb5a knockout mice. Here, we show that knockout of mitochondrial CYB5B, rather than CYB5A, blocks cholesterol biosynthesis at the sterol-C4 oxidation step in HeLa cells, causing an accumulation of testis meiosis-activating sterol (T-MAS) and dihydro-T-MAS. Surprisingly, liver-specific Cyb5b knockout (L-Cyb5b-/-) mice exhibit normal cholesterol metabolism. Further knockdown of Cyb5a in L-Cyb5b-/- (L-Cyb5b-/-/short hairpin [sh]Cyb5a) mice leads to a marked accumulation of T-MAS and dihydro-T-MAS, indicating that either CYB5A or CYB5B is required for sterol C4-demethylation. The L-Cyb5b-/-/shCyb5a mice are largely normal, with lower sterol regulatory element-binding protein (SREBP)-target gene expression during refeeding and higher liver triglyceride levels while fasting, as T-MAS and dihydro-T-MAS inhibit the SREBP pathway and activate the PPARγ pathway. In summary, CYB5A and CYB5B compensate for sterol C4-demethylation, and T-MAS and dihydro-T-MAS can modulate the SREBP and PPARγ pathways.
Collapse
Affiliation(s)
- Mei-Yan Ma
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430000, China
| | - Gang Deng
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430000, China
| | - Wen-Zhuo Zhu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430000, China
| | - Ming Sun
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430000, China
| | - Lu-Yi Jiang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430000, China
| | - Wei-Hui Li
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430000, China
| | - Yuan-Bin Liu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430000, China
| | - Lin Guo
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430000, China
| | - Bao-Liang Song
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430000, China.
| | - Xiaolu Zhao
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430000, China.
| |
Collapse
|
2
|
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
|
3
|
Clayton BLL, Barbar L, Sapar M, Kalpana K, Rao C, Migliori B, Rusielewicz T, Paull D, Brenner K, Moroziewicz D, Sand IK, Casaccia P, Tesar PJ, Fossati V. Patient iPSC models reveal glia-intrinsic phenotypes in multiple sclerosis. Cell Stem Cell 2024:S1934-5909(24)00288-1. [PMID: 39191254 DOI: 10.1016/j.stem.2024.08.002] [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: 07/27/2023] [Revised: 06/17/2024] [Accepted: 08/05/2024] [Indexed: 08/29/2024]
Abstract
Multiple sclerosis (MS) is an inflammatory and neurodegenerative disease of the central nervous system (CNS), resulting in neurological disability that worsens over time. While progress has been made in defining the immune system's role in MS pathophysiology, the contribution of intrinsic CNS cell dysfunction remains unclear. Here, we generated a collection of induced pluripotent stem cell (iPSC) lines from people with MS spanning diverse clinical subtypes and differentiated them into glia-enriched cultures. Using single-cell transcriptomic profiling and orthogonal analyses, we observed several distinguishing characteristics of MS cultures pointing to glia-intrinsic disease mechanisms. We found that primary progressive MS-derived cultures contained fewer oligodendrocytes. Moreover, MS-derived oligodendrocyte lineage cells and astrocytes showed increased expression of immune and inflammatory genes, matching those of glia from MS postmortem brains. Thus, iPSC-derived MS models provide a unique platform for dissecting glial contributions to disease phenotypes independent of the peripheral immune system and identify potential glia-specific targets for therapeutic intervention.
Collapse
Affiliation(s)
- Benjamin L L Clayton
- Institute for Glial Sciences, Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Lilianne Barbar
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Maria Sapar
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Kriti Kalpana
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Chandrika Rao
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Bianca Migliori
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Tomasz Rusielewicz
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Daniel Paull
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Katie Brenner
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Dorota Moroziewicz
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Ilana Katz Sand
- Corinne Goldsmith Dickinson Center for Multiple Sclerosis, Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10129, USA
| | - Patrizia Casaccia
- Neuroscience Initiative, Advanced Science Research Center at CUNY, New York, NY 10031, USA
| | - Paul J Tesar
- Institute for Glial Sciences, Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| | - Valentina Fossati
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA.
| |
Collapse
|
4
|
Al Jaf AIA, Peria S, Fabiano T, Ragnini-Wilson A. Remyelinating Drugs at a Crossroad: How to Improve Clinical Efficacy and Drug Screenings. Cells 2024; 13:1326. [PMID: 39195216 DOI: 10.3390/cells13161326] [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: 06/27/2024] [Revised: 08/01/2024] [Accepted: 08/06/2024] [Indexed: 08/29/2024] Open
Abstract
Axons wrapped around the myelin sheath enable fast transmission of neuronal signals in the Central Nervous System (CNS). Unfortunately, myelin can be damaged by injury, viral infection, and inflammatory and neurodegenerative diseases. Remyelination is a spontaneous process that can restore nerve conductivity and thus movement and cognition after a demyelination event. Cumulative evidence indicates that remyelination can be pharmacologically stimulated, either by targeting natural inhibitors of Oligodendrocyte Precursor Cells (OPCs) differentiation or by reactivating quiescent Neural Stem Cells (qNSCs) proliferation and differentiation in myelinating Oligodendrocytes (OLs). Although promising results were obtained in animal models for demyelination diseases, none of the compounds identified have passed all the clinical stages. The significant number of patients who could benefit from remyelination therapies reinforces the urgent need to reassess drug selection approaches and develop strategies that effectively promote remyelination. Integrating Artificial Intelligence (AI)-driven technologies with patient-derived cell-based assays and organoid models is expected to lead to novel strategies and drug screening pipelines to achieve this goal. In this review, we explore the current literature on these technologies and their potential to enhance the identification of more effective drugs for clinical use in CNS remyelination therapies.
Collapse
Affiliation(s)
- Aland Ibrahim Ahmed Al Jaf
- Department of Biology, University of Rome "Tor Vergata", Via della Ricerca Scientifica 1, 00133 Rome, Italy
| | - Simone Peria
- Department of Biology, University of Rome "Tor Vergata", Via della Ricerca Scientifica 1, 00133 Rome, Italy
| | - Tommaso Fabiano
- Department of Biology, University of Rome "Tor Vergata", Via della Ricerca Scientifica 1, 00133 Rome, Italy
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Antonella Ragnini-Wilson
- Department of Biology, University of Rome "Tor Vergata", Via della Ricerca Scientifica 1, 00133 Rome, Italy
| |
Collapse
|
5
|
Zveik O, Rechtman A, Ganz T, Vaknin-Dembinsky A. The interplay of inflammation and remyelination: rethinking MS treatment with a focus on oligodendrocyte progenitor cells. Mol Neurodegener 2024; 19:53. [PMID: 38997755 PMCID: PMC11245841 DOI: 10.1186/s13024-024-00742-8] [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: 05/29/2024] [Accepted: 07/01/2024] [Indexed: 07/14/2024] Open
Abstract
BACKGROUND Multiple sclerosis (MS) therapeutic goals have traditionally been dichotomized into two distinct avenues: immune-modulatory-centric interventions and pro-regenerative strategies. Oligodendrocyte progenitor cells (OPCs) were regarded for many years solely in concern to their potential to generate oligodendrocytes and myelin in the central nervous system (CNS). However, accumulating data elucidate the multifaceted roles of OPCs, including their immunomodulatory functions, positioning them as cardinal constituents of the CNS's immune landscape. MAIN BODY In this review, we will discuss how the two therapeutic approaches converge. We present a model by which (1) an inflammation is required for the appropriate pro-myelinating immune function of OPCs in the chronically inflamed CNS, and (2) the immune function of OPCs is crucial for their ability to differentiate and promote remyelination. This model highlights the reciprocal interactions between OPCs' pro-myelinating and immune-modulating functions. Additionally, we review the specific effects of anti- and pro-inflammatory interventions on OPCs, suggesting that immunosuppression adversely affects OPCs' differentiation and immune functions. CONCLUSION We suggest a multi-systemic therapeutic approach, which necessitates not a unidimensional focus but a harmonious balance between OPCs' pro-myelinating and immune-modulatory functions.
Collapse
Affiliation(s)
- Omri Zveik
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, 91120, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Ein-Kerem P.O.B. 12000, Jerusalem, 91120, Israel
| | - Ariel Rechtman
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, 91120, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Ein-Kerem P.O.B. 12000, Jerusalem, 91120, Israel
| | - Tal Ganz
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, 91120, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Ein-Kerem P.O.B. 12000, Jerusalem, 91120, Israel
| | - Adi Vaknin-Dembinsky
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, 91120, Israel.
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Ein-Kerem P.O.B. 12000, Jerusalem, 91120, Israel.
| |
Collapse
|
6
|
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
|
7
|
Tamada A, Muguruma K. Recapitulation and investigation of human brain development with neural organoids. IBRO Neurosci Rep 2024; 16:106-117. [PMID: 39007085 PMCID: PMC11240300 DOI: 10.1016/j.ibneur.2023.12.006] [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] [Indexed: 07/16/2024] Open
Abstract
Organoids are 3D cultured tissues derived from stem cells that resemble the structure of living organs. Based on the accumulated knowledge of neural development, neural organoids that recapitulate neural tissue have been created by inducing self-organized neural differentiation of stem cells. Neural organoid techniques have been applied to human pluripotent stem cells to differentiate 3D human neural tissues in culture. Various methods have been developed to generate neural tissues of different regions. Currently, neural organoid technology has several significant limitations, which are being overcome in an attempt to create neural organoids that more faithfully recapitulate the living brain. The rapidly advancing neural organoid technology enables the use of living human neural tissue as research material and contributes to our understanding of the development, structure and function of the human nervous system, and is expected to be used to overcome neurological diseases and for regenerative medicine.
Collapse
Affiliation(s)
- Atsushi Tamada
- Department of iPS Cell Applied Medicine, Faculty of Medicine, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Keiko Muguruma
- Department of iPS Cell Applied Medicine, Faculty of Medicine, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| |
Collapse
|
8
|
Liu X, Xin DE, Zhong X, Zhao C, Li Z, Zhang L, Dourson AJ, Lee L, Mishra S, Bayat AE, Nicholson E, Seibel WL, Yan B, Mason J, Turner BJ, Gonsalvez DG, Ong W, Chew SY, Ghosh B, Yoon SO, Xin M, He Z, Tchieu J, Wegner M, Nave KA, Franklin RJM, Dutta R, Trapp BD, Hu M, Smith MA, Jankowski MP, Barton SK, He X, Lu QR. Small-molecule-induced epigenetic rejuvenation promotes SREBP condensation and overcomes barriers to CNS myelin regeneration. Cell 2024; 187:2465-2484.e22. [PMID: 38701782 DOI: 10.1016/j.cell.2024.04.005] [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: 05/31/2023] [Revised: 01/01/2024] [Accepted: 04/04/2024] [Indexed: 05/05/2024]
Abstract
Remyelination failure in diseases like multiple sclerosis (MS) was thought to involve suppressed maturation of oligodendrocyte precursors; however, oligodendrocytes are present in MS lesions yet lack myelin production. We found that oligodendrocytes in the lesions are epigenetically silenced. Developing a transgenic reporter labeling differentiated oligodendrocytes for phenotypic screening, we identified a small-molecule epigenetic-silencing-inhibitor (ESI1) that enhances myelin production and ensheathment. ESI1 promotes remyelination in animal models of demyelination and enables de novo myelinogenesis on regenerated CNS axons. ESI1 treatment lengthened myelin sheaths in human iPSC-derived organoids and augmented (re)myelination in aged mice while reversing age-related cognitive decline. Multi-omics revealed that ESI1 induces an active chromatin landscape that activates myelinogenic pathways and reprograms metabolism. Notably, ESI1 triggered nuclear condensate formation of master lipid-metabolic regulators SREBP1/2, concentrating transcriptional co-activators to drive lipid/cholesterol biosynthesis. Our study highlights the potential of targeting epigenetic silencing to enable CNS myelin regeneration in demyelinating diseases and aging.
