1
|
Incontro S, Musella ML, Sammari M, Di Scala C, Fantini J, Debanne D. Lipids shape brain function through ion channel and receptor modulations: physiological mechanisms and clinical perspectives. Physiol Rev 2025; 105:137-207. [PMID: 38990068 DOI: 10.1152/physrev.00004.2024] [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: 01/16/2024] [Revised: 05/28/2024] [Accepted: 07/01/2024] [Indexed: 07/12/2024] Open
Abstract
Lipids represent the most abundant molecular type in the brain, with a fat content of ∼60% of the dry brain weight in humans. Despite this fact, little attention has been paid to circumscribe the dynamic role of lipids in brain function and disease. Membrane lipids such as cholesterol, phosphoinositide, sphingolipids, arachidonic acid, and endocannabinoids finely regulate both synaptic receptors and ion channels that ensure critical neural functions. After a brief introduction on brain lipids and their respective properties, we review here their role in regulating synaptic function and ion channel activity, action potential propagation, neuronal development, and functional plasticity and their contribution in the development of neurological and neuropsychiatric diseases. We also provide possible directions for future research on lipid function in brain plasticity and diseases.
Collapse
Affiliation(s)
| | | | - Malika Sammari
- UNIS, INSERM, Aix-Marseille Université, Marseille, France
| | | | | | | |
Collapse
|
2
|
Hasegawa S, Watanabe S, Fujimoto S, Kondo S, Nishi T. Characterization of soticlestat, a novel cholesterol 24-hydroxylase inhibitor, in acute and chronic neurodegeneration models. Neurosci Res 2024; 208:29-38. [PMID: 38897234 DOI: 10.1016/j.neures.2024.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 06/05/2024] [Accepted: 06/14/2024] [Indexed: 06/21/2024]
Abstract
We investigated whether soticlestat (TAK-935), a newly discovered cholesterol 24-hydroxylase (CH24H) inhibitor now in phase 3 clinical trials for Dravet and Lennox-Gastaut syndromes, has effects on neurodegeneration in both chronic and acute animal models associated with glutamate hyperexcitation. Soticlestat was administered at doses that approximately halve 24S-hydroxycholesterol in both experiments. In the kainic acid (KA)-induced acute hippocampal degeneration model, soticlestat ameliorated inflammatory cytokine expression, hippocampal degeneration, and memory impairment. We ruled out the possibility that soticlestat directly interferes with KA binding to the KA receptor, or that 24S-hydroxycholesterol modulates KA receptor signaling, by conducting receptor binding and cell death assays. In the PS19 chronic degeneration model of tauopathy, treatment effects were observed in neurodegeneration markers. Notably, there was a significant correlation between the levels of brain 24S-hydroxycholesterol and a proinflammatory cytokine, tumor necrosis factor-α, which is implicated in cognitive decline and lowering of seizure threshold. This is the first study demonstrating that CH24H inhibition can alleviate neurodegeneration concomitant with neuroinflammation. Herein, we discuss the interplay among 24S-hydroxycholesterol production, neuroinflammation, and excitotoxicity. Effects on neurodegeneration and neuroinflammation demonstrated in two preclinical models suggest that soticlestat is effective in ameliorating seizures and addressing cognitive dysfunction in seizure disorders.
Collapse
Affiliation(s)
- Shigeo Hasegawa
- Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 2-26-1 Muraoka Higashi, Fujisawa 251-8555, Japan.
| | - Sayuri Watanabe
- Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 2-26-1 Muraoka Higashi, Fujisawa 251-8555, Japan.
| | - Shinji Fujimoto
- Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 2-26-1 Muraoka Higashi, Fujisawa 251-8555, Japan.
| | - Shinichi Kondo
- Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 2-26-1 Muraoka Higashi, Fujisawa 251-8555, Japan.
| | - Toshiya Nishi
- Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 2-26-1 Muraoka Higashi, Fujisawa 251-8555, Japan.
| |
Collapse
|
3
|
Cho K, Kim GW. Decreased SREBP2 of the striatal cell relates to disrupted protein degradation in Huntington's disease. Brain Res 2024; 1846:149250. [PMID: 39313167 DOI: 10.1016/j.brainres.2024.149250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 09/14/2024] [Accepted: 09/20/2024] [Indexed: 09/25/2024]
Abstract
This study delineated the intricate relation between cholesterol metabolism, protein degradation mechanisms, and the pathogenesis of Huntington's disease (HD). Through investigations using both animal models and cellular systems, we have observed significant alterations in cholesterol levels, particularly in the striatum, which is the primary lesion site in HD. Our findings indicate the dysregulation of cholesterol metabolism-related factors, such as LDLR and SREBP2, in HD, which may contribute to disease progression. Additionally, we uncovered disruptions in protein degradation pathways, including decreased neddylated proteins and dysregulated autophagy, which further exacerbated HD pathology. Moreover, our study highlighted the potential therapeutic implications of targeting these pathways. By restoring cholesterol levels and modulating protein degradation mechanisms, particularly through interventions, such as MLN4924, we observed potential improvements in cellular function, as indicated by the increased BDNF levels. These insights underscore the importance of simultaneously addressing cholesterol metabolism and protein degradation to alleviate HD pathology. Collectively, this study provides a basic understanding of the interplay between the decrease of SREBP2 and the dysfunctional protein degradation system derived from disrupted cholesterol metabolism in HD and HD cells.
Collapse
Affiliation(s)
- Kyoungjoo Cho
- Department of Life Science, Kyonggi University, Suwon, South Korea
| | - Gyung Whan Kim
- Department of Neurology, College of Medicine, Yonsei University, Seoul, South Korea.
| |
Collapse
|
4
|
Ye D, Chukwu C, Yang Y, Hu Z, Chen H. Adeno-associated virus vector delivery to the brain: Technology advancements and clinical applications. Adv Drug Deliv Rev 2024; 211:115363. [PMID: 38906479 DOI: 10.1016/j.addr.2024.115363] [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: 12/20/2023] [Revised: 05/13/2024] [Accepted: 06/18/2024] [Indexed: 06/23/2024]
Abstract
Adeno-associated virus (AAV) vectors have emerged as a promising tool in the development of gene therapies for various neurological diseases, including Alzheimer's disease and Parkinson's disease. However, the blood-brain barrier (BBB) poses a significant challenge to successfully delivering AAV vectors to the brain. Strategies that can overcome the BBB to improve the AAV delivery efficiency to the brain are essential to successful brain-targeted gene therapy. This review provides an overview of existing strategies employed for AAV delivery to the brain, including direct intraparenchymal injection, intra-cerebral spinal fluid injection, intranasal delivery, and intravenous injection of BBB-permeable AAVs. Focused ultrasound has emerged as a promising technology for the noninvasive and spatially targeted delivery of AAV administered by intravenous injection. This review also summarizes each strategy's current preclinical and clinical applications in treating neurological diseases. Moreover, this review includes a detailed discussion of the recent advances in the emerging focused ultrasound-mediated AAV delivery. Understanding the state-of-the-art of these gene delivery approaches is critical for future technology development to fulfill the great promise of AAV in neurological disease treatment.
Collapse
Affiliation(s)
- Dezhuang Ye
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - Chinwendu Chukwu
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - Yaoheng Yang
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - Zhongtao Hu
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - Hong Chen
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO 63130, USA; Department of Neurosurgery, Washington University School of Medicine, Saint Louis, MO 63110 USA; Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| |
Collapse
|
5
|
Feringa FM, Hertog SJKD, Wang L, Derks RJE, Kruijff I, Erlebach L, Heijneman J, Miramontes R, Pömpner N, Blomberg N, Olivier-Jimenez D, Johansen LE, Cammack AJ, Giblin A, Toomey CE, Rose IVL, Yuan H, Ward M, Isaacs AM, Kampmann M, Kronenberg-Versteeg D, Lashley T, Thompson LM, Ori A, Mohammed Y, Giera M, van der Kant R. The Neurolipid Atlas: a lipidomics resource for neurodegenerative diseases uncovers cholesterol as a regulator of astrocyte reactivity impaired by ApoE4. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.01.601474. [PMID: 39005258 PMCID: PMC11244892 DOI: 10.1101/2024.07.01.601474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Lipid changes in the brain have been implicated in many neurodegenerative diseases including Alzheimer's Disease (AD), Parkinson's disease and Amyotrophic Lateral Sclerosis. To facilitate comparative lipidomic research across brain-diseases we established a data commons named the Neurolipid Atlas, that we have pre-populated with novel human, mouse and isogenic induced pluripotent stem cell (iPSC)-derived lipidomics data for different brain diseases. We show that iPSC-derived neurons, microglia and astrocytes display distinct lipid profiles that recapitulate in vivo lipotypes. Leveraging multiple datasets, we show that the AD risk gene ApoE4 drives cholesterol ester (CE) accumulation in human astrocytes recapitulating CE accumulation measured in the human AD brain. Multi-omic interrogation of iPSC-derived astrocytes revealed that cholesterol plays a major role in astrocyte interferon-dependent pathways such as the immunoproteasome and major histocompatibility complex (MHC) class I antigen presentation. We show that through enhanced cholesterol esterification ApoE4 suppresses immune activation of astrocytes. Our novel data commons, available at neurolipidatlas.com, provides a user-friendly tool and knowledge base for a better understanding of lipid dyshomeostasis in neurodegenerative diseases.
Collapse
Affiliation(s)
- Femke M Feringa
- Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Sascha J Koppes-den Hertog
- Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, the Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Lian Wang
- Leiden University Medical Center, Center for Proteomics and Metabolomics, Leiden, the Netherlands
| | - Rico J E Derks
- Leiden University Medical Center, Center for Proteomics and Metabolomics, Leiden, the Netherlands
| | - Iris Kruijff
- Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, the Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Lena Erlebach
- German Center for Neurodegenerative Diseases (DZNE) Tübingen, Tübingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Jorin Heijneman
- Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, the Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Ricardo Miramontes
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Nadine Pömpner
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany
| | - Niek Blomberg
- Leiden University Medical Center, Center for Proteomics and Metabolomics, Leiden, the Netherlands
| | - Damien Olivier-Jimenez
- Leiden University Medical Center, Center for Proteomics and Metabolomics, Leiden, the Netherlands
| | - Lill Eva Johansen
- Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Alexander J Cammack
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute at UCL, University College London, London, UK
| | - Ashling Giblin
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute at UCL, University College London, London, UK
| | - Christina E Toomey
- Department of Clinical and Molecular Neuroscience, Queen Square Institute of Neurology, University College London, London, UK
| | - Indigo V L Rose
- Institute for Neurodegenerative Diseases and Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Hebao Yuan
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Michael Ward
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Adrian M Isaacs
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute at UCL, University College London, London, UK
| | - Martin Kampmann
- Department of Biochemistry and Biophysics, Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
| | - Deborah Kronenberg-Versteeg
- German Center for Neurodegenerative Diseases (DZNE) Tübingen, Tübingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Tammaryn Lashley
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Leslie M Thompson
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Alessandro Ori
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany
| | - Yassene Mohammed
- Leiden University Medical Center, Center for Proteomics and Metabolomics, Leiden, the Netherlands
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC H3A 0G4, Canada
| | - Martin Giera
- Leiden University Medical Center, Center for Proteomics and Metabolomics, Leiden, the Netherlands
| | - Rik van der Kant
- Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, the Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, the Netherlands
| |
Collapse
|
6
|
Patel RV, Nanda P, Richardson RM. Neurosurgical gene therapy for central nervous system diseases. Neurotherapeutics 2024; 21:e00434. [PMID: 39191071 PMCID: PMC11445594 DOI: 10.1016/j.neurot.2024.e00434] [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: 05/11/2024] [Revised: 07/16/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024] Open
Abstract
Viral vector mediated gene therapies for neurodegenerative and neurodevelopmental conditions that require neurosurgical administration continue to expand. We systematically reviewed the National Institutes of Health (NIH) ClinicalTrials.gov database to identify all clinical trials studying in-vivo viral vector mediated gene therapies targeted to the CNS for neurodegenerative and neurodevelopmental diseases. We isolated studies which delivered therapies using neurosurgical approaches: intracisternal, intraventricular, and/or intraparenchymal. Clinical trials primarily registered in international countries were included if they were referenced by an NIH registered clinical trial. We performed a scoping review to identify the preclinical studies that supported each human clinical trial. Key preclinical and clinical data were aggregated to characterize vector capsid design, delivery methods, gene expression profile, and clinical benefit. A total of 64 clinical trials were identified in active, completed, terminated, and long-term follow-up stages. A range of CNS conditions across pediatric and adult populations are being studied with CNS targeted viral vector gene therapy, including Alzheimer's disease, Parkinson's disease, AADC deficiency, sphingolipidoses, mucopolysaccharidoses, neuronal ceroid lipofuscinoses, spinal muscular atrophy, adrenoleukodystrophy, Canavan disease, frontotemporal dementia, Huntington's disease, Rett syndrome, Dravet syndrome, mesial temporal lobe epilepsy, and glutaric acidemia. Adeno-associated viral vectors (AAVs) were utilized by the majority of tested therapies, with vector serotypes, regulatory elements, delivery methods, and vector monitoring varying based on the disease being studied. Intraparenchymal delivery has evolved significantly, with MRI-guided convection-enhanced delivery established as a gold standard method for pioneering novel gene targets.