Collapse
Affiliation(s)
- Xuezhao Liu
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Dazhuan Eric Xin
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Xiaowen Zhong
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, Ohio, 45229, USA
| | - Chuntao Zhao
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Zhidan Li
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Liguo Zhang
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Adam J Dourson
- Department of Anesthesia, Division of Pain Management, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Lindsay Lee
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Shreya Mishra
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Arman E Bayat
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Eva Nicholson
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - William L Seibel
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Bingfang Yan
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, Ohio, 45229, USA
| | - Joel Mason
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne 3052, Australia
| | - Bradley J Turner
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne 3052, Australia
| | - David G Gonsalvez
- Department of Anatomy and Developmental Biology, Monash University, Melbourne 3168, Australia
| | - William Ong
- School of Chemistry, Chemical Engineering, and Biotechnology Nanyang Technological University, Singapore 637459, Singapore
| | - Sing Yian Chew
- School of Chemistry, Chemical Engineering, and Biotechnology Nanyang Technological University, Singapore 637459, Singapore; Lee Kong Chian School of Medicine, School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Balaram Ghosh
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani Hyderabad Campus, Shamirpet, Hyderabad, India, 500078
| | - Sung Ok Yoon
- Department of Biological Chemistry and Pharmacology, Ohio State University, Columbus, Ohio
| | - Mei Xin
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Zhigang He
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, and Department of Neurology and Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Jason Tchieu
- Department of Pediatrics, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Michael Wegner
- Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Robin J M Franklin
- Altos Labs, Cambridge Institute of Science, Granta Park, Cambridge CB21 6GP, UK
| | - Ranjan Dutta
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Case Western Reserve University School of Medicine, Cleveland, OH 44195, USA
| | - Bruce D Trapp
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Case Western Reserve University School of Medicine, Cleveland, OH 44195, USA
| | - Ming Hu
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Matthew A Smith
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA; Rebecca D. Considine Research Institute, Akron Children's Hospital, Akron, OH, USA
| | - Michael P Jankowski
- Department of Anesthesia, Division of Pain Management, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Pediatric Pain Research Center, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Samantha K Barton
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne 3052, Australia
| | - Xuelian He
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P.R. China.
| | - Q Richard Lu
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
9
|
Liu H, Liu Y, Zhou Y, Chen X, Pan S, Zhou Q, Ji H, Zhu X. TM7SF2-induced lipid reprogramming promotes cell proliferation and migration via CPT1A/Wnt/β-Catenin axis in cervical cancer cells. Cell Death Discov 2024; 10:207. [PMID: 38693136 PMCID: PMC11063194 DOI: 10.1038/s41420-024-01975-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 04/14/2024] [Accepted: 04/18/2024] [Indexed: 05/03/2024] Open
Abstract
Cervical cancer poses a serious threat to women's health globally. Our previous studies found that upregulation of TM7SF2, which works as an enzyme involved in the process of cholesterol biosynthesis expression, was highly correlated with cervical cancer. However, the mechanistic basis of TM7SF2 promoting cervical cancer progression via lipid metabolism remains poorly understood. Therefore, quantification of fatty acids and lipid droplets were performed in vitro and in vivo. The protein-protein interaction was verified by Co-IP technique. The mechanism and underlying signaling pathway of TM7SF2 via CPT1A associated lipid metabolism in cervical cancer development were explored using Western blotting, IHC, colony formation, transwell assay, and wound healing assay. This study reported that overexpression of TM7SF2 increased fatty acids content and lipid droplets both in vivo and in vitro experiments. While knockout of TM7SF2 obviously attenuated this process. Moreover, TM7SF2 directly bonded with CPT1A, a key enzyme in fatty acid oxidation, and regulated CPT1A protein expression in cervical cancer cells. Notably, the proliferation and metastasis of cervical cancer cells were elevated when their CPT1A expression was upregulated. Then, rescue assay identified that CPT1A overexpressed could enhance the cell viability and migration in TM7SF2-knockout cells. Furthermore, depletion of TM7SF2 significantly inhibited WNT and β-catenin proteins expression, which was enhanced by CPT1A-overexpressed. The proliferation and migration of cervical cancer cells were reversed in CPT1A-overexpressed cells with the treatment of MSAB, an inhibitor of Wnt/β-Catenin pathway. This study put forward an idea that TM7SF2-induced lipid reprogramming promotes proliferation and migration via CPT1A/Wnt/β-Catenin axis in cervical cancer, underlying the progression of cervical cancer.
Collapse
Affiliation(s)
- Hejing Liu
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Yi Liu
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Yujia Zhou
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Xin Chen
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Shuya Pan
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Qingfeng Zhou
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Huihui Ji
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China.
| | - Xueqiong Zhu
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China.
| |
Collapse
|
10
|
Elitt MS, Tesar PJ. Pelizaeus-Merzbacher disease: on the cusp of myelin medicine. Trends Mol Med 2024; 30:459-470. [PMID: 38582621 PMCID: PMC11081862 DOI: 10.1016/j.molmed.2024.03.005] [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: 01/02/2024] [Revised: 03/05/2024] [Accepted: 03/08/2024] [Indexed: 04/08/2024]
Abstract
Pelizaeus-Merzbacher disease (PMD) is caused by mutations in the proteolipid protein 1 (PLP1) gene encoding proteolipid protein (PLP). As a major component of myelin, mutated PLP causes progressive neurodegeneration and eventually death due to severe white matter deficits. Medical care has long been limited to symptomatic treatments, but first-in-class PMD therapies with novel mechanisms now stand poised to enter clinical trials. Here, we review PMD disease mechanisms and outline rationale for therapeutic interventions, including PLP1 suppression, cell transplantation, iron chelation, and intracellular stress modulation. We discuss available preclinical data and their implications on clinical development. With several novel treatments on the horizon, PMD is on the precipice of a new era in the diagnosis and treatment of patients suffering from this debilitating disease.
Collapse
Affiliation(s)
- Matthew S Elitt
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Paul J Tesar
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| |
Collapse
|
11
|
Cohn EF, Clayton BLL, Madhavan M, Lee KA, Yacoub S, Fedorov Y, Scavuzzo MA, Paul Friedman K, Shafer TJ, Tesar PJ. Pervasive environmental chemicals impair oligodendrocyte development. Nat Neurosci 2024; 27:836-845. [PMID: 38528201 PMCID: PMC11088982 DOI: 10.1038/s41593-024-01599-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 02/05/2024] [Indexed: 03/27/2024]
Abstract
Exposure to environmental chemicals can impair neurodevelopment, and oligodendrocytes may be particularly vulnerable, as their development extends from gestation into adulthood. However, few environmental chemicals have been assessed for potential risks to oligodendrocytes. Here, using a high-throughput developmental screen in cultured cells, we identified environmental chemicals in two classes that disrupt oligodendrocyte development through distinct mechanisms. Quaternary compounds, ubiquitous in disinfecting agents and personal care products, were potently and selectively cytotoxic to developing oligodendrocytes, whereas organophosphate flame retardants, commonly found in household items such as furniture and electronics, prematurely arrested oligodendrocyte maturation. Chemicals from each class impaired oligodendrocyte development postnatally in mice and in a human 3D organoid model of prenatal cortical development. Analysis of epidemiological data showed that adverse neurodevelopmental outcomes were associated with childhood exposure to the top organophosphate flame retardant identified by our screen. This work identifies toxicological vulnerabilities for oligodendrocyte development and highlights the need for deeper scrutiny of these compounds' impacts on human health.
Collapse
Affiliation(s)
- Erin F Cohn
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Benjamin L L Clayton
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Mayur Madhavan
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Kristin A Lee
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Sara Yacoub
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Yuriy Fedorov
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Marissa A Scavuzzo
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Katie Paul Friedman
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Timothy J Shafer
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Paul J Tesar
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| |
Collapse
|
12
|
Goldfarb RB, Atala Pleshinger MJ, Yan DF, Adams DJ. Lipid-Restricted Culture Media Reveal Unexpected Cancer Cell Sensitivities. ACS Chem Biol 2024; 19:896-907. [PMID: 38506663 DOI: 10.1021/acschembio.3c00699] [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: 03/21/2024]
Abstract
Cancer cell culture models frequently rely on fetal bovine serum as a source of protein and lipid factors that support cell survival and proliferation; however, serum-containing media imperfectly mimic the in vivo cancer environment. Recent studies suggest that typical serum-containing cell culture conditions can mask cancer dependencies, for example, on cholesterol biosynthesis enzymes, that exist in vivo and emerge when cells are cultured in media that provide more realistic levels of lipids. Here, we describe a high-throughput screen that identified fenretinide and ivermectin as small molecules whose cytotoxicity is greatly enhanced in lipid-restricted media formulations. The mechanism of action studies indicates that ivermectin-induced cell death involves oxidative stress, while fenretinide likely targets delta 4-desaturase, sphingolipid 1, a lipid desaturase necessary for ceramide synthesis, to induce cell death. Notably, both fenretinide and ivermectin have previously demonstrated in vivo anticancer efficacy despite their low cytotoxicity under typical cell culture conditions. These studies suggest ceramide synthesis as a targetable vulnerability of cancer cells cultured under lipid-restricted conditions and reveal a general screening strategy for identifying additional cancer dependencies masked by the superabundance of medium lipids.
Collapse
Affiliation(s)
- Ralston B Goldfarb
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, United States
- Chemical Biology Program, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, United States
| | - Matthew J Atala Pleshinger
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, United States
- Chemical Biology Program, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, United States
| | - David F Yan
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, United States
- Chemical Biology Program, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, United States
| | - Drew J Adams
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, United States
- Chemical Biology Program, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, United States
| |
Collapse
|
13
|
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
|
14
|
Dorel R, Sun D, Carruthers N, Castanedo GM, Ung PMU, Factor DC, Li T, Baumann H, Janota D, Pang J, Salphati L, Meklemburg R, Korman AJ, Harper HE, Stubblefield S, Payandeh J, McHugh D, Lang BT, Tesar PJ, Dere E, Masureel M, Adams DJ, Volgraf M, Braun MG. Discovery and Optimization of Selective Brain-Penetrant EBP Inhibitors that Enhance Oligodendrocyte Formation. J Med Chem 2024. [PMID: 38470227 DOI: 10.1021/acs.jmedchem.3c02396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
The inhibition of emopamil binding protein (EBP), a sterol isomerase within the cholesterol biosynthesis pathway, promotes oligodendrocyte formation, which has been proposed as a potential therapeutic approach for treating multiple sclerosis. Herein, we describe the discovery and optimization of brain-penetrant, orally bioavailable inhibitors of EBP. A structure-based drug design approach from literature compound 1 led to the discovery of a hydantoin-based scaffold, which provided balanced physicochemical properties and potency and an improved in vitro safety profile. The long half-lives of early hydantoin-based EBP inhibitors in rodents prompted an unconventional optimization strategy, focused on increasing metabolic turnover while maintaining potency and a brain-penetrant profile. The resulting EBP inhibitor 11 demonstrated strong in vivo target engagement in the brain, as illustrated by the accumulation of EBP substrate zymostenol after repeated dosing. Furthermore, compound 11 enhanced the formation of oligodendrocytes in human cortical organoids, providing additional support for our therapeutic hypothesis.
Collapse
Affiliation(s)
- Ruth Dorel
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Dawei Sun
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Nicholas Carruthers
- Convelo Therapeutics, 11000 Cedar Avenue, Cleveland, Ohio 44106, United States
| | | | - Peter M-U Ung
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Daniel C Factor
- Convelo Therapeutics, 11000 Cedar Avenue, Cleveland, Ohio 44106, United States
| | - Tianbo Li
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Hannah Baumann
- Convelo Therapeutics, 11000 Cedar Avenue, Cleveland, Ohio 44106, United States
| | - Danielle Janota
- Convelo Therapeutics, 11000 Cedar Avenue, Cleveland, Ohio 44106, United States
| | - Jodie Pang
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Laurent Salphati
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Robert Meklemburg
- Convelo Therapeutics, 11000 Cedar Avenue, Cleveland, Ohio 44106, United States
| | - Allison J Korman
- Convelo Therapeutics, 11000 Cedar Avenue, Cleveland, Ohio 44106, United States
| | - Halie E Harper
- Convelo Therapeutics, 11000 Cedar Avenue, Cleveland, Ohio 44106, United States
| | | | - Jian Payandeh
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Daniel McHugh
- Convelo Therapeutics, 11000 Cedar Avenue, Cleveland, Ohio 44106, United States
| | - Bradley T Lang
- Convelo Therapeutics, 11000 Cedar Avenue, Cleveland, Ohio 44106, United States
| | - Paul J Tesar
- Convelo Therapeutics, 11000 Cedar Avenue, Cleveland, Ohio 44106, United States
| | - Edward Dere
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Matthieu Masureel
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Drew J Adams
- Convelo Therapeutics, 11000 Cedar Avenue, Cleveland, Ohio 44106, United States
| | - Matthew Volgraf
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | | |
Collapse
|
15
|
Göttle P, Dietrich M, Küry P. Multiple sclerosis drug repurposing for neuroregeneration. Neural Regen Res 2024; 19:507-508. [PMID: 37721276 PMCID: PMC10581586 DOI: 10.4103/1673-5374.380901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/07/2023] [Accepted: 06/20/2023] [Indexed: 09/19/2023] Open
Affiliation(s)
- Peter Göttle
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Michael Dietrich
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Patrick Küry
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| |
Collapse
|
16
|
Hill RA, Nishiyama A, Hughes EG. Features, Fates, and Functions of Oligodendrocyte Precursor Cells. Cold Spring Harb Perspect Biol 2024; 16:a041425. [PMID: 38052500 PMCID: PMC10910408 DOI: 10.1101/cshperspect.a041425] [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: 12/07/2023]
Abstract
Oligodendrocyte precursor cells (OPCs) are a central nervous system resident population of glia with a distinct molecular identity and an ever-increasing list of functions. OPCs generate oligodendrocytes throughout development and across the life span in most regions of the brain and spinal cord. This process involves a complex coordination of molecular checkpoints and biophysical cues from the environment that initiate the differentiation and integration of new oligodendrocytes that synthesize myelin sheaths on axons. Outside of their progenitor role, OPCs have been proposed to play other functions including the modulation of axonal and synaptic development and the participation in bidirectional signaling with neurons and other glia. Here, we review OPC identity and known functions and discuss recent findings implying other roles for these glial cells in brain physiology and pathology.
Collapse
Affiliation(s)
- Robert A Hill
- Department of Biological Sciences, Dartmouth College, Hanover, New Hampshire 03755, USA
| | - Akiko Nishiyama
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut 06269, USA
| | - Ethan G Hughes
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| |
Collapse
|
17
|
Cui SH, Suo N, Yang Y, Wu X, Guo SM, Xie X. The aminosteroid U73122 promotes oligodendrocytes generation and myelin formation. Acta Pharmacol Sin 2024; 45:490-501. [PMID: 37935896 PMCID: PMC10834981 DOI: 10.1038/s41401-023-01183-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/13/2023] [Indexed: 11/09/2023] Open
Abstract
Oligodendrocytes (OLs) are glial cells that ensheath neuronal axons and form myelin in the central nervous system (CNS). OLs are differentiated from oligodendrocyte precursor cells (OPCs) during development and myelin repair, which is often insufficient in the latter case in demyelinating diseases such as multiple sclerosis (MS). Many factors have been reported to regulate OPC-to-OL differentiation, including a number of G protein-coupled receptors (GPCRs). In an effort to search pathways downstream of GPCRs that might be involved in OPC differentiation, we discover that U73122, a phosphoinositide specific phospholipase C (PI-PLC) inhibitor, dramatically promotes OPC-to-OL differentiation and myelin regeneration in experimental autoimmune encephalomyelitis model. Unexpectedly, U73343, a close analog of U73122 which lacks PI-PLC inhibitory activity also promotes OL differentiation, while another reported PI-PLC inhibitor edelfosine does not have such effect, suggesting that U73122 and U73343 enhance OPC differentiation independent of PLC. Although the structures of U73122 and U73343 closely resemble 17β-estradiol, and both compounds do activate estrogen receptors Erα and Erβ with low efficacy and potency, further study indicates that these compounds do not act through Erα and/or Erβ to promote OPC differentiation. RNA-Seq and bioinformatic analysis indicate that U73122 and U73343 may regulate cholesterol biosynthesis. Further study shows both compounds increase 14-dehydrozymostenol, a steroid reported to promote OPC differentiation, in OPC culture. In conclusion, the aminosteroids U73122 and U73343 promote OPC-to-OL generation and myelin formation by regulating cholesterol biosynthesis pathway.