Collapse
Affiliation(s)
- Ruchit V Patel
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Pranav Nanda
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - R Mark Richardson
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
7
|
Fanaei H, Shoorijeh BT, Hafezinouri H, Mirzaei I, Parsi-Moud A. Impact of social isolation on corticosterone release and recovery after stroke in aged rats: A behavioral and biochemical analysis. Exp Gerontol 2024; 192:112453. [PMID: 38723916 DOI: 10.1016/j.exger.2024.112453] [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: 02/23/2024] [Revised: 04/29/2024] [Accepted: 05/06/2024] [Indexed: 06/01/2024]
Abstract
Social isolation (SI) after stroke reduces recovery. The aim of this study was to evaluate the effects of SI on corticosterone release and recovery after stroke in aged rats. A total of 64 male Wistar rats (aged 24 months) were used in the present study. All rats were housed in pairs for two weeks. After two weeks, rats were randomly assigned to one of four groups: (1) rats underwent sham surgery and kept socially isolated (control/social isolated (CO/SI) group); (2) rats underwent sham surgery and kept pair housed (control/pair housed (CO/PH) group); (3) rats underwent middle cerebral artery occlusion (MCAO) surgery and kept socially isolated (stroke/isolated (ST/SI) group); (4) rats underwent MCAO surgery and kept pair housed (stroke/pair housed (ST/PH)) group. Behaviors were assessed using the adhesive removal test, rotarod test and social interaction test at 1st, 7th, 14th and 21st days after stroke. Serum biochemical analysis was also performed on the behavioral testing days. Results showed THAT serum corticosterone and MDA levels in CO/PH group were significantly lower than CO/SI group. Serum BDNF levels in CO/PH group was significantly higher than CO/SI group. Serum corticosterone and MDA levels in ST/PH group were lower than ST/SI group. In ST/PH group, serum Total antioxidant capacity (TAC) and BDNF levels were significantly higher than ST/SI group. Biochemical analysis of certain regions of the brain (hippocampus, striatum and cerebral cortex) was performed on 21st day after stroke. In the hippocampus of CO/PH group, BDNF and TAC levels were significantly higher than CO/SI group. The hippocampal MDA level of CO/PH group were significantly lower than CO/SI group. BDNF and TAC levels in the hippocampus, striatum and cerebral cortex of ST/PH group were significantly higher and MDA level was significantly lower as compared with ST/SI group. Both ischemic groups showed sensorimotor recovery over a 21-day period, but recovery of ST/PH group was significantly greater than ST/SI group. Total social interaction time in ST/PH group was significantly longer than ST/SI group. Based on the results of this study, social interaction after stroke enhances histologic and sensorimotor recovery through reduction of HPA activity and corticosterone release, leading to increased TAC and BDNF levels.
Collapse
Affiliation(s)
- Hamed Fanaei
- Pregnancy Health Research Center, Department of Physiology, Zahedan University of Medical Sciences, Zahedan, Iran.
| | | | - Hamid Hafezinouri
- Laboratory Animal Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Ilia Mirzaei
- Student Research Committee, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Abolfazl Parsi-Moud
- Student Research Committee, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| |
Collapse
|
8
|
Katznelson A, Hernandez B, Fahning H, Zhang J, Burton A, Torres-Padilla ME, Plachta N, Zaret KS, McCarthy RL. Heterochromatin protein ERH represses alternative cell fates during early mammalian differentiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.06.597604. [PMID: 38895478 PMCID: PMC11185749 DOI: 10.1101/2024.06.06.597604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
During development, H3K9me3 heterochromatin is dynamically rearranged, silencing repeat elements and protein coding genes to restrict cell identity. Enhancer of Rudimentary Homolog (ERH) is an evolutionarily conserved protein originally characterized in fission yeast and recently shown to be required for H3K9me3 maintenance in human fibroblasts, but its function during development remains unknown. Here, we show that ERH is required for proper segregation of the inner cell mass and trophectoderm cell lineages during mouse development by repressing totipotent and alternative lineage programs. During human embryonic stem cell (hESC) differentiation into germ layer lineages, ERH is crucial for silencing naïve and pluripotency genes, transposable elements, and alternative lineage genes. Strikingly, ERH depletion in somatic cells reverts the H3K9me3 landscape to an hESC state and enables naïve and pluripotency gene and transposable element activation during iPSC reprogramming. Our findings reveal a role for ERH in initiation and maintenance of developmentally established gene repression.
Collapse
|
9
|
Audouard E, Khefif N, Gillet-Legrand B, Nobilleau F, Bouazizi O, Stanga S, Despres G, Alves S, Lamazière A, Cartier N, Piguet F. Modulation of Brain Cholesterol Metabolism through CYP46A1 Overexpression for Rett Syndrome. Pharmaceutics 2024; 16:756. [PMID: 38931878 PMCID: PMC11207948 DOI: 10.3390/pharmaceutics16060756] [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: 05/02/2024] [Revised: 05/27/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
Rett syndrome (RTT) is a rare neurodevelopmental disorder caused by mutation in the X-linked gene methyl-CpG-binding protein 2 (Mecp2), a ubiquitously expressed transcriptional regulator. RTT results in mental retardation and developmental regression that affects approximately 1 in 10,000 females. Currently, there is no curative treatment for RTT. Thus, it is crucial to develop new therapeutic approaches for children suffering from RTT. Several studies suggested that RTT is linked with defects in cholesterol homeostasis, but for the first time, therapeutic evaluation is carried out by modulating this pathway. Moreover, AAV-based CYP46A1 overexpression, the enzyme involved in cholesterol pathway, has been demonstrated to be efficient in several neurodegenerative diseases. Based on these data, we strongly believe that CYP46A1 could be a relevant therapeutic target for RTT. Herein, we evaluated the effects of intravenous AAVPHP.eB-hCYP46A1-HA delivery in male and female Mecp2-deficient mice. The applied AAVPHP.eB-hCYP46A1 transduced essential neurons of the central nervous system (CNS). CYP46A1 overexpression alleviates behavioral alterations in both male and female Mecp2 knockout mice and extends the lifespan in Mecp2-deficient males. Several parameters related to cholesterol pathway are improved and correction of mitochondrial activity is demonstrated in treated mice, which highlighted the clear therapeutic benefit of CYP46A1 through the neuroprotection effect. IV delivery of AAVPHP.eB-CYP46A1 is perfectly well tolerated with no inflammation observed in the CNS of the treated mice. Altogether, our results strongly suggest that CYP46A1 is a relevant target and overexpression could alleviate the phenotype of Rett patients.
Collapse
Affiliation(s)
- Emilie Audouard
- TIDU GENOV, Institut du Cerveau, ICM, F-75013 Paris, France;
- Institut du Cerveau, ICM, Inserm U 1127, CNRS UMR 7225, Sorbonne Université, F-75013 Paris, France (B.G.-L.); (S.A.)
| | - Nicolas Khefif
- Institut du Cerveau, ICM, Inserm U 1127, CNRS UMR 7225, Sorbonne Université, F-75013 Paris, France (B.G.-L.); (S.A.)
| | - Béatrix Gillet-Legrand
- Institut du Cerveau, ICM, Inserm U 1127, CNRS UMR 7225, Sorbonne Université, F-75013 Paris, France (B.G.-L.); (S.A.)
| | - Fanny Nobilleau
- Institut du Cerveau, ICM, Inserm U 1127, CNRS UMR 7225, Sorbonne Université, F-75013 Paris, France (B.G.-L.); (S.A.)
| | - Ouafa Bouazizi
- Institut du Cerveau, ICM, Inserm U 1127, CNRS UMR 7225, Sorbonne Université, F-75013 Paris, France (B.G.-L.); (S.A.)
| | - Serena Stanga
- Neuroscience Institute Cavalieri Ottolenghi, 10043 Orbassano, Italy
- Department of Neuroscience Rita Levi Montalcini, University of Turin, 10126 Turin, Italy
| | - Gaëtan Despres
- Saint Antoine Research Center, INSERM UMR 938, Département de Métabolomique Clinique, Hôpital Saint Antoine, AP-HP Sorbonne Université, F-75013 Paris, France
| | - Sandro Alves
- Institut du Cerveau, ICM, Inserm U 1127, CNRS UMR 7225, Sorbonne Université, F-75013 Paris, France (B.G.-L.); (S.A.)
| | - Antonin Lamazière
- Saint Antoine Research Center, INSERM UMR 938, Département de Métabolomique Clinique, Hôpital Saint Antoine, AP-HP Sorbonne Université, F-75013 Paris, France
| | - Nathalie Cartier
- Institut du Cerveau, ICM, Inserm U 1127, CNRS UMR 7225, Sorbonne Université, F-75013 Paris, France (B.G.-L.); (S.A.)
| | - Françoise Piguet
- TIDU GENOV, Institut du Cerveau, ICM, F-75013 Paris, France;
- Institut du Cerveau, ICM, Inserm U 1127, CNRS UMR 7225, Sorbonne Université, F-75013 Paris, France (B.G.-L.); (S.A.)
| |
Collapse
|
10
|
Mast N, Butts M, Pikuleva IA. Unbiased insights into the multiplicity of the CYP46A1 brain effects in 5XFAD mice treated with low dose-efavirenz. J Lipid Res 2024; 65:100555. [PMID: 38719151 PMCID: PMC11176809 DOI: 10.1016/j.jlr.2024.100555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/12/2024] [Accepted: 05/01/2024] [Indexed: 05/30/2024] Open
Abstract
Cytochrome P450 46A1 (CYP46A1) is the CNS-specific cholesterol 24-hydroxylase that controls cholesterol elimination and turnover in the brain. In mouse models, pharmacologic CYP46A1 activation with low-dose efavirenz or by gene therapy mitigates the manifestations of various brain disorders, neurologic, and nonneurologic, by affecting numerous, apparently unlinked biological processes. Accordingly, CYP46A1 is emerging as a promising therapeutic target; however, the mechanisms underlying the multiplicity of the brain CYP46A1 activity effects are currently not understood. We proposed the chain reaction hypothesis, according to which CYP46A1 is important for the three primary (unifying) processes in the brain (sterol flux through the plasma membranes, acetyl-CoA, and isoprenoid production), which in turn affect a variety of secondary processes. We already identified several processes secondary to changes in sterol flux and herein undertook a multiomics approach to compare the brain proteome, acetylproteome, and metabolome of 5XFAD mice (an Alzheimer's disease model), control and treated with low-dose efavirenz. We found that the latter had increased production of phospholipids from the corresponding lysophospholipids and a globally increased protein acetylation (including histone acetylation). Apparently, these effects were secondary to increased acetyl-CoA production. Signaling of small GTPases due to their altered abundance or abundance of their regulators could be affected as well, potentially via isoprenoid biosynthesis. In addition, the omics data related differentially abundant molecules to other biological processes either reported previously or new. Thus, we obtained unbiased mechanistic insights and identified potential players mediating the multiplicity of the CYP46A1 brain effects and further detailed our chain reaction hypothesis.
Collapse
Affiliation(s)
- Natalia Mast
- Department of Ophthalmology and Visual Science, Case Western Reserve University, Cleveland, OH, USA
| | - Makaya Butts
- Department of Ophthalmology and Visual Science, Case Western Reserve University, Cleveland, OH, USA
| | - Irina A Pikuleva
- Department of Ophthalmology and Visual Science, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
11
|
Serrano J, Martine L, Grosjean Y, Acar N, Alves G, Masson EAY. The importance of choosing the appropriate cholesterol quantification method: enzymatic assay versus gas chromatography. J Lipid Res 2024; 65:100561. [PMID: 38762123 PMCID: PMC11237936 DOI: 10.1016/j.jlr.2024.100561] [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: 02/06/2024] [Revised: 04/11/2024] [Accepted: 05/14/2024] [Indexed: 05/20/2024] Open
Abstract
Cholesterol is a major lipid of the animal realm with many biological roles. It is an important component of cellular membranes and a precursor of steroid hormones and bile acids. It is particularly abundant in nervous tissues, and dysregulation of cholesterol metabolism has been associated with neurodegenerative diseases such as Alzheimer's and Huntington's diseases. Deciphering the pathophysiological mechanisms of these disorders often involves animal models such as mice and Drosophila. Accurate quantification of cholesterol levels in the chosen models is a critical point of these studies. In the present work, we compare two common methods, gas chromatography coupled to flame-ionization detection (GC/FID) and a cholesterol oxidase-based fluorometric assay to measure cholesterol in mouse brains and Drosophila heads. Cholesterol levels measured by the two methods were similar for the mouse brain, which presents a huge majority of cholesterol in its sterol profile. On the contrary, depending on the method, measured cholesterol levels were very different for Drosophila heads, which present a complex sterol profile with a minority of cholesterol. We showed that the enzyme-based assay is not specific for cholesterol and detects other sterols as well. This method is therefore not suited for cholesterol measurement in models such as Drosophila. Alternatively, chromatographic methods, such as GC/FID, offer the required specificity for cholesterol quantification. Understanding the limitations of the quantification techniques is essential for reliable interpretation of the results in cholesterol-related research.
Collapse
Affiliation(s)
- Jeanne Serrano
- Eye & Nutrition Research Group, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne, Dijon, France; Sensory Perception & Glia-Neuron Interaction Research Group, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne, Dijon, France
| | - Lucy Martine
- Eye & Nutrition Research Group, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne, Dijon, France
| | - Yaël Grosjean
- Sensory Perception & Glia-Neuron Interaction Research Group, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne, Dijon, France
| | - Niyazi Acar
- Eye & Nutrition Research Group, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne, Dijon, France
| | - Georges Alves
- Sensory Perception & Glia-Neuron Interaction Research Group, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne, Dijon, France.
| | - Elodie A Y Masson
- Eye & Nutrition Research Group, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne, Dijon, France.
| |
Collapse
|
12
|
Lei X, Wang X, Xiong W, Xiao H, Wu Y, Huang T, Liang R, Li Y, Lin S. Cytochrome P450 Mining for Bufadienolide Diversification. ACS Chem Biol 2024; 19:1169-1179. [PMID: 38624108 DOI: 10.1021/acschembio.4c00089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
Bufadienolides are a class of steroids with a distinctive α-pyrone ring at C17, mostly produced by toads and consisting of over 100 orthologues. They exhibit potent cardiotonic and antitumor activities and are active ingredients of the traditional Chinese medicine Chansu and Cinobufacini. Direct extraction from toads is costly, and chemical synthesis is difficult, limiting the accessibility of active bufadienolides with diverse modifications and trace content. In this work, based on the transcriptome and genome analyses, using a yeast-based screening platform, we obtained eight cytochrome P450 (CYP) enzymes from toads, which catalyze the hydroxylation of bufalin and resibufogenin at different sites. Moreover, a reported fungal CYP enzyme Sth10 was found functioning in the modification of bufalin and resibufogenin at multiple sites. A total of 15 bufadienolides were produced and structurally identified, of which six were first discovered. All of the compounds were effective in inhibiting the proliferation of tumor cells, especially 19-hydroxy-bufalin (2) and 1β-hydroxy-bufalin (3), which were generated from bufalin hydroxylation catalyzed by CYP46A35. The catalytic efficiency of CYP46A35 was improved about six times and its substrate diversity was expanded to progesterone and testosterone, the common precursors for steroid drugs, achieving their efficient and site-specific hydroxylation. These findings elucidate the key modification process in the synthesis of bufadienolides by toads and provide an effective way for the synthesis of unavailable bufadienolides with site-specific modification and active potentials.
Collapse
Affiliation(s)
- Xiaolai Lei
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory on Metabolic & Developmental Sciences, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Xiaozheng Wang
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory on Metabolic & Developmental Sciences, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Weiliang Xiong
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory on Metabolic & Developmental Sciences, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Han Xiao
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory on Metabolic & Developmental Sciences, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Yingchun Wu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Tingting Huang
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory on Metabolic & Developmental Sciences, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Rubing Liang
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory on Metabolic & Developmental Sciences, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Yiming Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Shuangjun Lin
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory on Metabolic & Developmental Sciences, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
- Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| |
Collapse
|
13
|
Jia W, Li Y, Cheung KCP, Zheng X. Bile acid signaling in the regulation of whole body metabolic and immunological homeostasis. SCIENCE CHINA. LIFE SCIENCES 2024; 67:865-878. [PMID: 37515688 DOI: 10.1007/s11427-023-2353-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 04/23/2023] [Indexed: 07/31/2023]
Abstract
Bile acids (BAs) play a crucial role in nutrient absorption and act as key regulators of lipid and glucose metabolism and immune homeostasis. Through the enterohepatic circulation, BAs are synthesized, metabolized, and reabsorbed, with a portion entering the vascular circulation and distributing systemically. This allows BAs to interact with receptors in all major organs, leading to organ-organ interactions that regulate both local and global metabolic processes, as well as the immune system. This review focuses on the whole-body effects of BA-mediated metabolic and immunological regulation, including in the brain, heart, liver, intestine, eyes, skin, adipose tissue, and muscle. Targeting BA synthesis and receptor signaling is a promising strategy for the development of novel therapies for various diseases throughout the body.
Collapse
Affiliation(s)
- Wei Jia
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
| | - Yitao Li
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Kenneth C P Cheung
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Xiaojiao Zheng
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| |
Collapse
|
14
|
Kim JB, Kim SJ, So M, Kim DK, Noh HR, Kim BJ, Choi YR, Kim D, Koo H, Kim T, Woo HG, Park SM. Artificial intelligence-driven drug repositioning uncovers efavirenz as a modulator of α-synuclein propagation: Implications in Parkinson's disease. Biomed Pharmacother 2024; 174:116442. [PMID: 38513596 DOI: 10.1016/j.biopha.2024.116442] [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: 12/21/2023] [Revised: 03/09/2024] [Accepted: 03/15/2024] [Indexed: 03/23/2024] Open
Abstract
Parkinson's disease (PD) is a complex neurodegenerative disorder with an unclear etiology. Despite significant research efforts, developing disease-modifying treatments for PD remains a major unmet medical need. Notably, drug repositioning is becoming an increasingly attractive direction in drug discovery, and computational approaches offer a relatively quick and resource-saving method for identifying testable hypotheses that promote drug repositioning. We used an artificial intelligence (AI)-based drug repositioning strategy to screen an extensive compound library and identify potential therapeutic agents for PD. Our AI-driven analysis revealed that efavirenz and nevirapine, approved for treating human immunodeficiency virus infection, had distinct profiles, suggesting their potential effects on PD pathophysiology. Among these, efavirenz attenuated α-synuclein (α-syn) propagation and associated neuroinflammation in the brain of preformed α-syn fibrils-injected A53T α-syn Tg mice and α-syn propagation and associated behavioral changes in the C. elegans BiFC model. Through in-depth molecular investigations, we found that efavirenz can modulate cholesterol metabolism and mitigate α-syn propagation, a key pathological feature implicated in PD progression by regulating CYP46A1. This study opens new avenues for further investigation into the mechanisms underlying PD pathology and the exploration of additional drug candidates using advanced computational methodologies.
Collapse
Affiliation(s)
- Jae-Bong Kim
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea; Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea; Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
| | - Soo-Jeong Kim
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea
| | | | - Dong-Kyu Kim
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea
| | - Hye Rin Noh
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea; Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea; Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
| | - Beom Jin Kim
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea; Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea; Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
| | - Yu Ree Choi
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea
| | - Doyoon Kim
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea; Department of Physiology, Ajou University School of Medicine, Suwon, Korea
| | | | | | - Hyun Goo Woo
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea; Department of Physiology, Ajou University School of Medicine, Suwon, Korea
| | - Sang Myun Park
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea; Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea; Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea.
| |
Collapse
|
15
|
Chen W, Hu J, Chen J, Guo Y, Hong Y, Xia H. Spatio-temporal analysis of toxigenic genes expression in the growing Bufo gargarizans based on RNA sequencing data. Genomics 2024; 116:110847. [PMID: 38685287 DOI: 10.1016/j.ygeno.2024.110847] [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: 11/01/2023] [Revised: 03/26/2024] [Accepted: 04/19/2024] [Indexed: 05/02/2024]
Abstract
BACKGROUND Bufo gargarizans Cantor, a widely distributed amphibian species in Asia, produces and releases toxins through its retroauricular and granular glands. Although various tissues have been sequenced, the molecular mechanisms underlying the toxin production remain unclear. To elucidate these mechanisms, abdominal skin (non-toxic secretory glands) and retroauricular gland (toxic secreting glands) samples were collected at different time points (3, 6, 12, 24, and 36 months) for RNA sequencing (RNA-seq) and analysis. RESULTS In comparison to the S group during the same period, a total of 3053, 3026, 1516, 1028, and 2061 differentially expressed genes (DEGs) were identified across five developmental stages. Gene Ontology (GO) analysis revealed that DEGs were primarily enriched in biological processes including cellular processes, single-organism processes, metabolic processes, and biological regulation. In terms of cellular components, the DEGs were predominantly localized in the cell and cell parts, whereas molecular function indicated significant enrichment in binding and catalytic activity. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis showed that the metabolism and synthesis of various substances, such as lipid metabolism, cofactor and vitamin metabolism, tryptophan metabolism, steroid biosynthesis, and primary bile acid biosynthesis, were accompanied by the development of toads. Additionally, using trend analysis, we discovered candidate genes that were upregulated in the retroauricular glands during development, and the abundance of these genes in the abdominal skin was extremely low. Finally, we identified 26 genes that are likely to be involved in toxin production and that are likely to be involved in toxin anabolism. CONCLUSION Overall, these results provide new insights into the genes involved in toxin production in B. gargarizans, which will improve our understanding of the molecular mechanisms underlying toxigenic gene expression.
Collapse
Affiliation(s)
- Wenxiao Chen
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Jinghong Hu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Shandong Provincial Collaborative Innovation Center for Quality Control and Construction of the Whole Industrial Chain of Traditional Chinese Medicine, Jinan 250355, China.
| | - Jing Chen
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yuanyuan Guo
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yongjian Hong
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Houkai Xia
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Shandong Kang Yuan Tang Pharmaceutical Co., Ltd, College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| |
Collapse
|
16
|
Yu-Taeger L, El-Ayoubi A, Qi P, Danielyan L, Nguyen HHP. Intravenous MSC-Treatment Improves Impaired Brain Functions in the R6/2 Mouse Model of Huntington's Disease via Recovered Hepatic Pathological Changes. Cells 2024; 13:469. [PMID: 38534313 DOI: 10.3390/cells13060469] [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: 01/31/2024] [Revised: 03/02/2024] [Accepted: 03/06/2024] [Indexed: 03/28/2024] Open
Abstract
Huntington's disease (HD), a congenital neurodegenerative disorder, extends its pathological damages beyond the nervous system. The systematic manifestation of HD has been extensively described in numerous studies, including dysfunction in peripheral organs and peripheral inflammation. Gut dysbiosis and the gut-liver-brain axis have garnered greater emphasis in neurodegenerative research, and increased plasma levels of pro-inflammatory cytokines have been identified in HD patients and various in vivo models, correlating with disease progression. In the present study, we investigated hepatic pathological markers in the liver of R6/2 mice which convey exon 1 of the human mutant huntingtin gene. Furthermore, we evaluated the impact of intravenously administered Mesenchymal Stromal Cells (MSCs) on the liver enzymes, changes in hepatic inflammatory markers, as well as brain pathology and behavioral deficits in R6/2 mice. Our results revealed altered enzyme expression and increased levels of inflammatory mediators in the liver of R6/2 mice, which were significantly attenuated in the MSC-treated R6/2 mice. Remarkably, neuronal pathology and altered motor activities in the MSC-treated R6/2 mice were significantly ameliorated, despite the absence of MSCs in the postmortem brain. Our data highlight the importance of hepatic pathological changes in HD, providing a potential therapeutic approach. Moreover, the data open new perspectives for the search in blood biomarkers correlating with liver pathology in HD.