Collapse
Affiliation(s)
- Shi-Hao Cui
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Na Suo
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Ying Yang
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Xuan Wu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Shi-Meng Guo
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xin Xie
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China.
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China.
| |
Collapse
|
18
|
Franklin RJM, Bodini B, Goldman SA. Remyelination in the Central Nervous System. Cold Spring Harb Perspect Biol 2024; 16:a041371. [PMID: 38316552 PMCID: PMC10910446 DOI: 10.1101/cshperspect.a041371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
The inability of the mammalian central nervous system (CNS) to undergo spontaneous regeneration has long been regarded as a central tenet of neurobiology. However, while this is largely true of the neuronal elements of the adult mammalian CNS, save for discrete populations of granule neurons, the same is not true of its glial elements. In particular, the loss of oligodendrocytes, which results in demyelination, triggers a spontaneous and often highly efficient regenerative response, remyelination, in which new oligodendrocytes are generated and myelin sheaths are restored to denuded axons. Yet remyelination in humans is not without limitation, and a variety of demyelinating conditions are associated with sustained and disabling myelin loss. In this work, we will (1) review the biology of remyelination, including the cells and signals involved; (2) describe when remyelination occurs and when and why it fails, including the consequences of its failure; and (3) discuss approaches for therapeutically enhancing remyelination in demyelinating diseases of both children and adults, both by stimulating endogenous oligodendrocyte progenitor cells and by transplanting these cells into demyelinated brain.
Collapse
Affiliation(s)
- Robin J M Franklin
- Altos Labs Cambridge Institute of Science, Cambridge CB21 6GH, United Kingdom
| | - Benedetta Bodini
- Sorbonne Université, Paris Brain Institute, CNRS, INSERM, Paris 75013, France
- Saint-Antoine Hospital, APHP, Paris 75012, France
| | - Steven A Goldman
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York 14642, USA
- University of Copenhagen Faculty of Medicine, Copenhagen 2200, Denmark
| |
Collapse
|
19
|
Madan B, Wadia SR, Patnaik S, Harmston N, Tan E, Tan IBH, Nes WD, Petretto E, Virshup DM. The cholesterol biosynthesis enzyme FAXDC2 couples Wnt/β-catenin to RTK/MAPK signaling. J Clin Invest 2024; 134:e171222. [PMID: 38488003 PMCID: PMC10940096 DOI: 10.1172/jci171222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 01/17/2024] [Indexed: 03/18/2024] Open
Abstract
Wnts, cholesterol, and MAPK signaling are essential for development and adult homeostasis. Here, we report that fatty acid hydroxylase domain containing 2 (FAXDC2), a previously uncharacterized enzyme, functions as a methyl sterol oxidase catalyzing C4 demethylation in the Kandutsch-Russell branch of the cholesterol biosynthesis pathway. FAXDC2, a paralog of MSMO1, regulated the abundance of the specific C4-methyl sterols lophenol and dihydro-T-MAS. Highlighting its clinical relevance, FAXDC2 was repressed in Wnt/β-catenin-high cancer xenografts, in a mouse genetic model of Wnt activation, and in human colorectal cancers. Moreover, in primary human colorectal cancers, the sterol lophenol, regulated by FAXDC2, accumulated in the cancerous tissues and not in adjacent normal tissues. FAXDC2 linked Wnts to RTK/MAPK signaling. Wnt inhibition drove increased recycling of RTKs and activation of the MAPK pathway, and this required FAXDC2. Blocking Wnt signaling in Wnt-high cancers caused both differentiation and senescence; and this was prevented by knockout of FAXDC2. Our data show the integration of 3 ancient pathways, Wnts, cholesterol synthesis, and RTK/MAPK signaling, in cellular proliferation and differentiation.
Collapse
Affiliation(s)
- Babita Madan
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore
| | - Shawn R. Wadia
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore
| | - Siddhi Patnaik
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore
| | - Nathan Harmston
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore
- Science Division, Yale-NUS College, Singapore
| | - Emile Tan
- Department of Colorectal Surgery, Singapore General Hospital, Singapore
| | - Iain Bee Huat Tan
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore
- Department of Medical Oncology, National Cancer Centre, Singapore
| | - W. David Nes
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas, USA
| | - Enrico Petretto
- Center for Computational Biology and Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore
- Institute for Big Data and Artificial Intelligence in Medicine, School of Science, China Pharmaceutical University, Nanjing, China
| | - David M. Virshup
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore
- Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
20
|
de Medina P, Ayadi S, Diallo K, Buñay J, Pucheu L, Soulès R, Record M, Brillouet S, Vija L, Courbon F, Silvente-Poirot S, Poirot M. The Cholesterol-5,6-Epoxide Hydrolase: A Metabolic Checkpoint in Several Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1440:149-161. [PMID: 38036879 DOI: 10.1007/978-3-031-43883-7_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Cholesterol-5,6-epoxides (5,6-ECs) are oxysterols (OS) that have been linked to several pathologies including cancers and neurodegenerative diseases. 5,6-ECs can be produced from cholesterol by several mechanisms including reactive oxygen species, lipoperoxidation, and cytochrome P450 enzymes. 5,6-ECs exist as two different diastereoisomers: 5,6α-EC and 5,6β-EC with different metabolic fates. They can be produced as a mixture or as single products of epoxidation. The epoxide ring of 5,6α-EC and 5,6β-EC is very stable and 5,6-ECs are prone to hydration by the cholesterol-5,6-epoxide hydrolase (ChEH) to give cholestane-3β,5α,6β-triol, which can be further oxidized into oncosterone. 5,6α-EC is prone to chemical and enzymatic conjugation reactions leading to bioactive compounds such as dendrogenins, highlighting the existence of a new metabolic branch on the cholesterol pathway centered on 5,6α-EC. We will summarize in this chapter current knowledge on this pathway which is controlled by the ChEH.
Collapse
Affiliation(s)
- Philippe de Medina
- Cancer Research Center of Toulouse (CRCT), Inserm, CNRS, University of Toulouse, Team INOV: "Cholesterol Metabolism and Therapeutic Innovations", Toulouse, France
- Equipe labellisée par la Ligue Nationale contre le Cancer, Toulouse, France
- French Network for Nutrition Physical Activity and Cancer Research (NACRe Network), Jouy-en-Josas, France
| | - Silia Ayadi
- Cancer Research Center of Toulouse (CRCT), Inserm, CNRS, University of Toulouse, Team INOV: "Cholesterol Metabolism and Therapeutic Innovations", Toulouse, France
- Equipe labellisée par la Ligue Nationale contre le Cancer, Toulouse, France
- French Network for Nutrition Physical Activity and Cancer Research (NACRe Network), Jouy-en-Josas, France
| | - Khadijetou Diallo
- Cancer Research Center of Toulouse (CRCT), Inserm, CNRS, University of Toulouse, Team INOV: "Cholesterol Metabolism and Therapeutic Innovations", Toulouse, France
- Equipe labellisée par la Ligue Nationale contre le Cancer, Toulouse, France
- French Network for Nutrition Physical Activity and Cancer Research (NACRe Network), Jouy-en-Josas, France
| | - Julio Buñay
- Cancer Research Center of Toulouse (CRCT), Inserm, CNRS, University of Toulouse, Team INOV: "Cholesterol Metabolism and Therapeutic Innovations", Toulouse, France
- Equipe labellisée par la Ligue Nationale contre le Cancer, Toulouse, France
- French Network for Nutrition Physical Activity and Cancer Research (NACRe Network), Jouy-en-Josas, France
| | - Laly Pucheu
- Cancer Research Center of Toulouse (CRCT), Inserm, CNRS, University of Toulouse, Team INOV: "Cholesterol Metabolism and Therapeutic Innovations", Toulouse, France
- Equipe labellisée par la Ligue Nationale contre le Cancer, Toulouse, France
- French Network for Nutrition Physical Activity and Cancer Research (NACRe Network), Jouy-en-Josas, France
| | - Regis Soulès
- Cancer Research Center of Toulouse (CRCT), Inserm, CNRS, University of Toulouse, Team INOV: "Cholesterol Metabolism and Therapeutic Innovations", Toulouse, France
- Equipe labellisée par la Ligue Nationale contre le Cancer, Toulouse, France
- French Network for Nutrition Physical Activity and Cancer Research (NACRe Network), Jouy-en-Josas, France
| | - Michel Record
- Cancer Research Center of Toulouse (CRCT), Inserm, CNRS, University of Toulouse, Team INOV: "Cholesterol Metabolism and Therapeutic Innovations", Toulouse, France
- Equipe labellisée par la Ligue Nationale contre le Cancer, Toulouse, France
- French Network for Nutrition Physical Activity and Cancer Research (NACRe Network), Jouy-en-Josas, France
| | - Severine Brillouet
- Cancer Research Center of Toulouse (CRCT), Inserm, CNRS, University of Toulouse, Team INOV: "Cholesterol Metabolism and Therapeutic Innovations", Toulouse, France
- Equipe labellisée par la Ligue Nationale contre le Cancer, Toulouse, France
- French Network for Nutrition Physical Activity and Cancer Research (NACRe Network), Jouy-en-Josas, France
- Department of Radiopharmacy, Institut Universitaire du Cancer Toulouse - Oncopole, Toulouse, France
| | - Lavinia Vija
- Cancer Research Center of Toulouse (CRCT), Inserm, CNRS, University of Toulouse, Team INOV: "Cholesterol Metabolism and Therapeutic Innovations", Toulouse, France
- Equipe labellisée par la Ligue Nationale contre le Cancer, Toulouse, France
- French Network for Nutrition Physical Activity and Cancer Research (NACRe Network), Jouy-en-Josas, France
- Department of Medical Imaging, Institut Universitaire du Cancer Toulouse - Oncopole, Toulouse, France
| | - Frederic Courbon
- Cancer Research Center of Toulouse (CRCT), Inserm, CNRS, University of Toulouse, Team INOV: "Cholesterol Metabolism and Therapeutic Innovations", Toulouse, France
- Equipe labellisée par la Ligue Nationale contre le Cancer, Toulouse, France
- French Network for Nutrition Physical Activity and Cancer Research (NACRe Network), Jouy-en-Josas, France
- Department of Medical Imaging, Institut Universitaire du Cancer Toulouse - Oncopole, Toulouse, France
| | - Sandrine Silvente-Poirot
- Cancer Research Center of Toulouse (CRCT), Inserm, CNRS, University of Toulouse, Team INOV: "Cholesterol Metabolism and Therapeutic Innovations", Toulouse, France
- Equipe labellisée par la Ligue Nationale contre le Cancer, Toulouse, France
- French Network for Nutrition Physical Activity and Cancer Research (NACRe Network), Jouy-en-Josas, France
| | - Marc Poirot
- Cancer Research Center of Toulouse (CRCT), Inserm, CNRS, University of Toulouse, Team INOV: "Cholesterol Metabolism and Therapeutic Innovations", Toulouse, France.
- Equipe labellisée par la Ligue Nationale contre le Cancer, Toulouse, France.
- French Network for Nutrition Physical Activity and Cancer Research (NACRe Network), Jouy-en-Josas, France.
| |
Collapse
|
21
|
Area-Gomez E, Schon EA. Towards a Unitary Hypothesis of Alzheimer's Disease Pathogenesis. J Alzheimers Dis 2024; 98:1243-1275. [PMID: 38578892 DOI: 10.3233/jad-231318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2024]
Abstract
The "amyloid cascade" hypothesis of Alzheimer's disease (AD) pathogenesis invokes the accumulation in the brain of plaques (containing the amyloid-β protein precursor [AβPP] cleavage product amyloid-β [Aβ]) and tangles (containing hyperphosphorylated tau) as drivers of pathogenesis. However, the poor track record of clinical trials based on this hypothesis suggests that the accumulation of these peptides is not the only cause of AD. Here, an alternative hypothesis is proposed in which the AβPP cleavage product C99, not Aβ, is the main culprit, via its role as a regulator of cholesterol metabolism. C99, which is a cholesterol sensor, promotes the formation of mitochondria-associated endoplasmic reticulum (ER) membranes (MAM), a cholesterol-rich lipid raft-like subdomain of the ER that communicates, both physically and biochemically, with mitochondria. We propose that in early-onset AD (EOAD), MAM-localized C99 is elevated above normal levels, resulting in increased transport of cholesterol from the plasma membrane to membranes of intracellular organelles, such as ER/endosomes, thereby upregulating MAM function and driving pathology. By the same token, late-onset AD (LOAD) is triggered by any genetic variant that increases the accumulation of intracellular cholesterol that, in turn, boosts the levels of C99 and again upregulates MAM function. Thus, the functional cause of AD is upregulated MAM function that, in turn, causes the hallmark disease phenotypes, including the plaques and tangles. Accordingly, the MAM hypothesis invokes two key interrelated elements, C99 and cholesterol, that converge at the MAM to drive AD pathogenesis. From this perspective, AD is, at bottom, a lipid disorder.