Collapse
Affiliation(s)
- Libo Yu-Taeger
- Department of Human Genetics, Ruhr University of Bochum, D-44801 Bochum, Germany
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, D-72076 Tuebingen, Germany
| | - Ali El-Ayoubi
- Department of Human Genetics, Ruhr University of Bochum, D-44801 Bochum, Germany
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, D-72076 Tuebingen, Germany
| | - Pengfei Qi
- Department of Human Genetics, Ruhr University of Bochum, D-44801 Bochum, Germany
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, D-72076 Tuebingen, Germany
| | - Lusine Danielyan
- Department of Clinical Pharmacology, University Hospital of Tuebingen, D-72076 Tuebingen, Germany
- Departments of Biochemistry and Clinical Pharmacology, and Neuroscience Laboratory, Yerevan State Medical University, Yerevan 0025, Armenia
| | - Hoa Huu Phuc Nguyen
- Department of Human Genetics, Ruhr University of Bochum, D-44801 Bochum, Germany
- Department of Medical Chemistry, Yerevan State Medical University, Yerevan 0025, Armenia
| |
Collapse
|
17
|
Nunes MJ, Carvalho AN, Reis J, Costa D, Moutinho M, Mateus J, Mendes de Almeida R, Brito S, Risso D, Nunes S, Castro-Caldas M, Gama MJ, Rodrigues CMP, Xapelli S, Diógenes MJ, Cartier N, Chali F, Piguet F, Rodrigues E. Cholesterol redistribution triggered by CYP46A1 gene therapy improves major hallmarks of Niemann-Pick type C disease but is not sufficient to halt neurodegeneration. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166993. [PMID: 38142760 DOI: 10.1016/j.bbadis.2023.166993] [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: 07/21/2023] [Revised: 12/13/2023] [Accepted: 12/14/2023] [Indexed: 12/26/2023]
Abstract
Cholesterol 24-hydroxylase (CYP46A1) is an exclusively neuronal cytochrome P450 enzyme responsible for converting cholesterol into 24S-hydroxycholesterol, which serves as the primary pathway for eliminating cholesterol in the brain. We and others have shown that increased activity of CYP46A1 leads to reduced levels of cholesterol and has a positive effect on cognition. Therefore, we hypothesized that CYP46A1 could be a potential therapeutic target in Niemann-Pick type C (NPC) disease, a rare and fatal neurodegenerative disorder, characterized by cholesterol accumulation in endolysosomal compartments. Herein, we show that CYP46A1 ectopic expression, in cellular models of NPC and in Npc1tm(I1061T) mice by adeno-associated virus-mediated gene therapy improved NPC disease phenotype. Amelioration in functional, biochemical, molecular and neuropathological hallmarks of NPC disease were characterized. In vivo, CYP46A1 expression partially prevented weight loss and hepatomegaly, corrected the expression levels of genes involved in cholesterol homeostasis, and promoted a redistribution of brain cholesterol accumulated in late endosomes/lysosomes. Moreover, concomitant with the amelioration of cholesterol metabolism dysregulation, CYP46A1 attenuated microgliosis and lysosomal dysfunction in mouse cerebellum, favoring a pro-resolving phenotype. In vivo CYP46A1 ectopic expression improves important features of NPC disease and may represent a valid therapeutic approach to be used concomitantly with other drugs. However, promoting cholesterol redistribution does not appear to be enough to prevent Purkinje neuronal death in the cerebellum. This indicates that cholesterol buildup in neurons might not be the main cause of neurodegeneration in this human lipidosis.
Collapse
Affiliation(s)
- Maria João Nunes
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal; Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Andreia Neves Carvalho
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal; Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Joana Reis
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Daniela Costa
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Miguel Moutinho
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Joana Mateus
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Rita Mendes de Almeida
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Sara Brito
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Daniela Risso
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Sofia Nunes
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Margarida Castro-Caldas
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal; Department of Life Sciences, Faculty of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Maria João Gama
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal; Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Cecília M P Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal; Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Sara Xapelli
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Maria José Diógenes
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Nathalie Cartier
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, APHP, University Hospital Pitié Salpêtrière, Paris, France
| | - Farah Chali
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, APHP, University Hospital Pitié Salpêtrière, Paris, France
| | - Françoise Piguet
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, APHP, University Hospital Pitié Salpêtrière, Paris, France
| | - Elsa Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal; Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
18
|
Shu HJ, Ziolkowski LH, Salvatore SV, Benz AM, Wozniak DF, Yuede CM, Paul SM, Zorumski CF, Mennerick S. Effects of Complete and Partial Loss of the 24S-Hydroxycholesterol-Generating Enzyme Cyp46a1 on Behavior and Hippocampal Transcription in Mouse. Biomolecules 2024; 14:254. [PMID: 38540675 PMCID: PMC10968171 DOI: 10.3390/biom14030254] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/09/2024] [Accepted: 02/15/2024] [Indexed: 12/09/2024] Open
Abstract
Brain cholesterol metabolic products include neurosteroids and oxysterols, which play important roles in cellular physiology. In neurons, the cholesterol oxidation product, 24S-hydroxycholesterol (24S-HC), is a regulator of signaling and transcription. Here, we examined the behavioral effects of 24S-HC loss, using global and cell-selective genetic deletion of the synthetic enzyme CYP46A1. Mice that are globally deficient in CYP46A1 exhibited hypoactivity at young ages and unexpected increases in conditioned fear memory. Despite strong reductions in hippocampal 24S-HC in mice with selective loss of CYP46A1 in VGLUT1-positive cells, behavioral effects were not recapitulated in these conditional knockout mice. Global knockout produced strong, developmentally dependent transcriptional effects on select cholesterol metabolism genes. These included paradoxical changes in Liver X Receptor targets. Again, conditional knockout was insufficient to recapitulate most changes. Overall, our results highlight the complex effects of 24S-HC in an in vivo setting that are not fully predicted by known mechanisms. The results also demonstrate that the complete inhibition of enzymatic activity may be needed for a detectable, therapeutically relevant impact on gene expression and behavior.
Collapse
Affiliation(s)
- Hong-Jin Shu
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA (S.V.S.); (D.F.W.); (C.M.Y.); (S.M.P.)
| | - Luke H. Ziolkowski
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA (S.V.S.); (D.F.W.); (C.M.Y.); (S.M.P.)
| | - Sofia V. Salvatore
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA (S.V.S.); (D.F.W.); (C.M.Y.); (S.M.P.)
| | - Ann M. Benz
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA (S.V.S.); (D.F.W.); (C.M.Y.); (S.M.P.)
| | - David F. Wozniak
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA (S.V.S.); (D.F.W.); (C.M.Y.); (S.M.P.)
- Taylor Family Institute for Innovative Psychiatry Research, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Carla M. Yuede
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA (S.V.S.); (D.F.W.); (C.M.Y.); (S.M.P.)
| | - Steven M. Paul
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA (S.V.S.); (D.F.W.); (C.M.Y.); (S.M.P.)
- Taylor Family Institute for Innovative Psychiatry Research, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Charles F. Zorumski
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA (S.V.S.); (D.F.W.); (C.M.Y.); (S.M.P.)
- Taylor Family Institute for Innovative Psychiatry Research, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Steven Mennerick
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA (S.V.S.); (D.F.W.); (C.M.Y.); (S.M.P.)
- Taylor Family Institute for Innovative Psychiatry Research, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
19
|
Mast N, Li Y, Pikuleva IA. 7,8-Dihydroxy Efavirenz Is Not as Effective in CYP46A1 Activation In Vivo as Efavirenz or Its 8,14-Dihydroxy Metabolite. Int J Mol Sci 2024; 25:2242. [PMID: 38396919 PMCID: PMC10889178 DOI: 10.3390/ijms25042242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/08/2024] [Accepted: 02/10/2024] [Indexed: 02/25/2024] Open
Abstract
High dose (S)-efavirenz (EFV) inhibits the HIV reverse transcriptase enzyme and is used to lower HIV load. Low-dose EFV allosterically activates CYP46A1, the key enzyme for cholesterol elimination from the brain, and is investigated as a potential treatment for Alzheimer's disease. Simultaneously, we evaluate EFV dihydroxymetabolites for in vivo brain effects to compare with those of (S)-EFV. We have already tested (rac)-8,14dihydroxy EFV on 5XFAD mice, a model of Alzheimer's disease. Herein, we treated 5XFAD mice with (rac)-7,8dihydroxy EFV. In both sexes, the treatment modestly activated CYP46A1 in the brain and increased brain content of acetyl-CoA and acetylcholine. Male mice also showed a decrease in the brain levels of insoluble amyloid β40 peptides. However, the treatment had no effect on animal performance in different memory tasks. Thus, the overall brain effects of (rac)-7,8dihydroxy EFV were weaker than those of EFV and (rac)-8,14dihydroxy EFV and did not lead to cognitive improvements as were seen in treatments with EFV and (rac)-8,14dihydroxy EFV. An in vitro study assessing CYP46A1 activation in co-incubations with EFV and (rac)-7,8dihydroxy EFV or (rac)-8,14dihydroxy EFV was carried out and provided insight into the compound doses and ratios that could be used for in vivo co-treatments with EFV and its dihydroxymetabolite.
Collapse
Affiliation(s)
| | | | - Irina A. Pikuleva
- Department of Ophthalmology and Visual Science, Case Western Reserve University, Cleveland, OH 44106, USA; (N.M.); (Y.L.)
| |
Collapse
|
20
|
Masson EAY, Serrano J, Leger-Charnay E, Acar N. Cholesterol and oxysterols in retinal neuron-glia interactions: relevance for glaucoma. FRONTIERS IN OPHTHALMOLOGY 2024; 3:1303649. [PMID: 38983043 PMCID: PMC11182186 DOI: 10.3389/fopht.2023.1303649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 12/04/2023] [Indexed: 07/11/2024]
Abstract
Cholesterol is an essential component of cellular membranes, crucial for maintaining their structural and functional integrity. It is especially important for nervous tissues, including the retina, which rely on high amounts of plasma membranes for the transmission of the nervous signal. While cholesterol is by far the most abundant sterol, the retina also contains cholesterol precursors and metabolites, especially oxysterols, which are bioactive molecules. Cholesterol lack or excess is deleterious and some oxysterols are known for their effect on neuron survival. Cholesterol homeostasis must therefore be maintained. Retinal glial cells, especially Müller cells, the principal glial cells of the vertebrate retina, provide mechanical, nutritional, and metabolic support for the neighboring neurons. Several pieces of evidence indicate that Müller cells are major actors of cholesterol homeostasis in the retina, as it is known for other glial cells in the brain. This process is based on a close cooperation with neurons, and sterols can be signaling molecules participating in glia-neuron interactions. While some implication of cholesterol in age-related macular degeneration is now recognized, based on epidemiological and laboratory data, evidence for its role in glaucoma is still scarce. The association between cholesterolemia and glaucoma is controversial, but experimental data suggest that sterols could take part in the pathological processes. It has been demonstrated that Müller glial cells are implicated in the development of glaucoma through an ambivalent reactive retinal gliosis process. The early steps contribute to maintaining retinal homeostasis and favor the survival of ganglion cells, which are targeted during glaucoma. If gliosis persists, dysregulation of the neuroprotective functions, cytotoxic effects of gliotic Müller cells and disruption of glia-neuron interactions lead to an acceleration of ganglion cell death. Sterols could play a role in the glial cell response to glaucomatous injury. This represents an understudied but attractive topic to better understand glaucoma and conceive novel preventive or curative strategies. The present review describes the current knowledge on i) sterol metabolism in retinal glial cells, ii) the potential role of cholesterol in glaucoma, and iii) the possible relationships between cholesterol and oxysterols, glial cells and glaucoma. Focus is put on glia-neuron interactions.