Collapse
Affiliation(s)
- Estela Area-Gomez
- Department of Neurology, Columbia University, New York, NY, USA
- Centro de Investigaciones Biológicas "Margarita Salas", Spanish National Research Council, Madrid, Spain
| | - Eric A Schon
- Department of Neurology, Columbia University, New York, NY, USA
- Department of Genetics and Development>, Columbia University, New York, NY, USA
| |
Collapse
|
22
|
Mwema A, Muccioli GG, des Rieux A. Innovative drug delivery strategies to the CNS for the treatment of multiple sclerosis. J Control Release 2023; 364:435-457. [PMID: 37926243 DOI: 10.1016/j.jconrel.2023.10.052] [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: 07/05/2023] [Revised: 10/05/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023]
Abstract
Disorders of the central nervous system (CNS), such as multiple sclerosis (MS) represent a great emotional, financial and social burden. Despite intense efforts, great unmet medical needs remain in that field. MS is an autoimmune, chronic inflammatory demyelinating disease with no curative treatment up to date. The current therapies mostly act in the periphery and seek to modulate aberrant immune responses as well as slow down the progression of the disease. Some of these therapies are associated with adverse effects related partly to their administration route and show some limitations due to their rapid clearance and inability to reach the CNS. The scientific community have recently focused their research on developing MS therapies targeting different processes within the CNS. However, delivery of therapeutics to the CNS is mainly limited by the presence of the blood-brain barrier (BBB). Therefore, there is a pressing need to develop new drug delivery strategies that ensure CNS availability to capitalize on identified therapeutic targets. Several approaches have been developed to overcome or bypass the BBB and increase delivery of therapeutics to the CNS. Among these strategies, the use of alternative routes of administration, such as the nose-to-brain (N2B) pathway, offers a promising non-invasive option in the scope of MS, as it would allow a direct transport of the drugs from the nasal cavity to the brain. Moreover, the combination of bioactive molecules within nanocarriers bring forth new opportunities for MS therapies, allowing and/or increasing their transport to the CNS. Here we will review and discuss these alternative administration routes as well as the nanocarrier approaches useful to deliver drugs for MS.
Collapse
Affiliation(s)
- Ariane Mwema
- Université catholique de Louvain, UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue E. Mounier 73, 1200 Brussels, Belgium; Université catholique de Louvain, UCLouvain, Louvain Drug Research Institute, Bioanalysis and Pharmacology of Bioactive Lipids, Avenue E. Mounier 72, 1200 Brussels, Belgium
| | - Giulio G Muccioli
- Université catholique de Louvain, UCLouvain, Louvain Drug Research Institute, Bioanalysis and Pharmacology of Bioactive Lipids, Avenue E. Mounier 72, 1200 Brussels, Belgium.
| | - Anne des Rieux
- Université catholique de Louvain, UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue E. Mounier 73, 1200 Brussels, Belgium.
| |
Collapse
|
23
|
Sun S, Li J, Wang S, Li J, Ren J, Bao Z, Sun L, Ma X, Zheng F, Ma S, Sun L, Wang M, Yu Y, Ma M, Wang Q, Chen Z, Ma H, Wang X, Wu Z, Zhang H, Yan K, Yang Y, Zhang Y, Zhang S, Lei J, Teng ZQ, Liu CM, Bai G, Wang YJ, Li J, Wang X, Zhao G, Jiang T, Belmonte JCI, Qu J, Zhang W, Liu GH. CHIT1-positive microglia drive motor neuron ageing in the primate spinal cord. Nature 2023; 624:611-620. [PMID: 37907096 DOI: 10.1038/s41586-023-06783-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 10/25/2023] [Indexed: 11/02/2023]
Abstract
Ageing is a critical factor in spinal-cord-associated disorders1, yet the ageing-specific mechanisms underlying this relationship remain poorly understood. Here, to address this knowledge gap, we combined single-nucleus RNA-sequencing analysis with behavioural and neurophysiological analysis in non-human primates (NHPs). We identified motor neuron senescence and neuroinflammation with microglial hyperactivation as intertwined hallmarks of spinal cord ageing. As an underlying mechanism, we identified a neurotoxic microglial state demarcated by elevated expression of CHIT1 (a secreted mammalian chitinase) specific to the aged spinal cords in NHP and human biopsies. In the aged spinal cord, CHIT1-positive microglia preferentially localize around motor neurons, and they have the ability to trigger senescence, partly by activating SMAD signalling. We further validated the driving role of secreted CHIT1 on MN senescence using multimodal experiments both in vivo, using the NHP spinal cord as a model, and in vitro, using a sophisticated system modelling the human motor-neuron-microenvironment interplay. Moreover, we demonstrated that ascorbic acid, a geroprotective compound, counteracted the pro-senescent effect of CHIT1 and mitigated motor neuron senescence in aged monkeys. Our findings provide the single-cell resolution cellular and molecular landscape of the aged primate spinal cord and identify a new biomarker and intervention target for spinal cord degeneration.
Collapse
Affiliation(s)
- Shuhui Sun
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Jiaming Li
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Si Wang
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China
- Aging Biomarker Consortium, Beijing, China
- The Fifth People's Hospital of Chongqing, Chongqing, China
| | - Jingyi Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Aging Biomarker Consortium, Beijing, China
| | - Jie Ren
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Aging Biomarker Consortium, Beijing, China
- Key Laboratory of RNA Science and Engineering, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
| | - Zhaoshi Bao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- The Chinese Glioma Genome Atlas, Beijing, China
| | - Le Sun
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Xibo Ma
- MAIS, State Key Laboratory of Multimodal Artificial Intelligence Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
- College of Medicine and Biomedical Information Engineering, Northeastern University, Shenyang, China
| | - Fangshuo Zheng
- The Fifth People's Hospital of Chongqing, Chongqing, China
| | - Shuai Ma
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Aging Biomarker Consortium, Beijing, China
| | - Liang Sun
- Aging Biomarker Consortium, Beijing, China
- The MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Min Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Science and Technology of China, Hefei, China
| | - Yan Yu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Miyang Ma
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qiaoran Wang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhiyuan Chen
- MAIS, State Key Laboratory of Multimodal Artificial Intelligence Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - He Ma
- MAIS, State Key Laboratory of Multimodal Artificial Intelligence Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- College of Medicine and Biomedical Information Engineering, Northeastern University, Shenyang, China
| | - Xuebao Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zeming Wu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Hui Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Kaowen Yan
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Yuanhan Yang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yixin Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Sheng Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jinghui Lei
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zhao-Qian Teng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Chang-Mei Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ge Bai
- The MOE Frontier Research Center of Brain & Brain-Machine Integration, Zhejiang University School of Brain Science and Brain Medicine, Hangzhou, China
| | - Yan-Jiang Wang
- Aging Biomarker Consortium, Beijing, China
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Jian Li
- Aging Biomarker Consortium, Beijing, China
- The MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Xiaoqun Wang
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, New Cornerstone Science Laboratory, Beijing Normal University, Beijing, China
| | - Guoguang Zhao
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, China
- Clinical Research Center for Epilepsy Capital Medical University, Beijing, China
- Beijing Municipal Geriatric Medical Research Center, Beijing, China
| | - Tao Jiang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- The Chinese Glioma Genome Atlas, Beijing, China
- Beijing Neurosurgical Institute, Beijing, China
| | | | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Aging Biomarker Consortium, Beijing, China.
| | - Weiqi Zhang
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Aging Biomarker Consortium, Beijing, China.
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, China.
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China.
- Aging Biomarker Consortium, Beijing, China.
| |
Collapse
|
24
|
Vanherle S, Guns J, Loix M, Mingneau F, Dierckx T, Wouters F, Kuipers K, Vangansewinkel T, Wolfs E, Lins PP, Bronckaers A, Lambrichts I, Dehairs J, Swinnen JV, Verberk SGS, Haidar M, Hendriks JJA, Bogie JFJ. Extracellular vesicle-associated cholesterol supports the regenerative functions of macrophages in the brain. J Extracell Vesicles 2023; 12:e12394. [PMID: 38124258 PMCID: PMC10733568 DOI: 10.1002/jev2.12394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 11/15/2023] [Accepted: 11/23/2023] [Indexed: 12/23/2023] Open
Abstract
Macrophages play major roles in the pathophysiology of various neurological disorders, being involved in seemingly opposing processes such as lesion progression and resolution. Yet, the molecular mechanisms that drive their harmful and benign effector functions remain poorly understood. Here, we demonstrate that extracellular vesicles (EVs) secreted by repair-associated macrophages (RAMs) enhance remyelination ex vivo and in vivo by promoting the differentiation of oligodendrocyte precursor cells (OPCs). Guided by lipidomic analysis and applying cholesterol depletion and enrichment strategies, we find that EVs released by RAMs show markedly elevated cholesterol levels and that cholesterol abundance controls their reparative impact on OPC maturation and remyelination. Mechanistically, EV-associated cholesterol was found to promote OPC differentiation predominantly through direct membrane fusion. Collectively, our findings highlight that EVs are essential for cholesterol trafficking in the brain and that changes in cholesterol abundance support the reparative impact of EVs released by macrophages in the brain, potentially having broad implications for therapeutic strategies aimed at promoting repair in neurodegenerative disorders.
Collapse
Affiliation(s)
- Sam Vanherle
- Department of Immunology and Infection, Biomedical Research InstituteHasselt UniversityDiepenbeekBelgium
- University MS Center HasseltPeltBelgium
| | - Jeroen Guns
- Department of Immunology and Infection, Biomedical Research InstituteHasselt UniversityDiepenbeekBelgium
- University MS Center HasseltPeltBelgium
| | - Melanie Loix
- Department of Immunology and Infection, Biomedical Research InstituteHasselt UniversityDiepenbeekBelgium
- University MS Center HasseltPeltBelgium
| | - Fleur Mingneau
- Department of Immunology and Infection, Biomedical Research InstituteHasselt UniversityDiepenbeekBelgium
- University MS Center HasseltPeltBelgium
| | - Tess Dierckx
- Department of Immunology and Infection, Biomedical Research InstituteHasselt UniversityDiepenbeekBelgium
- University MS Center HasseltPeltBelgium
| | - Flore Wouters
- Department of Immunology and Infection, Biomedical Research InstituteHasselt UniversityDiepenbeekBelgium
- University MS Center HasseltPeltBelgium
| | - Koen Kuipers
- Department of Immunology and Infection, Biomedical Research InstituteHasselt UniversityDiepenbeekBelgium
- University MS Center HasseltPeltBelgium
| | - Tim Vangansewinkel
- Department of Cardio and Organs Systems, Biomedical Research InstituteHasselt UniversityDiepenbeekBelgium
- VIB, Center for Brain & Disease Research, Laboratory of NeurobiologyUniversity of LeuvenLeuvenBelgium
| | - Esther Wolfs
- Department of Cardio and Organs Systems, Biomedical Research InstituteHasselt UniversityDiepenbeekBelgium
| | - Paula Pincela Lins
- Department of Cardio and Organs Systems, Biomedical Research InstituteHasselt UniversityDiepenbeekBelgium
- Health DepartmentFlemish Institute for Technological ResearchMolBelgium
| | - Annelies Bronckaers
- Department of Cardio and Organs Systems, Biomedical Research InstituteHasselt UniversityDiepenbeekBelgium
| | - Ivo Lambrichts
- Department of Cardio and Organs Systems, Biomedical Research InstituteHasselt UniversityDiepenbeekBelgium
| | - Jonas Dehairs
- Department of Oncology, Laboratory of Lipid Metabolism and Cancer, Leuven Cancer InstituteUniversity of LeuvenLeuvenBelgium
| | - Johannes V. Swinnen
- Department of Oncology, Laboratory of Lipid Metabolism and Cancer, Leuven Cancer InstituteUniversity of LeuvenLeuvenBelgium
| | - Sanne G. S. Verberk
- Department of Immunology and Infection, Biomedical Research InstituteHasselt UniversityDiepenbeekBelgium
- University MS Center HasseltPeltBelgium
| | - Mansour Haidar
- Department of Immunology and Infection, Biomedical Research InstituteHasselt UniversityDiepenbeekBelgium
- University MS Center HasseltPeltBelgium
| | - Jerome J. A. Hendriks
- Department of Immunology and Infection, Biomedical Research InstituteHasselt UniversityDiepenbeekBelgium
- University MS Center HasseltPeltBelgium
| | - Jeroen F. J. Bogie
- Department of Immunology and Infection, Biomedical Research InstituteHasselt UniversityDiepenbeekBelgium
- University MS Center HasseltPeltBelgium
| |
Collapse
|
25
|
Jagielska A, Radzwill K, Espinosa-Hoyos D, Yang M, Kowsari K, Farley JE, Giera S, Byrne A, Sheng G, Fang NX, Dodge JC, Pedraza CE, Van Vliet KJ. Artificial axons as a biomimetic 3D myelination platform for the discovery and validation of promyelinating compounds. Sci Rep 2023; 13:19529. [PMID: 37945646 PMCID: PMC10636046 DOI: 10.1038/s41598-023-44675-6] [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: 02/24/2023] [Accepted: 10/11/2023] [Indexed: 11/12/2023] Open
Abstract
Multiple sclerosis (MS), a chronic neurodegenerative disease driven by damage to the protective myelin sheath, is currently incurable. Today, all clinically available treatments modulate the immune-mediated symptoms of the disease but they fail to stop neurodegeneration in many patients. Remyelination, the regenerative process of myelin repair by oligodendrocytes, which is considered a necessary step to protect demyelinated axons and stop neuronal death, is impaired in MS patients. One of the major obstacles to finding effective remyelinating drugs is the lack of biomimetic drug screening platforms that enable quantification of compounds' potential to stimulate 3D myelination in the physiologically relevant axon-like environment. To address this need, we built a unique myelination drug discovery platform, by expanding our previously developed technology, artificial axons (AAs), which enables 3D-printing of synthetic axon mimics with the geometry and mechanical properties closely resembling those of biological axons. This platform allows for high-throughput phenotypic myelination assay based on quantification of 3D wrapping of myelin membrane around axons in response to compounds. Here, we demonstrate quantification of 3D myelin wrapping by rat oligodendrocytes around the axon mimics in response to a small library of known pro-myelinating compounds. This assay shows pro-myelinating activity for all tested compounds consistent with the published in vitro and in vivo data, demonstrating predictive power of AA platform. We find that stimulation of myelin wrapping by these compounds is dose-dependent, providing a facile means to quantify the compounds' potency and efficacy in promoting myelin wrapping. Further, the ranking of relative efficacy among these compounds differs in this 3D axon-like environment as compared to a traditional oligodendrocyte 2D differentiation assay quantifying area of deposited myelin membrane. Together, we demonstrate that the artificial axons platform and associated phenotypic myelin wrapping assay afford direct evaluation of myelin wrapping by oligodendrocytes in response to soluble compounds in an axon-like environment, providing a predictive tool for the discovery of remyelinating therapies.