Collapse
Affiliation(s)
- Elodie A Y Masson
- Eye and Nutrition Research Group, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne, Dijon, France
| | - Jeanne Serrano
- Eye and Nutrition Research Group, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne, Dijon, France
- Sensory Perception, Glia/Neuron Interaction Research Group, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne, Dijon, France
| | - Elise Leger-Charnay
- Eye and Nutrition Research Group, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne, Dijon, France
| | - Niyazi Acar
- Eye and Nutrition Research Group, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne, Dijon, France
| |
Collapse
|
21
|
Ghosh S, Ghzaiel I, Vejux A, Meaney S, Nag S, Lizard G, Tripathi G, Naez F, Paul S. Impact of Oxysterols in Age-Related Disorders and Strategies to Alleviate Adverse Effects. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1440:163-191. [PMID: 38036880 DOI: 10.1007/978-3-031-43883-7_9] [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
Oxysterols or cholesterol oxidation products are a class of molecules with the sterol moiety, derived from oxidative reaction of cholesterol through enzymatic and non-enzymatic processes. They are widely reported in animal-origin foods and prove significant involvement in the regulation of cholesterol homeostasis, lipid transport, cellular signaling, and other physiological processes. Reports of oxysterol-mediated cytotoxicity are in abundance and thus consequently implicated in several age-related and lifestyle disorders such as cardiovascular diseases, bone disorders, pancreatic disorders, age-related macular degeneration, cataract, neurodegenerative disorders such as Alzheimer's and Parkinson's disease, and some types of cancers. In this chapter, we attempt to review a selection of physiologically relevant oxysterols, with a focus on their formation, properties, and roles in health and disease, while also delving into the potential of natural and synthetic molecules along with bacterial enzymes for mitigating oxysterol-mediated cell damage.
Collapse
Affiliation(s)
- Shubhrima Ghosh
- Trinity Translational Medicine Institute, School of Medicine, Trinity College Dublin, Dublin 8, Ireland
| | - Imen Ghzaiel
- Bio-PeroxIL Laboratory, EA7270, University of Bourgogne & Inserm, Dijon, France
- Faculty of Medicine, Laboratory 'Nutrition, Functional Food and Vascular Health' (LR12ES05), University of Monastir, Monastir, Tunisia
| | - Anne Vejux
- Bio-PeroxIL Laboratory, EA7270, University of Bourgogne & Inserm, Dijon, France
| | - Steve Meaney
- School of Biological, Health and Sports Sciences, Technological University Dublin, Dublin 7, Ireland
| | - Sagnik Nag
- Department of Bio-Sciences, School of Biosciences & Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - Gérard Lizard
- Bio-PeroxIL Laboratory, EA7270, University of Bourgogne & Inserm, Dijon, France
| | - Garima Tripathi
- Department of Bio-Sciences, School of Biosciences & Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - Falal Naez
- Department of Microbiology, Vijaygarh Jyotish Ray College, University of Calcutta, Kolkata, India
| | - Srijita Paul
- Department of Microbiology, Gurudas College, Kolkata, West Bengal, India
| |
Collapse
|
22
|
Du J, Chen Z, Chen X, Zhang J, Wang Y, Zhao T, Wang D, Wang C, Chen Y, Meng Q, Sun H, Liu K, Wu J. Inhibition of Glycyrrhiza Polysaccharide on Human Cytochrome P450 46A1 in vitro and in vivo: Implications in Treating Neurological Diseases. Curr Drug Metab 2024; 25:227-234. [PMID: 38797896 DOI: 10.2174/0113892002305873240520072802] [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: 01/23/2024] [Revised: 04/14/2024] [Accepted: 04/18/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND Cytochrome P450 (CYP) 46A1, also known as cholesterol 24S-hydroxylase, is essential for maintaining the homeostasis of cholesterol in the brain and serves as a therapeutic target of neurodegenerative disorders and excitatory neurotoxicity. N-methyl-d-aspartate receptor (NMDAR) is a prototypical receptor for the excitatory neurotransmitter glutamate and can be specifically regulated by 24S-hydroxycholesterol (24S-HC). Glycyrrhiza is one of the most widely used herbs with broad clinical applications, which has several pharmacological activities, such as clearing heat and detoxifying, moistening the lung and relieving cough, analgesic, neuroprotective outcomes, and regulating a variety of drug activities. Glycyrrhiza is a commonly used herb for the treatment of epileptic encephalopathy. However, whether glycyrrhiza can interfere with the activity of CYP46A1 remains unknown. OBJECTIVE This study aimed to investigate the regulating effects of glycyrrhiza polysaccharides (GP) on CYP46A1-mediated cholesterol conversion, as well as in the modulation of related proteins. MATERIALS AND METHODS The effects of glycyrrhiza polysaccharide (GP) on the activity of CYP46A1 were investigated in vivo and in vitro. Moreover, the potential regulatory effects of GP on the expressions of CYP46A1, HMG-CoA reductase (HMGCR), and NMDAR were also detected. RESULTS The in vitro results demonstrated that glycyrrhiza polysaccharide (GP), as the main water-soluble active component of glycyrrhiza, remarkably inhibited the activity of CYP46A1 in a non-competitive mode with a Ki value of 0.7003 mg/ml. Furthermore, the in vivo experiments verified that GP markedly decreased the contents of 24S-HC in rat plasma and brain tissues as compared to the control. More importantly, the protein expressions of CYP46A1, GluN2A, GluN2B, and HMG-CoA reductase (HMGCR) in rat brains were all downregulated, whereas the mRNA expressions of CYP46A1 and HMGCR were not significantly changed after treatment with GP. CONCLUSION GP exhibits a significant inhibitory effect on CYP46A1 activity in vitro and in vivo, and the protein expressions of CYP46A1, HMGCR, and NMDAR are also inhibited by GP, which are of considerable clinical significance for GP's potential therapeutic role in treating neurological diseases.
Collapse
Affiliation(s)
- Jie Du
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Zujia Chen
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Xiaodong Chen
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Jiahui Zhang
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Yaojun Wang
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Tingting Zhao
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Dalong Wang
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Changyuan Wang
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Liaoning Dalian Medical University, Dalian, Liaoning, China
| | - Yanwei Chen
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Qiang Meng
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Liaoning Dalian Medical University, Dalian, Liaoning, China
| | - Huijun Sun
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Liaoning Dalian Medical University, Dalian, Liaoning, China
| | - Kexin Liu
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Liaoning Dalian Medical University, Dalian, Liaoning, China
| | - Jingjing Wu
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Liaoning Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
23
|
Binda CS, Lelos MJ, Rosser AE, Massey TH. Using gene or cell therapies to treat Huntington's disease. HANDBOOK OF CLINICAL NEUROLOGY 2024; 205:193-215. [PMID: 39341655 DOI: 10.1016/b978-0-323-90120-8.00014-9] [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: 10/01/2024]
Abstract
Huntington's disease is caused by a CAG repeat expansion in the first exon of the HTT gene, leading to the production of gain-of-toxic-function mutant huntingtin protein species and consequent transcriptional dysregulation and disrupted cell metabolism. The brunt of the disease process is borne by the striatum from the earliest disease stages, with striatal atrophy beginning approximately a decade prior to the onset of neurologic signs. Although the expanded CAG repeat in the HTT gene is necessary and sufficient to cause HD, other genes can influence the age at onset of symptoms and how they progress. Many of these modifier genes have roles in DNA repair and are likely to modulate the stability of the CAG repeat in somatic cells. Currently, there are no disease-modifying treatments for HD that can be prescribed to patients and few symptomatic treatments, but there is a lot of interest in therapeutics that can target the pathogenic pathways at the DNA and RNA levels, some of which have reached the stage of human studies. In contrast, cell therapies aim to replace key neural cells lost to the disease process and/or to support the host vulnerable striatum by direct delivery of cells to the brain. Ultimately it may be possible to combine gene and cell therapies to both slow disease processes and provide some level of neural repair. In this chapter we consider the current status of these therapeutic strategies along with their prospects and challenges.
Collapse
Affiliation(s)
- Caroline S Binda
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom; UK Dementia Research Institute at Cardiff, Cardiff University, Cardiff, United Kingdom
| | - Mariah J Lelos
- Brain Repair Group, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Anne E Rosser
- Brain Repair Group, School of Biosciences, Cardiff University, Cardiff, United Kingdom; BRAIN Unit, Neuroscience and Mental Health Research Institute, Cardiff, United Kingdom.
| | - Thomas H Massey
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom; UK Dementia Research Institute at Cardiff, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
24
|
Messedi M, Makni-Ayadi F. 24S-Hydroxycholesterol in Neuropsychiatric Diseases: Schizophrenia, Autism Spectrum Disorder, and Bipolar Disorder. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1440:293-304. [PMID: 38036886 DOI: 10.1007/978-3-031-43883-7_15] [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
Neuropsychiatric diseases (NPDs) are severe, debilitating psychiatric conditions that affect the nervous system. These are among the most challenging disorders in medicine. Some examples include Alzheimer's, anxiety disorders, autism spectrum disorder, bipolar disorder, and schizophrenia. NPDs represent an ever-increasing burden on public health and are prevalent throughout the world. For most of these diseases, the particular etiopathogeneses are still enigmatic. NPDs are also associated with structural and functional changes in the brain, along with altered neurotransmitter and neuroendocrine systems.Approximately 25% of the total human body cholesterol is located in the brain. Its involvement in neuronal functions starts in the early growth stages and remains important throughout adulthood. It is also an integral part of the neuronal membrane, ensuring membrane lipid organization and regulating membrane fluidity. The main mechanism for removing cholesterol from the brain is cholesterol 24-hydroxylation by cytochrome P450 46A1 (CYP46A1), an enzyme specifically found in the central nervous system. Although research on 24S-OHC and its role in neuropsychiatric diseases is still in its early stages, this oxidized cholesterol metabolite is thought to play a crucial role in the etiology of NPDs. 24S-OHC can affect neurons, astrocytes, oligodendrocytes, and vascular cells. In addition to regulating the homeostasis of cholesterol in the brain, this oxysterol is involved in neurotransmission, oxidative stress, and inflammation. The role of 24S-OHC in NPDs has been found to be controversial in terms of the findings so far. There are several intriguing discrepancies in the data gathered so far regarding 24S-OHC and NPDs. In fact, 24S-OHC levels were reported to have decreased in a number of NPDs and increased in others.Hence, in this chapter, we first summarize the available data regarding 24S-OHC as a biomarker in NPDs, including schizophrenia, autism spectrum disorder, and bipolar disorder. Then, we present a brief synopsis of the pharmacological targeting of 24S-OHC levels through the modulation of CYP46A1 activity.
Collapse
Affiliation(s)
- Meriam Messedi
- Research Laboratory "Molecular Basis of Human Diseases", LR19ES13, Sfax Medicine School, University of Sfax, Sfax, Tunisia
| | - Fatma Makni-Ayadi
- Research Laboratory "Molecular Basis of Human Diseases", LR19ES13, Sfax Medicine School, University of Sfax, Sfax, Tunisia
- Department of Clinical biochemistry, Habib Bourguiba Hospital, Sfax, Tunisia
| |
Collapse
|
25
|
Urano Y, Noguchi N. Enzymatically Formed Oxysterols and Cell Death. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1440:193-211. [PMID: 38036881 DOI: 10.1007/978-3-031-43883-7_10] [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
The side-chain hydroxylation of cholesterol by specific enzymes produces 24(S)-hydroxycholesterol, 25-hydroxycholesterol, 27-hydroxycholesterol, and other products. These enzymatically formed side-chain oxysterols act as intermediates in the biosynthesis of bile acids and serve as signaling molecules that regulate cholesterol homeostasis. Besides these intracellular functions, an imbalance in oxysterol homeostasis is implicated in pathophysiology. Furthermore, growing evidence reveals that oxysterols affect cell proliferation and cause cell death. This chapter provides an overview of the pathophysiological role of side-chain oxysterols in developing human diseases. We also summarize our understanding of the molecular mechanisms underlying the induction of various forms of cell death by side-chain oxysterols.
Collapse
Affiliation(s)
- Yasuomi Urano
- Department of Medical Life Systems, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe, Kyoto, Japan.
| | - Noriko Noguchi
- Department of Medical Life Systems, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe, Kyoto, Japan
| |
Collapse
|
26
|
Vigne S, Pot C. Implication of Oxysterols and Phytosterols in Aging and Human Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1440:231-260. [PMID: 38036883 DOI: 10.1007/978-3-031-43883-7_12] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Cholesterol is easily oxidized and can be transformed into numerous oxidation products, among which oxysterols. Phytosterols are plant sterols related to cholesterol. Both oxysterols and phytosterols can have an impact on human health and diseases.Cholesterol is a member of the sterol family that plays essential roles in biological processes, including cell membrane stability and myelin formation. Cholesterol can be metabolized into several molecules including bile acids, hormones, and oxysterols. On the other hand, phytosterols are plant-derived compounds structurally related to cholesterol, which can also have an impact on human health. Here, we review the current knowledge about the role of oxysterols and phytosterols on human health and focus on the impact of their pathways on diseases of the central nervous system (CNS), autoimmune diseases, including inflammatory bowel diseases (IBD), vascular diseases, and cancer in both experimental models and human studies. We will first discuss the implications of oxysterols and then of phytosterols in different human diseases.