Collapse
Affiliation(s)
- Anna Jagielska
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Artificial Axon Labs, Boston, MA, USA.
| | | | - Daniela Espinosa-Hoyos
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Sanofi, Cambridge, MA, USA
| | - Mingyu Yang
- Harvard-MIT Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kavin Kowsari
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Merck, Rahway, NJ, USA
| | - Jonathan E Farley
- Sanofi, Cambridge, MA, USA
- Alnylam Pharmaceuticals, Cambridge, MA, USA
| | | | | | | | - Nicholas X Fang
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- The University of Hong Kong, Hong Kong, China
| | | | | | - Krystyn J Van Vliet
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Cornell University, Ithaca, NY, USA.
| |
Collapse
|
26
|
Zhang R, Wang Y, Wu A, Wang J, Zhang J. Strategies of targeting CYP51 for IFIs therapy: Emerging prospects, opportunities and challenges. Eur J Med Chem 2023; 259:115658. [PMID: 37480712 DOI: 10.1016/j.ejmech.2023.115658] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/08/2023] [Accepted: 07/14/2023] [Indexed: 07/24/2023]
Abstract
CYP51, a monooxygenase associated with the sterol synthesis pathway, is responsible for the catalysis of the 14-methyl hydroxylation reaction of lanosterol precursors. This enzyme is widely present in microorganisms, plants, and mammals. In mammals, CYP51 plays a role in cholesterol production, oligodendrocyte formation, oocyte maturation, and spermatogenesis. In fungal cells, CYP51 is an enzyme that synthesizes membrane sterols. By inhibiting fungal CYP51, ergosterol synthesis can be inhibited and ergosterol membrane fluidity is altered, resulting in fungal cell apoptosis. Thus, targeting CYP51 is a reliable antifungal strategy with important implications for the treatment of invasive fungal infections (IFIs). Many CYP51 inhibitors have been approved by the FDA for clinical treatment. However, several limitations of CYP51 inhibitors remain to be resolved, including fungal resistance, hepatotoxicity, and drug-drug interactions. New broad-spectrum, anti-resistant, highly selective CYP51 inhibitors are expected to be developed to enhance clinical efficacy and minimize adverse effects. Herein, we summarize the structural features and biological functions of CYP51 and emphatically analyze the structure-activity relationship (SAR) and therapeutic potential of different chemical types of small-molecule CYP51 inhibitors. We also discuss the latest progress of novel strategies, providing insights into new drugs targeting CYP51 for clinical practice.
Collapse
Affiliation(s)
- Ruofei Zhang
- Department of Neurology, Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yuxi Wang
- Department of Neurology, Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Aijia Wu
- Department of Neurology, Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jiaxing Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, 38163, Tennessee, United States
| | - Jifa Zhang
- Department of Neurology, Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
27
|
Tu WB, Christofk HR, Plath K. Nutrient regulation of development and cell fate decisions. Development 2023; 150:dev199961. [PMID: 37260407 PMCID: PMC10281554 DOI: 10.1242/dev.199961] [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] [Indexed: 06/02/2023]
Abstract
Diet contributes to health at all stages of life, from embryonic development to old age. Nutrients, including vitamins, amino acids, lipids and sugars, have instructive roles in directing cell fate and function, maintaining stem cell populations, tissue homeostasis and alleviating the consequences of aging. This Review highlights recent findings that illuminate how common diets and specific nutrients impact cell fate decisions in healthy and disease contexts. We also draw attention to new models, technologies and resources that help to address outstanding questions in this emerging field and may lead to dietary approaches that promote healthy development and improve disease treatments.
Collapse
Affiliation(s)
- William B. Tu
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Heather R. Christofk
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Kathrin Plath
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
28
|
Chen K, Cambi F, Kozai TDY. Pro-myelinating clemastine administration improves recording performance of chronically implanted microelectrodes and nearby neuronal health. Biomaterials 2023; 301:122210. [PMID: 37413842 PMCID: PMC10528716 DOI: 10.1016/j.biomaterials.2023.122210] [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: 02/06/2023] [Revised: 06/08/2023] [Accepted: 06/19/2023] [Indexed: 07/08/2023]
Abstract
Intracortical microelectrodes have become a useful tool in neuroprosthetic applications in the clinic and to understand neurological disorders in basic neurosciences. Many of these brain-machine interface technology applications require successful long-term implantation with high stability and sensitivity. However, the intrinsic tissue reaction caused by implantation remains a major failure mechanism causing loss of recorded signal quality over time. Oligodendrocytes remain an underappreciated intervention target to improve chronic recording performance. These cells can accelerate action potential propagation and provides direct metabolic support for neuronal health and functionality. However, implantation injury causes oligodendrocyte degeneration and leads to progressive demyelination in surrounding brain tissue. Previous work highlighted that healthy oligodendrocytes are necessary for greater electrophysiological recording performance and the prevention of neuronal silencing around implanted microelectrodes over the chronic implantation period. Thus, we hypothesize that enhancing oligodendrocyte activity with a pharmaceutical drug, Clemastine, will prevent the chronic decline of microelectrode recording performance. Electrophysiological evaluation showed that the promyelination Clemastine treatment significantly elevated the signal detectability and quality, rescued the loss of multi-unit activity, and increased functional interlaminar connectivity over 16-weeks of implantation. Additionally, post-mortem immunohistochemistry showed that increased oligodendrocyte density and myelination coincided with increased survival of both excitatory and inhibitory neurons near the implant. Overall, we showed a positive relationship between enhanced oligodendrocyte activity and neuronal health and functionality near the chronically implanted microelectrode. This study shows that therapeutic strategy that enhance oligodendrocyte activity is effective for integrating the functional device interface with brain tissue over chronic implantation period.
Collapse
Affiliation(s)
- Keying Chen
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, Pittsburgh, PA, USA
| | - Franca Cambi
- Veterans Administration Pittsburgh, Pittsburgh, PA, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Takashi D Y Kozai
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, Pittsburgh, PA, USA; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA; McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA; NeuroTech Center, University of Pittsburgh Brain Institute, Pittsburgh, PA, USA.
| |
Collapse
|
29
|
Sun MA, Yang R, Liu H, Wang W, Song X, Hu B, Reynolds N, Roso K, Chen LH, Greer PK, Keir ST, McLendon RE, Cheng SY, Bigner DD, Ashley DM, Pirozzi CJ, He Y. Repurposing Clemastine to Target Glioblastoma Cell Stemness. Cancers (Basel) 2023; 15:4619. [PMID: 37760589 PMCID: PMC10526458 DOI: 10.3390/cancers15184619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 09/08/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Brain tumor-initiating cells (BTICs) and tumor cell plasticity promote glioblastoma (GBM) progression. Here, we demonstrate that clemastine, an over-the-counter drug for treating hay fever and allergy symptoms, effectively attenuated the stemness and suppressed the propagation of primary BTIC cultures bearing PDGFRA amplification. These effects on BTICs were accompanied by altered gene expression profiling indicative of their more differentiated states, resonating with the activity of clemastine in promoting the differentiation of normal oligodendrocyte progenitor cells (OPCs) into mature oligodendrocytes. Functional assays for pharmacological targets of clemastine revealed that the Emopamil Binding Protein (EBP), an enzyme in the cholesterol biosynthesis pathway, is essential for BTIC propagation and a target that mediates the suppressive effects of clemastine. Finally, we showed that a neural stem cell-derived mouse glioma model displaying predominantly proneural features was similarly susceptible to clemastine treatment. Collectively, these results identify pathways essential for maintaining the stemness and progenitor features of GBMs, uncover BTIC dependency on EBP, and suggest that non-oncology, low-toxicity drugs with OPC differentiation-promoting activity can be repurposed to target GBM stemness and aid in their treatment.
Collapse
Affiliation(s)
- Michael A. Sun
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA; (M.A.S.); (R.Y.); (H.L.); (W.W.); (N.R.); (K.R.); (L.H.C.); (P.K.G.); (S.T.K.); (R.E.M.); (D.D.B.); (D.M.A.)
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
- Pathology Graduate Program, Duke University Medical Center, Durham, NC 27710, USA
| | - Rui Yang
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA; (M.A.S.); (R.Y.); (H.L.); (W.W.); (N.R.); (K.R.); (L.H.C.); (P.K.G.); (S.T.K.); (R.E.M.); (D.D.B.); (D.M.A.)
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Heng Liu
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA; (M.A.S.); (R.Y.); (H.L.); (W.W.); (N.R.); (K.R.); (L.H.C.); (P.K.G.); (S.T.K.); (R.E.M.); (D.D.B.); (D.M.A.)
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
- Pathology Graduate Program, Duke University Medical Center, Durham, NC 27710, USA
| | - Wenzhe Wang
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA; (M.A.S.); (R.Y.); (H.L.); (W.W.); (N.R.); (K.R.); (L.H.C.); (P.K.G.); (S.T.K.); (R.E.M.); (D.D.B.); (D.M.A.)
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Xiao Song
- The Ken & Ruth Davee Department of Neurology, Lou and Jean Malnati Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; (X.S.); (B.H.); (S.-Y.C.)
| | - Bo Hu
- The Ken & Ruth Davee Department of Neurology, Lou and Jean Malnati Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; (X.S.); (B.H.); (S.-Y.C.)
| | - Nathan Reynolds
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA; (M.A.S.); (R.Y.); (H.L.); (W.W.); (N.R.); (K.R.); (L.H.C.); (P.K.G.); (S.T.K.); (R.E.M.); (D.D.B.); (D.M.A.)
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Kristen Roso
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA; (M.A.S.); (R.Y.); (H.L.); (W.W.); (N.R.); (K.R.); (L.H.C.); (P.K.G.); (S.T.K.); (R.E.M.); (D.D.B.); (D.M.A.)
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Lee H. Chen
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA; (M.A.S.); (R.Y.); (H.L.); (W.W.); (N.R.); (K.R.); (L.H.C.); (P.K.G.); (S.T.K.); (R.E.M.); (D.D.B.); (D.M.A.)
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Paula K. Greer
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA; (M.A.S.); (R.Y.); (H.L.); (W.W.); (N.R.); (K.R.); (L.H.C.); (P.K.G.); (S.T.K.); (R.E.M.); (D.D.B.); (D.M.A.)
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Stephen T. Keir
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA; (M.A.S.); (R.Y.); (H.L.); (W.W.); (N.R.); (K.R.); (L.H.C.); (P.K.G.); (S.T.K.); (R.E.M.); (D.D.B.); (D.M.A.)
- Department of Neurosurgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Roger E. McLendon
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA; (M.A.S.); (R.Y.); (H.L.); (W.W.); (N.R.); (K.R.); (L.H.C.); (P.K.G.); (S.T.K.); (R.E.M.); (D.D.B.); (D.M.A.)
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
- Department of Neurosurgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Shi-Yuan Cheng
- The Ken & Ruth Davee Department of Neurology, Lou and Jean Malnati Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; (X.S.); (B.H.); (S.-Y.C.)
| | - Darell D. Bigner
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA; (M.A.S.); (R.Y.); (H.L.); (W.W.); (N.R.); (K.R.); (L.H.C.); (P.K.G.); (S.T.K.); (R.E.M.); (D.D.B.); (D.M.A.)
- Department of Neurosurgery, Duke University Medical Center, Durham, NC 27710, USA
| | - David M. Ashley
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA; (M.A.S.); (R.Y.); (H.L.); (W.W.); (N.R.); (K.R.); (L.H.C.); (P.K.G.); (S.T.K.); (R.E.M.); (D.D.B.); (D.M.A.)
- Department of Neurosurgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Christopher J. Pirozzi
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA; (M.A.S.); (R.Y.); (H.L.); (W.W.); (N.R.); (K.R.); (L.H.C.); (P.K.G.); (S.T.K.); (R.E.M.); (D.D.B.); (D.M.A.)
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Yiping He
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA; (M.A.S.); (R.Y.); (H.L.); (W.W.); (N.R.); (K.R.); (L.H.C.); (P.K.G.); (S.T.K.); (R.E.M.); (D.D.B.); (D.M.A.)