Collapse
Affiliation(s)
- Solenne Vigne
- Laboratories of Neuroimmunology, Service of Neurology and Neuroscience Research Center, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Epalinges, Lausanne, Switzerland
| | - Caroline Pot
- Laboratories of Neuroimmunology, Service of Neurology and Neuroscience Research Center, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Epalinges, Lausanne, Switzerland.
| |
Collapse
|
27
|
Brunet de Courssou JB, Deiva K. Les thérapies géniques en neurologie. PRATIQUE NEUROLOGIQUE - FMC 2023; 14:208-224. [DOI: 10.1016/j.praneu.2023.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
28
|
Tsitsou-Kampeli A, Suzzi S, Kenigsbuch M, Satomi A, Strobelt R, Singer O, Feldmesser E, Purnapatre M, Colaiuta SP, David E, Cahalon L, Hahn O, Wyss-Coray T, Shaul Y, Amit I, Schwartz M. Cholesterol 24-hydroxylase at the choroid plexus contributes to brain immune homeostasis. Cell Rep Med 2023; 4:101278. [PMID: 37944529 PMCID: PMC10694665 DOI: 10.1016/j.xcrm.2023.101278] [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: 11/15/2022] [Revised: 05/26/2023] [Accepted: 10/11/2023] [Indexed: 11/12/2023]
Abstract
The choroid plexus (CP) plays a key role in remotely controlling brain function in health, aging, and disease. Here, we report that CP epithelial cells express the brain-specific cholesterol 24-hydroxylase (CYP46A1) and that its levels are decreased under different mouse and human brain conditions, including amyloidosis, aging, and SARS-CoV-2 infection. Using primary mouse CP cell cultures, we demonstrate that the enzymatic product of CYP46A1, 24(S)-hydroxycholesterol, downregulates inflammatory transcriptomic signatures within the CP, found here to be elevated across multiple neurological conditions. In vitro, the pro-inflammatory cytokine tumor necrosis factor α (TNF-α) downregulates CYP46A1 expression, while overexpression of CYP46A1 or its pharmacological activation in mouse CP organ cultures increases resilience to TNF-α. In vivo, overexpression of CYP46A1 in the CP in transgenic mice with amyloidosis is associated with better cognitive performance and decreased brain inflammation. Our findings suggest that CYP46A1 expression in the CP impacts the role of this niche as a guardian of brain immune homeostasis.
Collapse
Affiliation(s)
| | - Stefano Suzzi
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel.
| | - Mor Kenigsbuch
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel; Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Akisawa Satomi
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel; Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Romano Strobelt
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Oded Singer
- Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Ester Feldmesser
- Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | | | | | - Eyal David
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Liora Cahalon
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Oliver Hahn
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Yosef Shaul
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Ido Amit
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Michal Schwartz
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
29
|
Meza U, Romero-Méndez C, Sánchez-Armáss S, Rodríguez-Menchaca AA. Role of rafts in neurological disorders. Neurologia 2023; 38:671-680. [PMID: 37858892 DOI: 10.1016/j.nrleng.2023.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 01/01/2021] [Indexed: 10/21/2023] Open
Abstract
INTRODUCTION Rafts are protein-lipid structural nanodomains involved in efficient signal transduction and the modulation of physiological processes of the cell plasma membrane. Raft disruption in the nervous system has been associated with a wide range of disorders. DEVELOPMENT We review the concept of rafts, the nervous system processes in which they are involved, and their role in diseases such as Parkinson's disease, Alzheimer disease, and Huntington disease. CONCLUSIONS Based on the available evidence, preservation and/or reconstitution of rafts is a promising treatment strategy for a wide range of neurological disorders.
Collapse
Affiliation(s)
- U Meza
- Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México.
| | - C Romero-Méndez
- Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México.
| | - S Sánchez-Armáss
- Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México.
| | - A A Rodríguez-Menchaca
- Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México.
| |
Collapse
|
30
|
Lalloyer F, Mogilenko DA, Verrijken A, Haas JT, Lamazière A, Kouach M, Descat A, Caron S, Vallez E, Derudas B, Gheeraert C, Baugé E, Despres G, Dirinck E, Tailleux A, Dombrowicz D, Van Gaal L, Eeckhoute J, Lefebvre P, Goossens JF, Francque S, Staels B. Roux-en-Y gastric bypass induces hepatic transcriptomic signatures and plasma metabolite changes indicative of improved cholesterol homeostasis. J Hepatol 2023; 79:898-909. [PMID: 37230231 DOI: 10.1016/j.jhep.2023.05.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 04/18/2023] [Accepted: 05/08/2023] [Indexed: 05/27/2023]
Abstract
BACKGROUND & AIMS Roux-en-Y gastric bypass (RYGB), the most effective surgical procedure for weight loss, decreases obesity and ameliorates comorbidities, such as non-alcoholic fatty liver (NAFLD) and cardiovascular (CVD) diseases. Cholesterol is a major CVD risk factor and modulator of NAFLD development, and the liver tightly controls its metabolism. How RYGB surgery modulates systemic and hepatic cholesterol metabolism is still unclear. METHODS We studied the hepatic transcriptome of 26 patients with obesity but not diabetes before and 1 year after undergoing RYGB. In parallel, we measured quantitative changes in plasma cholesterol metabolites and bile acids (BAs). RESULTS RYGB surgery improved systemic cholesterol metabolism and increased plasma total and primary BA levels. Transcriptomic analysis revealed specific alterations in the liver after RYGB, with the downregulation of a module of genes implicated in inflammation and the upregulation of three modules, one associated with BA metabolism. A dedicated analysis of hepatic genes related to cholesterol homeostasis pointed towards increased biliary cholesterol elimination after RYGB, associated with enhancement of the alternate, but not the classical, BA synthesis pathway. In parallel, alterations in the expression of genes involved in cholesterol uptake and intracellular trafficking indicate improved hepatic free cholesterol handling. Finally, RYGB decreased plasma markers of cholesterol synthesis, which correlated with an improvement in liver disease status after surgery. CONCLUSIONS Our results identify specific regulatory effects of RYGB on inflammation and cholesterol metabolism. RYGB alters the hepatic transcriptome signature, likely improving liver cholesterol homeostasis. These gene regulatory effects are reflected by systemic post-surgery changes of cholesterol-related metabolites, corroborating the beneficial effects of RYGB on both hepatic and systemic cholesterol homeostasis. IMPACT AND IMPLICATIONS Roux-en-Y gastric bypass (RYGB) is a widely used bariatric surgery procedure with proven efficacy in body weight management, combatting cardiovascular disease (CVD) and non-alcoholic fatty liver disease (NAFLD). RYGB exerts many beneficial metabolic effects, by lowering plasma cholesterol and improving atherogenic dyslipidemia. Using a cohort of patients undergoing RYGB, studied before and 1 year after surgery, we analyzed how RYGB modulates hepatic and systemic cholesterol and bile acid metabolism. The results of our study provide important insights on the regulation of cholesterol homeostasis after RYGB and open avenues that could guide future monitoring and treatment strategies targeting CVD and NAFLD in obesity.
Collapse
Affiliation(s)
- Fanny Lalloyer
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Denis A Mogilenko
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France; Department of Medicine, Department of Pathology, Microbiology and Immunology, Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Ann Verrijken
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, 2610, Wilrijk, Antwerp, Belgium; Department of Endocrinology, Diabetology and Metabolism, Antwerp University Hospital, 2650, Edegem, Antwerp, Belgium
| | - Joel T Haas
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Antonin Lamazière
- Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint Antoine, Clinical Metabolomic Department, Sorbonne Université, Inserm, F-75012, Paris, France
| | - Mostafa Kouach
- University of Lille, CHU Lille, EA 7365-GRITA-Groupe de Recherche sur les formes Injectables et les Technologies Associées, F-59000, Lille, France
| | - Amandine Descat
- University of Lille, CHU Lille, EA 7365-GRITA-Groupe de Recherche sur les formes Injectables et les Technologies Associées, F-59000, Lille, France
| | - Sandrine Caron
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Emmanuelle Vallez
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Bruno Derudas
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Céline Gheeraert
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Eric Baugé
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Gaëtan Despres
- Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint Antoine, Clinical Metabolomic Department, Sorbonne Université, Inserm, F-75012, Paris, France
| | - Eveline Dirinck
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, 2610, Wilrijk, Antwerp, Belgium; Department of Endocrinology, Diabetology and Metabolism, Antwerp University Hospital, 2650, Edegem, Antwerp, Belgium
| | - Anne Tailleux
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - David Dombrowicz
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Luc Van Gaal
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, 2610, Wilrijk, Antwerp, Belgium; Department of Endocrinology, Diabetology and Metabolism, Antwerp University Hospital, 2650, Edegem, Antwerp, Belgium
| | - Jerôme Eeckhoute
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Philippe Lefebvre
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France
| | - Jean-François Goossens
- University of Lille, CHU Lille, EA 7365-GRITA-Groupe de Recherche sur les formes Injectables et les Technologies Associées, F-59000, Lille, France
| | - Sven Francque
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, 2610, Wilrijk, Antwerp, Belgium; Department of Gastroenterology and Hepatology, Antwerp University Hospital, ERN RARE-LIVER, 2650, Edegem, Antwerp, Belgium
| | - Bart Staels
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000, Lille, France.
| |
Collapse
|
31
|
Valenza M, Birolini G, Cattaneo E. The translational potential of cholesterol-based therapies for neurological disease. Nat Rev Neurol 2023; 19:583-598. [PMID: 37644213 DOI: 10.1038/s41582-023-00864-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2023] [Indexed: 08/31/2023]
Abstract
Cholesterol is an important metabolite and membrane component and is enriched in the brain owing to its role in neuronal maturation and function. In the adult brain, cholesterol is produced locally, predominantly by astrocytes. When cholesterol has been used, recycled and catabolized, the derivatives are excreted across the blood-brain barrier. Abnormalities in any of these steps can lead to neurological dysfunction. Here, we examine how precise interactions between cholesterol production and its use and catabolism in neurons ensures cholesterol homeostasis to support brain function. As an example of a neurological disease associated with cholesterol dyshomeostasis, we summarize evidence from animal models of Huntington disease (HD), which demonstrate a marked reduction in cholesterol biosynthesis with clinically relevant consequences for synaptic activity and cognition. In addition, we examine the relationship between cholesterol loss in the brain and cognitive decline in ageing. We then present emerging therapeutic strategies to restore cholesterol homeostasis, focusing on evidence from HD mouse models.
Collapse
Affiliation(s)
- Marta Valenza
- Department of Biosciences, University of Milan, Milan, Italy.
- Istituto Nazionale di Genetica Molecolare 'Romeo ed Enrica Invernizzi', Milan, Italy.
| | - Giulia Birolini
- Department of Biosciences, University of Milan, Milan, Italy
- Istituto Nazionale di Genetica Molecolare 'Romeo ed Enrica Invernizzi', Milan, Italy
| | - Elena Cattaneo
- Department of Biosciences, University of Milan, Milan, Italy.
- Istituto Nazionale di Genetica Molecolare 'Romeo ed Enrica Invernizzi', Milan, Italy.
| |
Collapse
|
32
|
Birolini G, Valenza M, Ottonelli I, Talpo F, Minoli L, Cappelleri A, Bombaci M, Caccia C, Canevari C, Trucco A, Leoni V, Passoni A, Favagrossa M, Nucera MR, Colombo L, Paltrinieri S, Bagnati R, Duskey JT, Caraffi R, Vandelli MA, Taroni F, Salmona M, Scanziani E, Biella G, Ruozi B, Tosi G, Cattaneo E. Chronic cholesterol administration to the brain supports complete and long-lasting cognitive and motor amelioration in Huntington's disease. Pharmacol Res 2023; 194:106823. [PMID: 37336430 PMCID: PMC10463277 DOI: 10.1016/j.phrs.2023.106823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/14/2023] [Accepted: 06/16/2023] [Indexed: 06/21/2023]
Abstract
Evidence that Huntington's disease (HD) is characterized by impaired cholesterol biosynthesis in the brain has led to strategies to increase its level in the brain of the rapidly progressing R6/2 mouse model, with a positive therapeutic outcome. Here we tested the long-term efficacy of chronic administration of cholesterol to the brain of the slowly progressing zQ175DN knock-in HD mice in preventing ("early treatment") or reversing ("late treatment") HD symptoms. To do this we used the most advanced formulation of cholesterol loaded brain-permeable nanoparticles (NPs), termed hybrid-g7-NPs-chol, which were injected intraperitoneally. We show that one cycle of treatment with hybrid-g7-NPs-chol, administered in the presymptomatic ("early treatment") or symptomatic ("late treatment") stages is sufficient to normalize cognitive defects up to 5 months, as well as to improve other behavioral and neuropathological parameters. A multiple cycle treatment combining both early and late treatments ("2 cycle treatment") lasting 6 months generates therapeutic effects for more than 11 months, without severe adverse reactions. Sustained cholesterol delivery to the brain of zQ175DN mice also reduces mutant Huntingtin aggregates in both the striatum and cortex and completely normalizes synaptic communication in the striatal medium spiny neurons compared to saline-treated HD mice. Furthermore, through a meta-analysis of published and current data, we demonstrated the power of hybrid-g7-NPs-chol and other strategies able to increase brain cholesterol biosynthesis, to reverse cognitive decline and counteract the formation of mutant Huntingtin aggregates. These results demonstrate that cholesterol delivery via brain-permeable NPs is a therapeutic option to sustainably reverse HD-related behavioral decline and neuropathological signs over time, highlighting the therapeutic potential of cholesterol-based strategies in HD patients. DATA AVAILABILITY: This study does not include data deposited in public repositories. Data are available on request to the corresponding authors.
Collapse
Affiliation(s)
- Giulia Birolini
- Department of Biosciences, University of Milan, 20133 Milan, Italy; Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi", 20122 Milan, Italy
| | - Marta Valenza
- Department of Biosciences, University of Milan, 20133 Milan, Italy; Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi", 20122 Milan, Italy.
| | - Ilaria Ottonelli
- Nanotech Lab, Te.Far.T.I. Center, Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Francesca Talpo
- Department of Biology and Biotechnologies, University of Pavia, 27100 Pavia, Italy
| | - Lucia Minoli
- Pathology Department, Evotec, 37135 Verona, Italy; Mouse & Animal Pathology Lab (MAPLab), Fondazione UniMi, 20139 Milan, Italy
| | - Andrea Cappelleri
- Dipartimento di Medicina Veterinaria e Scienze Animali, Università degli Studi di Milano, 26900 Lodi, Italy; Mouse & Animal Pathology Lab (MAPLab), Fondazione UniMi, 20139 Milan, Italy
| | - Mauro Bombaci
- Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi", 20122 Milan, Italy
| | - Claudio Caccia
- Unit of Medical Genetics and Neurogenetics. Fondazione IRCCS Istituto Neurologico Carlo Besta, 20131 Milan, Italy
| | - Caterina Canevari
- Department of Biology and Biotechnologies, University of Pavia, 27100 Pavia, Italy
| | - Arianna Trucco
- Department of Biology and Biotechnologies, University of Pavia, 27100 Pavia, Italy
| | - Valerio Leoni
- Laboratory of Clinical Chemistry, Hospital Pio XI of Desio, ASST-Brianza and Department of Medicine and Surgery, University of Milano Bicocca, 20900 Monza, Italy
| | - Alice Passoni
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
| | - Monica Favagrossa
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
| | - Maria Rosaria Nucera
- Department of Biosciences, University of Milan, 20133 Milan, Italy; Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi", 20122 Milan, Italy
| | - Laura Colombo
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
| | - Saverio Paltrinieri
- Dipartimento di Medicina Veterinaria e Scienze Animali, Università degli Studi di Milano, 26900 Lodi, Italy
| | - Renzo Bagnati
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
| | - Jason Thomas Duskey
- Nanotech Lab, Te.Far.T.I. Center, Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Riccardo Caraffi
- Nanotech Lab, Te.Far.T.I. Center, Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Maria Angela Vandelli
- Nanotech Lab, Te.Far.T.I. Center, Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Franco Taroni
- Unit of Medical Genetics and Neurogenetics. Fondazione IRCCS Istituto Neurologico Carlo Besta, 20131 Milan, Italy
| | - Mario Salmona
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
| | - Eugenio Scanziani
- Dipartimento di Medicina Veterinaria e Scienze Animali, Università degli Studi di Milano, 26900 Lodi, Italy; Mouse & Animal Pathology Lab (MAPLab), Fondazione UniMi, 20139 Milan, Italy
| | - Gerardo Biella
- Department of Biology and Biotechnologies, University of Pavia, 27100 Pavia, Italy
| | - Barbara Ruozi
- Nanotech Lab, Te.Far.T.I. Center, Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Giovanni Tosi
- Nanotech Lab, Te.Far.T.I. Center, Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Elena Cattaneo
- Department of Biosciences, University of Milan, 20133 Milan, Italy; Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi", 20122 Milan, Italy.
| |
Collapse
|
33
|
Pinchaud K, Masson C, Dayre B, Mounier C, Gilles JF, Vanhoutte P, Caboche J, Betuing S. Cell-Type Specific Regulation of Cholesterogenesis by CYP46A1 Re-Expression in zQ175 HD Mouse Striatum. Int J Mol Sci 2023; 24:11001. [PMID: 37446179 DOI: 10.3390/ijms241311001] [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: 06/09/2023] [Revised: 06/22/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Cholesterol metabolism dysregulation is associated with several neurological disorders. In Huntington's disease (HD), several enzymes involved in cholesterol metabolism are downregulated, among which the neuronal cholesterol 24-hydroxylase, CYP46A1, is of particular interest. The restoration of CYP46A1 expression in striatal neurons of HD mouse models is beneficial for motor behavior, cholesterol metabolism, transcriptomic activity, and alleviates neuropathological hallmarks induced by mHTT. Among the genes regulated after CYP46A1 restoration, those involved in cholesterol synthesis and efflux may explain the positive effect of CYP46A1 on cholesterol precursor metabolites. Since cholesterol homeostasis results from a fine-tuning between neurons and astrocytes, we quantified the distribution of key genes regulating cholesterol metabolism and efflux in astrocytes and neurons using in situ hybridization coupled with S100β and NeuN immunostaining, respectively. Neuronal expression of CYP46A1 in the striatum of HD zQ175 mice increased key cholesterol synthesis driver genes (Hmgcr, Dhcr24), specifically in neurons. This effect was associated with an increase of the srebp2 transcription factor gene that regulates most of the genes encoding for cholesterol enzymes. However, the cholesterol efflux gene, ApoE, was specifically upregulated in astrocytes by CYP46A1, probably though a paracrine effect. In summary, the neuronal expression of CYP46A1 has a dual and specific effect on neurons and astrocytes, regulating cholesterol metabolism. The neuronal restoration of CYP46A1 in HD paves the way for future strategies to compensate for mHTT toxicity.
Collapse
Affiliation(s)
- Katleen Pinchaud
- Neuroscience Paris Seine, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8246/INSERM U1130, Sorbonne Université, 75005 Paris, France
| | - Chloé Masson
- Neuroscience Paris Seine, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8246/INSERM U1130, Sorbonne Université, 75005 Paris, France
| | - Baptiste Dayre
- Neuroscience Paris Seine, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8246/INSERM U1130, Sorbonne Université, 75005 Paris, France
| | - Coline Mounier
- Neuroscience Paris Seine, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8246/INSERM U1130, Sorbonne Université, 75005 Paris, France
| | - Jean-François Gilles
- Imaging Facility, Institut de Biologie Paris-Seine (IBPS), Sorbonne Université, 75005 Paris, France
| | - Peter Vanhoutte
- Neuroscience Paris Seine, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8246/INSERM U1130, Sorbonne Université, 75005 Paris, France
| | - Jocelyne Caboche
- Neuroscience Paris Seine, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8246/INSERM U1130, Sorbonne Université, 75005 Paris, France
| | - Sandrine Betuing
- Neuroscience Paris Seine, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8246/INSERM U1130, Sorbonne Université, 75005 Paris, France
| |
Collapse
|
34
|
Mast N, El-Darzi N, Li Y, Pikuleva IA. Quantitative characterizations of the cholesterol-related pathways in the retina and brain of hamsters. J Lipid Res 2023:100401. [PMID: 37330011 PMCID: PMC10394389 DOI: 10.1016/j.jlr.2023.100401] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/08/2023] [Accepted: 06/13/2023] [Indexed: 06/19/2023] Open
Abstract
The retina and brain are separated from the systemic circulation by the anatomical barriers, which are permeable (the outer blood-retinal barrier) and impermeable (the blood-brain and inner blood-retina barriers) to cholesterol. Herein we investigated whether the whole-body cholesterol maintenance affects cholesterol homeostasis in the retina and brain. We used hamsters, whose whole-body cholesterol handling is more similar to those in humans than in mice and conducted separate administrations of deuterated water and deuterated cholesterol. We assessed the quantitative significance of the retinal and brain pathways of cholesterol input and compared the results with those from our previous studies in mice. The utility of the measurements in the plasma of deuterated 24-hydroxycholesterol, the major cholesterol elimination product from the brain, was investigated as well. We established that despite a 7-fold higher serum LDL to HDL ratio and other cholesterol-related differences, in situ biosynthesis remained the major source of cholesterol for hamster retina, although its quantitative significance was reduced to 53% as compared to 72-78% in mouse retina. In the brain, the principal pathway of cholesterol input was also the same, in situ biosynthesis, accounting for 94% of the total brain cholesterol input (96% in mice); the interspecies differences pertained to the absolute rates of the total cholesterol input and turnover. We documented the correlations between deuterium enrichments of the brain 24-hydroxycholesterol, brain cholesterol, and plasma 24-hydroxycholesterol, which suggested that deuterium enrichment of plasma 24-hydroxycholesteol could be an in vivo marker of cholesterol elimination and turnover in the brain.
Collapse
Affiliation(s)
- Natalia Mast
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH USA
| | - Nicole El-Darzi
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH USA
| | - Yong Li
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH USA
| | - Irina A Pikuleva
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH USA.
| |
Collapse
|
35
|
Xing C, Huang X, Wang D, Yu D, Hou S, Cui H, Song L. Roles of bile acids signaling in neuromodulation under physiological and pathological conditions. Cell Biosci 2023; 13:106. [PMID: 37308953 PMCID: PMC10258966 DOI: 10.1186/s13578-023-01053-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 05/13/2023] [Indexed: 06/14/2023] Open
Abstract
Bile acids (BA) are important physiological molecules not only mediating nutrients absorption and metabolism in peripheral tissues, but exerting neuromodulation effect in the central nerve system (CNS). The catabolism of cholesterol to BA occurs predominantly in the liver by the classical and alternative pathways, or in the brain initiated by the neuronal-specific enzyme CYP46A1 mediated pathway. Circulating BA could cross the blood brain barrier (BBB) and reach the CNS through passive diffusion or BA transporters. Brain BA might trigger direct signal through activating membrane and nucleus receptors or affecting activation of neurotransmitter receptors. Peripheral BA may also provide the indirect signal to the CNS via farnesoid X receptor (FXR) dependent fibroblast growth factor 15/19 (FGF15/19) pathway or takeda G protein coupled receptor 5 (TGR5) dependent glucagon-like peptide-1 (GLP-1) pathway. Under pathological conditions, alterations in BA metabolites have been discovered as potential pathogenic contributors in multiple neurological disorders. Attractively, hydrophilic ursodeoxycholic acid (UDCA), especially tauroursodeoxycholic acid (TUDCA) can exert neuroprotective roles by attenuating neuroinflammation, apoptosis, oxidative or endoplasmic reticulum stress, which provides promising therapeutic effects for treatment of neurological diseases. This review summarizes recent findings highlighting the metabolism, crosstalk between brain and periphery, and neurological functions of BA to elucidate the important role of BA signaling in the brain under both physiological and pathological conditions.
Collapse
Affiliation(s)
- Chen Xing
- Beijing Institute of Basic Medical Sciences, Taiping Road #27, Beijing, 100850, China.
| | - Xin Huang
- Beijing Institute of Basic Medical Sciences, Taiping Road #27, Beijing, 100850, China
| | - Dongxue Wang
- Beijing Institute of Basic Medical Sciences, Taiping Road #27, Beijing, 100850, China
- College of Pharmacy, Jiamusi University, Jiamusi, 154007, China
| | - Dengjun Yu
- Beijing Institute of Basic Medical Sciences, Taiping Road #27, Beijing, 100850, China
- College of Pharmacy, Jiamusi University, Jiamusi, 154007, China
| | - Shaojun Hou
- Beijing Institute of Basic Medical Sciences, Taiping Road #27, Beijing, 100850, China
- Anhui Medical University, Heifei, 230032, China
| | - Haoran Cui
- Beijing Institute of Basic Medical Sciences, Taiping Road #27, Beijing, 100850, China
| | - Lung Song
- Beijing Institute of Basic Medical Sciences, Taiping Road #27, Beijing, 100850, China.