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
30
|
Yin J, Fu J, Shao Y, Xu J, Li H, Chen C, Zhao Y, Zheng Z, Yu C, Zheng L, Wang B. CYP51-mediated cholesterol biosynthesis is required for the proliferation of CD4 + T cells in Sjogren's syndrome. Clin Exp Med 2023; 23:1691-1711. [PMID: 36413274 DOI: 10.1007/s10238-022-00939-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 11/02/2022] [Indexed: 11/23/2022]
Abstract
CYtochrome P450, family 51 (CYP51) is an important enzyme for de novo cholesterol synthesis in mammalian cells. In the present study, we found that the expression of CYP51 positively correlated with CD4+ T cell activation both in vivo and in vitro. The addition of ketoconazole, a pharmacological inhibitor of CYP51, prevented the proliferation and activation of anti-CD3/CD28-expanded mouse CD4+ T cells in a dose-dependent fashion. Liquid chromatography-tandem mass spectrometry indicated an increase in levels of lanosterol in T cells treated with ketoconazole during activation. Ketoconazole-induced blockade of the cholesterol synthesis pathway also caused Sterol regulatory element binding protein 2 (SREBP2) activation in CD4+ T cells. Additionally, ketoconazole treatment elicited an integrated stress response in T cells that up-regulated activating transcription factor 4 (ATF4) and DNA-damage inducible transcript 3 (DDIT3/CHOP) at the translational level. Furthermore, treatment with ketoconazole significantly decreased the amount of CD4+ T cells infiltrating lesions in the submandibular glands of NOD/Ltj mice. In summary, our results suggest that CYP51 plays an essential role in the proliferation and survival of CD4+ T cells, which makes ketoconazole an inhibitor of CD4+ T cell proliferation and of the SS-like autoimmune response through regulating the biosynthesis of cholesterol and inducing the integrated stress response.
Collapse
Affiliation(s)
- Junhao Yin
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology & National Clinical Research Center for Oral Disease, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Jiayao Fu
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology & National Clinical Research Center for Oral Disease, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Yanxiong Shao
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology & National Clinical Research Center for Oral Disease, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Jiabao Xu
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology & National Clinical Research Center for Oral Disease, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Hui Li
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology & National Clinical Research Center for Oral Disease, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Changyu Chen
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology & National Clinical Research Center for Oral Disease, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Yijie Zhao
- Department of Oral and Maxillofacial Surgery, Shanghai Stomatological Hospital, Fudan University, 1258 Fuxin Zhong Road, Shanghai, China
| | - Zhanglong Zheng
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Chuangqi Yu
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology & National Clinical Research Center for Oral Disease, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Lingyan Zheng
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China.
- National Center for Stomatology & National Clinical Research Center for Oral Disease, Shanghai, China.
- Shanghai Key Laboratory of Stomatology, Shanghai, China.
| | - Baoli Wang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China.
- National Center for Stomatology & National Clinical Research Center for Oral Disease, Shanghai, China.
- Shanghai Key Laboratory of Stomatology, Shanghai, China.
| |
Collapse
|
31
|
Battis K, Xiang W, Winkler J. The Bidirectional Interplay of α-Synuclein with Lipids in the Central Nervous System and Its Implications for the Pathogenesis of Parkinson's Disease. Int J Mol Sci 2023; 24:13270. [PMID: 37686080 PMCID: PMC10487772 DOI: 10.3390/ijms241713270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
The alteration and aggregation of alpha-synuclein (α-syn) play a crucial role in neurodegenerative diseases collectively termed as synucleinopathies, including Parkinson's disease (PD). The bidirectional interaction of α-syn with lipids and biomembranes impacts not only α-syn aggregation but also lipid homeostasis. Indeed, lipid composition and metabolism are severely perturbed in PD. One explanation for lipid-associated alterations may involve structural changes in α-syn, caused, for example, by missense mutations in the lipid-binding region of α-syn as well as post-translational modifications such as phosphorylation, acetylation, nitration, ubiquitination, truncation, glycosylation, and glycation. Notably, different strategies targeting the α-syn-lipid interaction have been identified and are able to reduce α-syn pathology. These approaches include the modulation of post-translational modifications aiming to reduce the aggregation of α-syn and modify its binding properties to lipid membranes. Furthermore, targeting enzymes involved in various steps of lipid metabolism and exploring the neuroprotective potential of lipids themselves have emerged as novel therapeutic approaches. Taken together, this review focuses on the bidirectional crosstalk of α-syn and lipids and how alterations of this interaction affect PD and thereby open a window for therapeutic interventions.
Collapse
Affiliation(s)
| | | | - Jürgen Winkler
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany; (K.B.); (W.X.)
| |
Collapse
|
32
|
Clayton BL, Barbar L, Sapar M, Rusielewicz T, Kalpana K, Migliori B, Paull D, Brenner K, Moroziewicz D, Sand IK, Casaccia P, Tesar PJ, Fossati V. Patient iPSC models reveal glia-intrinsic phenotypes in multiple sclerosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.01.551553. [PMID: 37577713 PMCID: PMC10418164 DOI: 10.1101/2023.08.01.551553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Multiple sclerosis (MS) is considered an inflammatory and neurodegenerative disease of the central nervous system, typically resulting in significant neurological disability that worsens over time. While considerable progress has been made in defining the immune system's role in MS pathophysiology, the contribution of intrinsic CNS-cell dysfunction remains unclear. Here, we generated the largest reported collection of iPSC lines from people with MS spanning diverse clinical subtypes and differentiated them into glia-enriched cultures. Using single-cell transcriptomic profiling, we observed several distinguishing characteristics of MS cultures pointing to glia-intrinsic disease mechanisms. We found that iPSC-derived cultures from people with primary progressive MS contained fewer oligodendrocytes. Moreover, iPSC-oligodendrocyte lineage cells and astrocytes from people with MS showed increased expression of immune and inflammatory genes that match those of glial cells from MS postmortem brains. Thus, iPSC-derived MS models provide a unique platform for dissecting glial contributions to disease phenotypes independent of the peripheral immune system and identify potential glia-specific targets for therapeutic intervention.
Collapse
Affiliation(s)
- Benjamin L.L. Clayton
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
- These authors contributed equally
| | - Lilianne Barbar
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
- Current affiliation: Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, 63105, USA
- These authors contributed equally
| | - Maria Sapar
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Tomasz Rusielewicz
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Kriti Kalpana
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Bianca Migliori
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | | | - Daniel Paull
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Katie Brenner
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Dorota Moroziewicz
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Ilana Katz Sand
- Corinne Goldsmith Dickinson Center for Multiple Sclerosis, Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10129, USA
| | | | - Paul J. Tesar
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Valentina Fossati
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| |
Collapse
|
33
|
Dominicis A, Del Giovane A, Torreggiani M, Recchia AD, Ciccarone F, Ciriolo MR, Ragnini-Wilson A. N-Acetylaspartate Drives Oligodendroglial Differentiation via Histone Deacetylase Activation. Cells 2023; 12:1861. [PMID: 37508525 PMCID: PMC10378218 DOI: 10.3390/cells12141861] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/09/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
An unmet clinical goal in demyelinating pathologies is to restore the myelin sheath prior to neural degeneration. N-acetylaspartate (NAA) is an acetylated derivative form of aspartate, abundant in the healthy brain but severely reduced during traumatic brain injury and in patients with neurodegenerative pathologies. How extracellular NAA variations impact the remyelination process and, thereby, the ability of oligodendrocytes to remyelinate axons remains unexplored. Here, we evaluated the remyelination properties of the oligodendroglial (OL) mouse cell line Oli-neuM under different concentrations of NAA using a combination of biochemical, qPCR, immunofluorescence assays, and in vitro engagement tests, at NAA doses compatible with those observed in healthy brains and during brain injury. We observed that oligodendroglia cells respond to decreasing levels of NAA by stimulating differentiation and promoting gene expression of myelin proteins in a temporally regulated manner. Low doses of NAA potently stimulate Oli-neuM to engage with synthetic axons. Furthermore, we show a concentration-dependent expression of specific histone deacetylases essential for MBP gene expression under NAA or Clobetasol treatment. These data are consistent with the idea that oligodendrocytes respond to lowering the NAA concentration by activating the remyelination process via deacetylase activation.
Collapse
Affiliation(s)
| | - Alice Del Giovane
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Matteo Torreggiani
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | | | - Fabio Ciccarone
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
- IRCCS San Raffaele, 00166 Rome, Italy
| | - Maria Rosa Ciriolo
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
- IRCCS San Raffaele, 00166 Rome, Italy
| | | |
Collapse
|
34
|
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
|
35
|
Scavuzzo MA, Letai KC, Maeno-Hikichi Y, Wulftange WJ, Shah IK, Rameshbabu JS, Tomar A, Shick HE, Shah AK, Xiong Y, Cohn EF, Allan KC, Tesar PJ. Enteric glial hub cells coordinate intestinal motility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.07.544052. [PMID: 37333182 PMCID: PMC10274798 DOI: 10.1101/2023.06.07.544052] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Enteric glia are the predominant cell type in the enteric nervous system yet their identities and roles in gastrointestinal function are not well classified. Using our optimized single nucleus RNA-sequencing method, we identified distinct molecular classes of enteric glia and defined their morphological and spatial diversity. Our findings revealed a functionally specialized biosensor subtype of enteric glia that we call "hub cells." Deletion of the mechanosensory ion channel PIEZO2 from adult enteric glial hub cells, but not other subtypes of enteric glia, led to defects in intestinal motility and gastric emptying in mice. These results provide insight into the multifaceted functions of different enteric glial cell subtypes in gut health and emphasize that therapies targeting enteric glia could advance the treatment of gastrointestinal diseases.
Collapse
Affiliation(s)
- Marissa A. Scavuzzo
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Katherine C. Letai
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Yuka Maeno-Hikichi
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - William J. Wulftange
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Isha K. Shah
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Jeyashri S. Rameshbabu
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Alka Tomar
- 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
| | - Aakash K. Shah
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Ying Xiong
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Erin F. Cohn
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Kevin C. Allan
- 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
| |
Collapse
|
36
|
Molina-Gonzalez I, Holloway RK, Jiwaji Z, Dando O, Kent SA, Emelianova K, Lloyd AF, Forbes LH, Mahmood A, Skripuletz T, Gudi V, Febery JA, Johnson JA, Fowler JH, Kuhlmann T, Williams A, Chandran S, Stangel M, Howden AJM, Hardingham GE, Miron VE. Astrocyte-oligodendrocyte interaction regulates central nervous system regeneration. Nat Commun 2023; 14:3372. [PMID: 37291151 PMCID: PMC10250470 DOI: 10.1038/s41467-023-39046-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 05/18/2023] [Indexed: 06/10/2023] Open
Abstract
Failed regeneration of myelin around neuronal axons following central nervous system damage contributes to nerve dysfunction and clinical decline in various neurological conditions, for which there is an unmet therapeutic demand. Here, we show that interaction between glial cells - astrocytes and mature myelin-forming oligodendrocytes - is a determinant of remyelination. Using in vivo/ ex vivo/ in vitro rodent models, unbiased RNA sequencing, functional manipulation, and human brain lesion analyses, we discover that astrocytes support the survival of regenerating oligodendrocytes, via downregulation of the Nrf2 pathway associated with increased astrocytic cholesterol biosynthesis pathway activation. Remyelination fails following sustained astrocytic Nrf2 activation in focally-lesioned male mice yet is restored by either cholesterol biosynthesis/efflux stimulation, or Nrf2 inhibition using the existing therapeutic Luteolin. We identify that astrocyte-oligodendrocyte interaction regulates remyelination, and reveal a drug strategy for central nervous system regeneration centred on targeting this interaction.
Collapse
Affiliation(s)
- Irene Molina-Gonzalez
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh Medical School, Edinburgh, EH16 4TJ, UK
- United Kingdom Multiple Sclerosis Society Edinburgh Centre for Multiple Sclerosis Research, University of Edinburgh, Edinburgh, EH16 4TJ, UK
- Center for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK
- Medical Research Council Centre for Reproductive Health, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Rebecca K Holloway
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh Medical School, Edinburgh, EH16 4TJ, UK
- United Kingdom Multiple Sclerosis Society Edinburgh Centre for Multiple Sclerosis Research, University of Edinburgh, Edinburgh, EH16 4TJ, UK
- Center for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK
- Medical Research Council Centre for Reproductive Health, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Zoeb Jiwaji
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh Medical School, Edinburgh, EH16 4TJ, UK
- Center for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - Owen Dando
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh Medical School, Edinburgh, EH16 4TJ, UK
- Center for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - Sarah A Kent
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh Medical School, Edinburgh, EH16 4TJ, UK
- United Kingdom Multiple Sclerosis Society Edinburgh Centre for Multiple Sclerosis Research, University of Edinburgh, Edinburgh, EH16 4TJ, UK
- Center for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK
- Wellcome Trust Translational Neuroscience PhD programme, Edinburgh, UK
| | - Katie Emelianova
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh Medical School, Edinburgh, EH16 4TJ, UK
- Center for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - Amy F Lloyd
- Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| | - Lindsey H Forbes
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh Medical School, Edinburgh, EH16 4TJ, UK
- United Kingdom Multiple Sclerosis Society Edinburgh Centre for Multiple Sclerosis Research, University of Edinburgh, Edinburgh, EH16 4TJ, UK
- Center for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK
- Medical Research Council Centre for Reproductive Health, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Ayisha Mahmood
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh Medical School, Edinburgh, EH16 4TJ, UK
- United Kingdom Multiple Sclerosis Society Edinburgh Centre for Multiple Sclerosis Research, University of Edinburgh, Edinburgh, EH16 4TJ, UK
- Center for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK
- Medical Research Council Centre for Reproductive Health, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Thomas Skripuletz
- Department of Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Medizinische Hochschule Hannover, Hannover, 30625, Germany
| | - Viktoria Gudi
- Department of Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Medizinische Hochschule Hannover, Hannover, 30625, Germany
| | - James A Febery
- Center for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - Jeffrey A Johnson
- Division of Pharmaceutical Sciences, University of Wisconsin, Madison, WI, 53705, USA
- Molecular and Environmental Toxicology Centre, University of Wisconsin, Madison, WI, 53706, USA
- Center for Neuroscience, University of Wisconsin, Madison, WI, 53705, USA
- Waisman Centre, University of Wisconsin, Madison, WI, 53705, USA
| | - Jill H Fowler
- Center for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - Tanja Kuhlmann
- Institute of Neuropathology, University Hospital Muenster, Muenster, D-48129, Germany
| | - Anna Williams
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh Medical School, Edinburgh, EH16 4TJ, UK
- United Kingdom Multiple Sclerosis Society Edinburgh Centre for Multiple Sclerosis Research, University of Edinburgh, Edinburgh, EH16 4TJ, UK
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, EH16 5UU, UK
| | - Siddharthan Chandran
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh Medical School, Edinburgh, EH16 4TJ, UK
- United Kingdom Multiple Sclerosis Society Edinburgh Centre for Multiple Sclerosis Research, University of Edinburgh, Edinburgh, EH16 4TJ, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - Martin Stangel
- Department of Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Medizinische Hochschule Hannover, Hannover, 30625, Germany
| | - Andrew J M Howden
- Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| | - Giles E Hardingham
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh Medical School, Edinburgh, EH16 4TJ, UK
- United Kingdom Multiple Sclerosis Society Edinburgh Centre for Multiple Sclerosis Research, University of Edinburgh, Edinburgh, EH16 4TJ, UK
- Center for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - Veronique E Miron
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh Medical School, Edinburgh, EH16 4TJ, UK.
- United Kingdom Multiple Sclerosis Society Edinburgh Centre for Multiple Sclerosis Research, University of Edinburgh, Edinburgh, EH16 4TJ, UK.
- Center for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK.
- Medical Research Council Centre for Reproductive Health, University of Edinburgh, Edinburgh, EH16 4TJ, UK.
- BARLO Multiple Sclerosis Centre, St.Michael's Hospital, Toronto, ON, M5B 1W8, Canada.
- Keenan Centre for Biomedical Research at St.Michael's Hospital, Toronto, ON, M5B 1T8, Canada.
- Department of Immunology, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
37
|
Watson LA, Meharena HS. From neurodevelopment to neurodegeneration: utilizing human stem cell models to gain insight into Down syndrome. Front Genet 2023; 14:1198129. [PMID: 37323671 PMCID: PMC10267712 DOI: 10.3389/fgene.2023.1198129] [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: 03/31/2023] [Accepted: 05/09/2023] [Indexed: 06/17/2023] Open
Abstract
Down syndrome (DS), caused by triplication of chromosome 21, is the most frequent aneuploidy observed in the human population and represents the most common genetic form of intellectual disability and early-onset Alzheimer's disease (AD). Individuals with DS exhibit a wide spectrum of clinical presentation, with a number of organs implicated including the neurological, immune, musculoskeletal, cardiac, and gastrointestinal systems. Decades of DS research have illuminated our understanding of the disorder, however many of the features that limit quality of life and independence of individuals with DS, including intellectual disability and early-onset dementia, remain poorly understood. This lack of knowledge of the cellular and molecular mechanisms leading to neurological features of DS has caused significant roadblocks in developing effective therapeutic strategies to improve quality of life for individuals with DS. Recent technological advances in human stem cell culture methods, genome editing approaches, and single-cell transcriptomics have provided paradigm-shifting insights into complex neurological diseases such as DS. Here, we review novel neurological disease modeling approaches, how they have been used to study DS, and what questions might be addressed in the future using these innovative tools.
Collapse
Affiliation(s)
- L. Ashley Watson
- Developmental and Cognitive Genomics Research Laboratory, Division of Biological Sciences, Section of Neurobiology, University of California, San Diego, La Jolla, CA, United States
| | - Hiruy S. Meharena
- Developmental and Cognitive Genomics Research Laboratory, Division of Biological Sciences, Section of Neurobiology, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
38
|
Jansen MI, Castorina A. Identification of Key Genes and Regulatory Pathways in Multiple Sclerosis Brain Samples: A Meta-Analysis of Micro-Array Datasets. Int J Mol Sci 2023; 24:ijms24119361. [PMID: 37298310 DOI: 10.3390/ijms24119361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 05/11/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disorder of the central nervous system (CNS) whose aetiology is only partly understood. Investigating the intricate transcriptional changes occurring in MS brains is critical to unravel novel pathogenic mechanisms and therapeutic targets. Unfortunately, this process is often hindered by the difficulty in retrieving an adequate number of samples. However, by merging data from publicly available datasets, it is possible to identify alterations in gene expression profiles and regulatory pathways that were previously overlooked. Here, we merged microarray gene expression profiles obtained from CNS white matter samples taken from MS donors to identify novel differentially expressed genes (DEGs) linked with MS. Data from three independent datasets (GSE38010, GSE32915, and GSE108000) were combined and used to detect novel DEGs using the Stouffer's Z-score method. Corresponding regulatory pathways were analysed using the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway databases. Finally, top up- and down-regulated transcripts were validated by real-time quantitative PCR (qPCR) using an independent set of white matter tissue samples obtained from MS donors with different disease subtypes. There were a total of 1446 DEGs, of which 742 were up-regulated and 704 genes were down-regulated. DEGs were associated with several myelin-related pathways and protein metabolism pathways. Validation studies of selected top up- or down-regulated genes highlighted MS subtype-specific differences in the expression of some of the identified genes, underlining a more complex scenario of white matter pathology amongst people afflicted by this devastating disease.
Collapse
Affiliation(s)
- Margo I Jansen
- Laboratory of Cellular and Molecular Neuroscience (LCMN), School of Life Sciences, Faculty of Science, University of Technology Sydney, P.O. Box 123, Broadway, NSW 2007, Australia
| | - Alessandro Castorina
- Laboratory of Cellular and Molecular Neuroscience (LCMN), School of Life Sciences, Faculty of Science, University of Technology Sydney, P.O. Box 123, Broadway, NSW 2007, Australia
| |
Collapse
|
39
|
Qian L, Scott NA, Capell-Hattam IM, Draper EA, Fenton NM, Luu W, Sharpe LJ, Brown AJ. Cholesterol synthesis enzyme SC4MOL is fine-tuned by sterols and targeted for degradation by the E3 ligase MARCHF6. J Lipid Res 2023; 64:100362. [PMID: 36958722 PMCID: PMC10176258 DOI: 10.1016/j.jlr.2023.100362] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 02/09/2023] [Accepted: 02/25/2023] [Indexed: 03/25/2023] Open
Abstract
Cholesterol biosynthesis is a highly regulated pathway, with over 20 enzymes controlled at the transcriptional and post-translational level. Whilst some enzymes remain stable, increased sterol levels can trigger degradation of several synthesis enzymes via the ubiquitin-proteasome system. Of note, we previously identified four cholesterol synthesis enzymes as substrates for one E3 ubiquitin ligase, membrane-associated RING-CH-type finger 6 (MARCHF6). Whether MARCHF6 targets the cholesterol synthesis pathway at other points is unknown. In addition, the post-translational regulation of many cholesterol synthesis enzymes, including the C4-demethylation complex (sterol-C4-methyl oxidase-like, SC4MOL; NAD(P) dependent steroid dehydrogenase-like, NSDHL; hydroxysteroid 17-beta dehydrogenase, HSD17B7) is largely uncharacterized. Using cultured mammalian cell-lines (human-derived and Chinese Hamster Ovary cells), we show SC4MOL, the first acting enzyme of C4-demethylation, is a MARCHF6 substrate, and is rapidly turned over and sensitive to sterols. Sterol depletion stabilizes SC4MOL protein levels, whilst sterol excess downregulates both transcript and protein levels. Furthermore, we found SC4MOL depletion by siRNA results in a significant decrease in total cell cholesterol. Thus, our work indicates SC4MOL is the most regulated enzyme in the C4-demethylation complex. Our results further implicate MARCHF6 as a crucial post-translational regulator of cholesterol synthesis, with this E3 ubiquitin ligase controlling levels of at least five enzymes of the pathway.
Collapse
Affiliation(s)
- Lydia Qian
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, New South Wales 2052, Australia
| | - Nicola A Scott
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, New South Wales 2052, Australia
| | - Isabelle M Capell-Hattam
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, New South Wales 2052, Australia
| | - Eliza A Draper
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, New South Wales 2052, Australia
| | - Nicole M Fenton
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, New South Wales 2052, Australia
| | - Winnie Luu
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, New South Wales 2052, Australia
| | - Laura J Sharpe
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, New South Wales 2052, Australia
| | - Andrew J Brown
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, New South Wales 2052, Australia.
| |
Collapse
|
40
|
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: 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: 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
|
41
|
Wang J, Shi R, Yang Q, Chen Z, Wang J, Gong Z, Chen S, Wang N. Characterization and potential function of 7-dehydrocholesterol reductase (dhcr7) and lathosterol 5-desaturase (sc5d) in Cynoglossus semilaevis sexual size dimorphism. Gene X 2023; 853:147089. [PMID: 36470484 DOI: 10.1016/j.gene.2022.147089] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 11/17/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022] Open
Abstract
The typical sexual size dimorphism (SSD) phenomenon of Chinese tongue sole (Cynoglossus semilaevis) seriously restricts the sustainable development of the fishing industry. Previous transcriptome analysis has found a close relationship between the steroid biosynthesis and C. semilaevis SSD. The 7-dehydrocholesterol reductase (dhcr7) and lathosterol 5-desaturase (sc5d) are two genes in the steroid biosynthesis pathway, playing important roles in lipid synthesis, cellular metabolism, and growth. The present study assessed their roles in the mechanism of C. semilaevis SSD. The quantitative polymerase chain reaction (qPCR) results showed that C. semilaevis dhcr7 was mainly expressed in female livers, and C. semilaevis sc5d was highly expressed in female livers and gonads. Dual-luciferase experiment showed that dhcr7 and sc5d promoters had strong transcriptional activity. The transcription factors E2F transcription factor 1 (E2F1), and CCAAT enhancer binding protein alpha (C/EBPα) significantly regulated the transcriptional activity of dhcr7 and sc5d promoters, respectively. Furthermore, small interfering RNA (siRNA) knockdown results showed that expression levels of several genes [SREBF chaperone (scap), membrane-bound transcription factor peptidase, site 1 (mbtps1), fatty acid synthase (fasn), sonic hedgehog (shh), bone morphogenetic protein 2b (bmp2b) and AKT serine/threonine kinase 1 (akt1)] were suppressed. Protein subcellular localization results indicated that Dhcr7 and Sc5d were both specifically distributed in the cytoplasm, with co-localization been observed. The present study provides evidence that dhcr7 and sc5d might regulate C. semilaevis sexual size dimorphism by involving in energy homeostasis and cell cycle, or by affecting PI3K-Akt and Shh signaling pathways. The detailed roles of these steroid biosynthesis genes regulating C. semilaevis SSD needed more information.
Collapse
Affiliation(s)
- Jialin Wang
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Pilot National Laboratory for Marine Science and Technology, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, China
| | - Rui Shi
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Pilot National Laboratory for Marine Science and Technology, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, China
| | - Qian Yang
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Pilot National Laboratory for Marine Science and Technology, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, China
| | - Zhangfan Chen
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Pilot National Laboratory for Marine Science and Technology, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; Key Laboratory for Sustainable Development of Marine Fisheries, Ministry of Agriculture and Rural Affairs, Qingdao 266071, China; Shandong Key Laboratory of Marine Fisheries Biotechnology and Genetic Breeding, Qingdao 266071, China
| | - Jiacheng Wang
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Pilot National Laboratory for Marine Science and Technology, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, China
| | - Zhihong Gong
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Pilot National Laboratory for Marine Science and Technology, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; College of Marine Life, Ocean University of China, Qingdao 266100, China
| | - Songlin Chen
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Pilot National Laboratory for Marine Science and Technology, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; Key Laboratory for Sustainable Development of Marine Fisheries, Ministry of Agriculture and Rural Affairs, Qingdao 266071, China; Shandong Key Laboratory of Marine Fisheries Biotechnology and Genetic Breeding, Qingdao 266071, China.
| | - Na Wang
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Pilot National Laboratory for Marine Science and Technology, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; Key Laboratory for Sustainable Development of Marine Fisheries, Ministry of Agriculture and Rural Affairs, Qingdao 266071, China; Shandong Key Laboratory of Marine Fisheries Biotechnology and Genetic Breeding, Qingdao 266071, China.
| |
Collapse
|
42
|
Cohn EF, Clayton BL, Madhavan M, Yacoub S, Federov Y, Paul-Friedman K, Shafer TJ, Tesar PJ. Pervasive environmental chemicals impair oligodendrocyte development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.10.528042. [PMID: 36798415 PMCID: PMC9934656 DOI: 10.1101/2023.02.10.528042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
Exposure to environmental chemicals can impair neurodevelopment1-4. Oligodendrocytes that wrap around axons to boost neurotransmission may be particularly vulnerable to chemical toxicity as they develop throughout fetal development and into adulthood5,6. However, few environmental chemicals have been assessed for potential risks to oligodendrocyte development. Here, we utilized a high-throughput developmental screen and human cortical brain organoids, which revealed environmental chemicals in two classes that disrupt oligodendrocyte development through distinct mechanisms. Quaternary compounds, ubiquitous in disinfecting agents, hair conditioners, and fabric softeners, were potently and selectively cytotoxic to developing oligodendrocytes through activation of the integrated stress response. Organophosphate flame retardants, commonly found in household items such as furniture and electronics, were non-cytotoxic but prematurely arrested oligodendrocyte maturation. Chemicals from each class impaired human oligodendrocyte development in a 3D organoid model of prenatal cortical development. In analysis of epidemiological data from the CDC's National Health and Nutrition Examination Survey, adverse neurodevelopmental outcomes were associated with childhood exposure to the top organophosphate flame retardant identified by our oligodendrocyte toxicity platform. Collectively, our work identifies toxicological vulnerabilities specific to oligodendrocyte development and highlights common household chemicals with high exposure risk to children that warrant deeper scrutiny for their impact on human health.
Collapse
Affiliation(s)
- Erin F. Cohn
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Benjamin L.L. Clayton
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Mayur Madhavan
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Sara Yacoub
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Yuriy Federov
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Katie Paul-Friedman
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Timothy J. Shafer
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Paul J. Tesar
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| |
Collapse
|
43
|
Chen K, Cambi F, Kozai TDY. Pro-myelinating Clemastine administration improves recording performance of chronically implanted microelectrodes and nearby neuronal health. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.31.526463. [PMID: 36778360 PMCID: PMC9915570 DOI: 10.1101/2023.01.31.526463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Intracortical microelectrodes have become a useful tool in neuroprosthetic applications in the clinic and to understand neurological disorders in basic neurosciences. Many of these brain-machine interface technology applications require successful long-term implantation with high stability and sensitivity. However, the intrinsic tissue reaction caused by implantation remains a major failure mechanism causing loss of recorded signal quality over time. Oligodendrocytes remain an underappreciated intervention target to improve chronic recording performance. These cells can accelerate action potential propagation and provides direct metabolic support for neuronal health and functionality. However, implantation injury causes oligodendrocyte degeneration and leads to progressive demyelination in surrounding brain tissue. Previous work highlighted that healthy oligodendrocytes are necessary for greater electrophysiological recording performance and the prevention of neuronal silencing around implanted microelectrodes over chronic implantation. Thus, we hypothesize that enhancing oligodendrocyte activity with a pharmaceutical drug, Clemastine, will prevent the chronic decline of microelectrode recording performance. Electrophysiological evaluation showed that the promyelination Clemastine treatment significantly elevated the signal detectability and quality, rescued the loss of multi-unit activity, and increased functional interlaminar connectivity over 16-weeks of implantation. Additionally, post-mortem immunohistochemistry showed that increased oligodendrocyte density and myelination coincided with increased survival of both excitatory and inhibitory neurons near the implant. Overall, we showed a positive relationship between enhanced oligodendrocyte activity and neuronal health and functionality near the chronically implanted microelectrode. This study shows that therapeutic strategy that enhance oligodendrocyte activity is effective for integrating the functional device interface with brain tissue over chronic implantation period. Abstract Figure
Collapse
|
44
|
Lipidomics analysis in drug discovery and development. Curr Opin Chem Biol 2023; 72:102256. [PMID: 36586190 DOI: 10.1016/j.cbpa.2022.102256] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 11/08/2022] [Accepted: 11/28/2022] [Indexed: 12/30/2022]
Abstract
Despite being a relatively new addition to the Omics' landscape, lipidomics is increasingly being recognized as an important tool for the identification of druggable targets and biochemical markers. In this review we present recent advances of lipid analysis in drug discovery and development. We cover current state of the art technologies which are constantly evolving to meet demands in terms of sensitivity and selectivity. A careful selection of important examples is then provided, illustrating the versatility of lipidomics analysis in the drug discovery and development process. Integration of lipidomics with other omics', stem-cell technologies, and metabolic flux analysis will open new avenues for deciphering pathophysiological mechanisms and the discovery of novel targets and biomarkers.
Collapse
|
45
|
Harrington AW, Liu C, Phillips N, Nepomuceno D, Kuei C, Chang J, Chen W, Sutton SW, O'Malley D, Pham L, Yao X, Sun S, Bonaventure P. Identification and characterization of select oxysterols as ligands for GPR17. Br J Pharmacol 2023; 180:401-421. [PMID: 36214386 DOI: 10.1111/bph.15969] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 09/28/2022] [Accepted: 09/30/2022] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND AND PURPOSE G-protein coupled receptor 17 (GPR17) is an orphan receptor involved in the process of myelination, due to its ability to inhibit the maturation of oligodendrocyte progenitor cells (OPCs) into myelinating oligodendrocytes. Despite multiple claims that the biological ligand has been identified, it remains an orphan receptor. EXPERIMENTAL APPROACH Seventy-seven oxysterols were screened in a cell-free [35 S]GTPγS binding assay using membranes from cells expressing GPR17. The positive hits were characterized using adenosine 3',5' cyclic monophosphate (cAMP), inositol monophosphate (IP1) and calcium mobilization assays, with results confirmed in rat primary oligodendrocytes. Rat and pig brain extracts were separated by high-performance liquid chromatography (HPLC) and endogenous activator(s) were identified in receptor activation assays. Gene expression studies of GPR17, and CYP46A1 (cytochrome P450 family 46 subfamily A member 1) enzymes responsible for the conversion of cholesterol into specific oxysterols, were performed using quantitative real-time PCR. KEY RESULTS Five oxysterols were able to stimulate GPR17 activity, including the brain cholesterol, 24(S)-hydroxycholesterol (24S-HC). A specific brain fraction from rat and pig extracts containing 24S-HC activates GPR17 in vitro. Expression of Gpr17 during mouse brain development correlates with the expression of Cyp46a1 and the levels of 24S-HC itself. Other active oxysterols have low brain concentrations below effective ranges. CONCLUSIONS AND IMPLICATIONS Oxysterols, including but not limited to 24S-HC, could be physiological activators for GPR17 and thus potentially regulate OPC differentiation and myelination through activation of the receptor.
Collapse
Affiliation(s)
| | - Changlu Liu
- Janssen Research & Development, LLC, San Diego, California, USA
| | - Naomi Phillips
- Janssen Research & Development, LLC, San Diego, California, USA
| | | | - Chester Kuei
- Janssen Research & Development, LLC, San Diego, California, USA
| | - Joseph Chang
- Janssen Research & Development, LLC, San Diego, California, USA
| | - Weixuan Chen
- Janssen Research & Development, LLC, San Diego, California, USA
| | - Steven W Sutton
- Janssen Research & Development, LLC, San Diego, California, USA
| | - Daniel O'Malley
- Janssen Research & Development, LLC, San Diego, California, USA
| | - Ly Pham
- Janssen Research & Development, LLC, San Diego, California, USA
| | - Xiang Yao
- Janssen Research & Development, LLC, San Diego, California, USA
| | - Siquan Sun
- Janssen Research & Development, LLC, San Diego, California, USA
| | | |
Collapse
|
46
|
Wang Y, Yang X, Cao Y, Li X, Xu R, Yan J, Guo Z, Sun S, Sun X, Wu Y. Electroacupuncture promotes remyelination and alleviates cognitive deficit via promoting OPC differentiation in a rat model of subarachnoid hemorrhage. Metab Brain Dis 2023; 38:687-698. [PMID: 36383326 DOI: 10.1007/s11011-022-01102-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 10/10/2022] [Indexed: 11/17/2022]
Abstract
Subarachnoid hemorrhage (SAH) is a devastating cerebral vascular disease which causes neurological deficits including long-term cognitive deficit. Demyelination of white matter is correlated with cognitive deficit in SAH. Electroacupuncture (EA) is a traditional Chinese medical treatment which protects against cognitive deficit in varies of neurological diseases. However, whether EA exerts protective effect on cognitive function in SAH has not been investigated. The underlying mechanism of remyelination regulated by EA remains unclear. This study aimed to investigate the protective effects of EA on cognitive deficit in a rat model of SAH. SAH was induced in SD rats (n = 72) by endovascular perforation. Rats in EA group received EA treatment (10 min per day) under isoflurane anesthesia after SAH. Rats in SAH and sham groups received the same isoflurane anesthesia with no treatment. The mortality rate, neurological score, cognitive function, cerebral blood flow (CBF), and remyelination in sham, SAH and EA groups were assessed at 21 d after SAH.EA treatment alleviated cognitive deficits and myelin injury of rats compared with that in SAH group. Moreover, EA treatment enhanced remyelination in white matter and promoted the differentiation of OPCs after SAH. EA treatment inhibited the expression of Id2 and promoted the expression of SOX10 in oligodendrocyte cells. Additionally, the cerebral blood flow (CBF) of rats was increased by EA compared with that in SAH group. EA treatment exerts protective effect against cognitive deficit in the late phase of SAH. The underlying mechanisms involve promoting oligodendrocyte progenitor cell (OPC) differentiation and remyelination in white matter via regulating the expression of Id2 and SOX10. The improvement of CBF may also account for the protective effect of EA on cognitive function. EA treatment is a potential therapy for the treatment of cognitive deficit after SAH.
Collapse
Affiliation(s)
- Yingwen Wang
- Departement of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, NO.1 of Youyi Road, Yuzhong District, Chongqing, China
| | - Xiaomin Yang
- Departement of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, NO.1 of Youyi Road, Yuzhong District, Chongqing, China
| | - Yunchuan Cao
- Departement of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, NO.1 of Youyi Road, Yuzhong District, Chongqing, China
| | - Xiaoguo Li
- Departement of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, NO.1 of Youyi Road, Yuzhong District, Chongqing, China
| | - Rui Xu
- Departement of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, NO.1 of Youyi Road, Yuzhong District, Chongqing, China
| | - Jin Yan
- Departement of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, NO.1 of Youyi Road, Yuzhong District, Chongqing, China
| | - Zongduo Guo
- Departement of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, NO.1 of Youyi Road, Yuzhong District, Chongqing, China
| | - Shanquan Sun
- Institute of Neuroscience, Chongqing Medical University, Chongqing, China
| | - Xiaochuan Sun
- Departement of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, NO.1 of Youyi Road, Yuzhong District, Chongqing, China.
| | - Yue Wu
- Departement of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, NO.1 of Youyi Road, Yuzhong District, Chongqing, China.
| |
Collapse
|
47
|
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: 2.0] [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
|
48
|
Korade Z, Tallman KA, Kim HYH, Balog M, Genaro-Mattos TC, Pattnaik A, Mirnics K, Pattnaik AK, Porter NA. Dose-Response Effects of 7-Dehydrocholesterol Reductase Inhibitors on Sterol Profiles and Vesicular Stomatitis Virus Replication. ACS Pharmacol Transl Sci 2022; 5:1086-1096. [PMID: 36407960 PMCID: PMC9667548 DOI: 10.1021/acsptsci.2c00051] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Indexed: 11/29/2022]
Abstract
Cholesterol is ubiquitous in cells; it plays a critical role in membrane structure and transport as well as in intracellular trafficking processes. There are suggestions that cholesterol metabolism is linked to innate immunity with inhibitors of DHCR7, the last enzyme in the cholesterol pathway, suggested to have potential as viral therapeutics nearly a decade ago. In fact, there are a number of highly prescribed pharmaceuticals that are off-target inhibitors of DHCR7, causing increased cellular levels of 7-dehydrodesmosterol (7-DHD) and 7-dehydrocholesterol (7-DHC). We report here dose-response studies of six such inhibitors on late-stage cholesterol biosynthesis in Neuro2a cells as well as their effect on infection of vesicular stomatitis virus (VSV). Four of the test compounds are FDA-approved drugs (cariprazine, trazodone, metoprolol, and tamoxifen), one (ifenprodil) has been the object of a recent Phase 2b COVID trial, and one (AY9944) is an experimental compound that has seen extensive use as a DHCR7 inhibitor. The three FDA-approved drugs inhibit replication of a GFP-tagged VSV with efficacies that mirror their effect on DHCR7. Ifenprodil and AY9944 have complex inhibitory profiles, acting on both DHCR7 and DHCR14, while tamoxifen does not inhibit DHCR7 and is toxic to Neuro2a at concentrations where it inhibits the Δ7-Δ8 isomerase of the cholesterol pathway. VSV itself affects the sterol profile in Neuro2a cells, showing a dose-response increase of dehydrolathosterol and lathosterol, the substrates for DHCR7, with a corresponding decrease in desmosterol and cholesterol. 7-DHD and 7-DHC are orders of magnitude more vulnerable to free radical chain oxidation than other sterols as well as polyunsaturated fatty esters, and the effect of these sterols on viral infection is likely a reflection of this fact of Nature.
Collapse
Affiliation(s)
- Zeljka Korade
- Department
of Pediatrics, Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Keri A. Tallman
- Department
of Chemistry, Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Hye-Young H. Kim
- Department
of Chemistry, Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Marta Balog
- Munroe-Meyer
Institute for Genetics and Rehabilitation, University of Nebraska Medical Center, Omaha, Nebraska 68105, United States
- Department
of Medical Biology and Genetics, Faculty of Medicine, J. J. Strossmayer University of Osijek, Osijek 31000, Croatia
| | - Thiago C. Genaro-Mattos
- Munroe-Meyer
Institute for Genetics and Rehabilitation, University of Nebraska Medical Center, Omaha, Nebraska 68105, United States
| | - Aryamav Pattnaik
- Nebraska
Center for Virology and School of Veterinary Medicine and Biomedical
Sciences, University of Nebraska-Lincoln, Lincoln 68583, United States
| | - Károly Mirnics
- Munroe-Meyer
Institute for Genetics and Rehabilitation, University of Nebraska Medical Center, Omaha, Nebraska 68105, United States
| | - Asit K. Pattnaik
- Nebraska
Center for Virology and School of Veterinary Medicine and Biomedical
Sciences, University of Nebraska-Lincoln, Lincoln 68583, United States
| | - Ned A. Porter
- Department
of Chemistry, Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37235, United States
| |
Collapse
|
49
|
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: 7] [Impact Index Per Article: 3.5] [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
|
50
|
Beyer BA, Lairson LL. Promoting remyelination: A case study in regenerative medicine. Curr Opin Chem Biol 2022; 70:102201. [PMID: 36037558 DOI: 10.1016/j.cbpa.2022.102201] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 07/08/2022] [Accepted: 07/14/2022] [Indexed: 11/03/2022]
Abstract
Therapeutics that modulate regenerative mechanisms by targeting the activity of endogenous (adult) stem cell populations have the potential to revolutionize medicine. In many human disease states, capacity to repair damaged tissue underlies progressive decline and disease progression. Recent insights derived from efforts aimed at promoting remyelination for the treatment of multiple sclerosis (MS) highlight the importance of considering the limiting factors and underlying mechanisms associated with all aspects of disease onset, progression and recovery, during both the discovery and clinical stages of developing a regenerative medicine. This perspective presents general considerations for the development of regenerative therapies, using remyelination as a case study.
Collapse
Affiliation(s)
- Brittney A Beyer
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Luke L Lairson
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, 92037, USA.
| |
Collapse
|