- Anhui Medical University, Heifei, 230032, China.
| |
Collapse
|
36
|
Frajerman A, Chaumette B, Farabos D, Despres G, Simonard C, Lamazière A, Krebs MO, Kebir O. Membrane Lipids in Ultra-High-Risk Patients: Potential Predictive Biomarkers of Conversion to Psychosis. Nutrients 2023; 15:2215. [PMID: 37432345 DOI: 10.3390/nu15092215] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/01/2023] [Accepted: 05/04/2023] [Indexed: 07/12/2023] Open
Abstract
Alterations in membrane lipids are reported in schizophrenia. However, no conclusion can be drawn regarding the extended and predictive value of these alterations in persons at ultra-high risk of psychosis (UHR). Recent studies suggested that sterols' impact on psychiatric disorders was underestimated. Here, we simultaneously explored sterols, fatty acids (FA), and phospholipids (PL) in UHR persons for the first time. We analysed erythrocyte membrane lipids in 61 UHR persons, including 29 who later converted to psychosis (UHR-C) and 32 who did not (UHC-NC). We used gas chromatography for FA and liquid chromatography tandem with mass spectrometry for sterols and phospholipids. Among UHR individuals, elevated baseline membrane linoleic acid level was associated with conversion to psychosis (26.1% vs. 60.5%, p = 0.02). Combining sterols, FA, and PL membrane composition improved the prediction of psychosis onset (AUC = 0.73). This is the first report showing that membrane sterol participates, with other membrane lipids, in modulating the risk of psychosis. It suggests that membrane lipids could be used as biomarkers for personalised medicine in UHR patients.
Collapse
Affiliation(s)
- Ariel Frajerman
- Institute of Psychiatry and Neuroscience of Paris (IPNP), Université de Paris, INSERM U1266, F-75014 Paris, France
| | - Boris Chaumette
- Institute of Psychiatry and Neuroscience of Paris (IPNP), Université de Paris, INSERM U1266, F-75014 Paris, France
- GHU Paris Psychiatrie et Neurosciences, F-75674 Paris, France
- Department of Psychiatry, McGill University, Montréal, QC H3A 0G4, Canada
| | - Dominique Farabos
- INSERM UMR S 938, Département METOMICS, Centre de Recherche Saint-Antoine, Sorbonne Université, AP-HP, F-75012 Paris, France
| | - Gaétan Despres
- INSERM UMR S 938, Département METOMICS, Centre de Recherche Saint-Antoine, Sorbonne Université, AP-HP, F-75012 Paris, France
| | - Christelle Simonard
- INSERM UMR S 938, Département METOMICS, Centre de Recherche Saint-Antoine, Sorbonne Université, AP-HP, F-75012 Paris, France
| | - Antonin Lamazière
- INSERM UMR S 938, Département METOMICS, Centre de Recherche Saint-Antoine, Sorbonne Université, AP-HP, F-75012 Paris, France
| | - Marie-Odile Krebs
- Institute of Psychiatry and Neuroscience of Paris (IPNP), Université de Paris, INSERM U1266, F-75014 Paris, France
- GHU Paris Psychiatrie et Neurosciences, F-75674 Paris, France
| | - Oussama Kebir
- Institute of Psychiatry and Neuroscience of Paris (IPNP), Université de Paris, INSERM U1266, F-75014 Paris, France
- GHU Paris Psychiatrie et Neurosciences, F-75674 Paris, France
| |
Collapse
|
37
|
Alavi MS, Karimi G, Ghanimi HA, Roohbakhsh A. The potential of CYP46A1 as a novel therapeutic target for neurological disorders: An updated review of mechanisms. Eur J Pharmacol 2023; 949:175726. [PMID: 37062503 DOI: 10.1016/j.ejphar.2023.175726] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 04/03/2023] [Accepted: 04/14/2023] [Indexed: 04/18/2023]
Abstract
Cholesterol is a key component of the cell membrane that impacts the permeability, fluidity, and functions of membrane-bound proteins. It also participates in synaptogenesis, synaptic function, axonal growth, dendrite outgrowth, and microtubule stability. Cholesterol biosynthesis and metabolism are in balance in the brain. Its metabolism in the brain is mediated mainly by CYP46A1 or cholesterol 24-hydroxylase. It is responsible for eliminating about 80% of the cholesterol excess from the human brain. CYP46A1 converts cholesterol to 24S-hydroxycholesterol (24HC) that readily crosses the blood-brain barrier and reaches the liver for the final elimination process. Studies show that cholesterol and 24HC levels change during neurological diseases and conditions. So, it was hypothesized that inhibition or activation of CYP46A1 would be an effective therapeutic strategy. Accordingly, preclinical studies, using genetic and pharmacological interventions, assessed the role of CYP46A1 in main neurodegenerative disorders such as Parkinson's disease, Huntington's disease, Alzheimer's disease, multiple sclerosis, spinocerebellar ataxias, and amyotrophic lateral sclerosis. In addition, its role in seizures and brain injury was evaluated. The recent development of soticlestat, as a selective and potent CYP46A1 inhibitor, with significant anti-seizure effects in preclinical and clinical studies, suggests the importance of this target for future drug developments. Previous studies have shown that both activation and inhibition of CYP46A1 are of therapeutic value. This article, using recent studies, highlights the role of CYP46A1 in various brain diseases and insults.
Collapse
Affiliation(s)
- Mohaddeseh Sadat Alavi
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gholamreza Karimi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Ali Roohbakhsh
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
38
|
Wu W, Ren T, Cao X, Gao J. Hepatic transcriptome analysis reveals that elovl5 deletion promotes PUFA biosynthesis and deposition. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2023; 46:101076. [PMID: 37080058 DOI: 10.1016/j.cbd.2023.101076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 04/02/2023] [Accepted: 04/02/2023] [Indexed: 04/22/2023]
Abstract
The safe and low-cost acquisition of polyunsaturated fatty acids (PUFAs) has become a research hotspot. Fatty acyl elongase 5 (Elovl5), a rate-limiting enzyme for fatty acid elongation, is principally in charge of extending C18 and C20 PUFA substrates. However, the role of elovl5 in regulating pathways and genes involved in PUFA synthesis remain largely unknown. Here, hepatic transcriptome analysis of wild-type and elovl5 knockout (elovl5-/-) zebrafish was performed to identify the potential regulatory targets related to PUFA deposition and synthesis. There were 1579 differentially expressed genes (DEGs), of which 787 had their expression levels increased while 792 had the opposite effect. Peroxisome proliferators-activated receptors (PPAR) signaling pathway was considerably enriched in DEGs, according to the KEGG analysis, in which fatp2, fabp7, and pparδ were engaged in PUFA absorption and deposition. Additionally, transcriptome analysis also revealed that cyp46a1 and cyp2r1 were implicated in the synthesis of bile acids and the metabolism of vitamin D, thus indirectly participating in PUFA biosynthesis and deposition. Finally, the DEGs, which improve PUFA level following elovl5 deletion, were verified through feeding experiment with two prepared diets soybean oil diet and linolenic acid oil diet. This study revealed potential regulatory targets that improve PUFA level after elovl5 deletion in teleosts.
Collapse
Affiliation(s)
- Wenpeng Wu
- College of Fisheries, Engineering Research Center of Green development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | - Tianying Ren
- College of Fisheries, Engineering Research Center of Green development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiaojuan Cao
- College of Fisheries, Engineering Research Center of Green development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China; College of Fisheries, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Huazhong Agricultural University, Wuhan 430070, China
| | - Jian Gao
- College of Fisheries, Engineering Research Center of Green development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China; College of Fisheries, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
39
|
Abstract
Huntington's disease (HD) is a fatal, monogenic, autosomal dominant neurodegenerative disease caused by a polyglutamine-encoding CAG expansion in the huntingtin (HTT) gene that results in mutant huntingtin proteins (mHTT) in cells throughout the body. Although large parts of the central nervous system (CNS) are affected, the striatum is especially vulnerable and undergoes marked atrophy. Astrocytes are abundant within the striatum and contain mHTT in HD, as well as in mouse models of the disease. We focus on striatal astrocytes and summarize how they participate in, and contribute to, molecular pathophysiology and disease-related phenotypes in HD model mice. Where possible, reference is made to pertinent astrocyte alterations in human HD. Astrocytic dysfunctions related to cellular morphology, extracellular ion and neurotransmitter homeostasis, and metabolic support all accompany the development and progression of HD, in both transgenic mouse and human cellular and chimeric models of HD. These findings reveal the potential for the therapeutic targeting of astrocytes so as to restore synaptic as well as tissue homeostasis in HD. Elucidation of the mechanisms by which astrocytes contribute to HD pathogenesis may inform a broader understanding of the role of glial pathology in neurodegenerative disorders and, by so doing, enable new strategies of glial-directed therapeutics.
Collapse
Affiliation(s)
- Baljit S. Khakh
- Department of Physiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
- Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Steven A. Goldman
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York, USA
- Center for Translational Neuromedicine, University of Copenhagen Faculty of Health and Medical Sciences, Copenhagen, Denmark
| |
Collapse
|
40
|
Cataldi GG, Elorza SD, Toledano-Zaragoza A, de Olmos S, Cragnolini AB, Martín MG. Cholesterol-24-hydroxylase (CYP46) in the old brain: Analysis of positive populations and factors triggering its expression in astrocytes. J Comp Neurol 2023; 531:486-499. [PMID: 36453552 DOI: 10.1002/cne.25436] [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: 02/18/2022] [Revised: 11/01/2022] [Accepted: 11/04/2022] [Indexed: 12/05/2022]
Abstract
Cholesterol-24-hydroxylase (CYP46), a member of the cytochrome P450 superfamily of enzymes, is selectively expressed in the brain and is mainly responsible for cholesterol turnover in the central nervous system. Although increased cyp46A1 gene expression has been linked to cognitive alterations in aging and observed in neurodegenerative diseases and after traumatic brain injury, a detailed characterization of the brain regions and cell types in which CYP46 is expressed in old individuals has not been performed. Using immunohistochemistry and immunofluorescence, we investigated the specific regions and cell populations in the brain, in which cyp46A1 is expressed in 24-month-old mice. We found that CYP46 is localized in the same neuronal populations in young and old brains, mainly in the hippocampus, in cortical layers, and in Purkinje neurons of the cerebellum. No increase in CYP46 levels was found in astrocytes in old mice brains, in primary astrocyte-neuron cocultures aged in vitro, or in primary cultures of senescent astrocytes. However, interleukin-6 treatment strongly induced cyp46A1 expression in reactive astrocytes characterized by high GFAP levels but had no effect in nonactivated astrocytes. Our data suggest that cholesterol-24-hydroxylase expression is triggered in reactive astrocytes in response to proinflammatory signals, probably as part of a response mechanism to injury.
Collapse
Affiliation(s)
- Gabriel G Cataldi
- Instituto Ferreyra, INIMEC-CONICET-UNC, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Setiembre D Elorza
- Instituto Ferreyra, INIMEC-CONICET-UNC, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Ana Toledano-Zaragoza
- Molecular Neuropathology Unit, Physiological and Pathological Processes Program, Centro de Biología Molecular Severo Ochoa (CBMSO-CSIC), Madrid, Spain
| | - Soledad de Olmos
- Instituto Ferreyra, INIMEC-CONICET-UNC, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Andrea B Cragnolini
- Instituto de Investigaciones Biológicas y Tecnológicas, CONICET-UNC, Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Mauricio G Martín
- Instituto Ferreyra, INIMEC-CONICET-UNC, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
41
|
Estevez-Fraga C, Tabrizi SJ, Wild EJ. Huntington's Disease Clinical Trials Corner: August 2023. J Huntingtons Dis 2023; 12:169-185. [PMID: 37483021 PMCID: PMC10473124 DOI: 10.3233/jhd-239001] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/04/2023] [Indexed: 07/25/2023]
Abstract
In this edition of the Huntington's Disease Clinical Trials Corner, we expand on the GENERATION HD2 (tominersen) and on the Asklepios Biopharmaceutical/BrainVectis trial with AB-1001. We also comment on the recent findings from the PROOF-HD trial, and list all currently registered and ongoing clinical trials in Huntington's disease.
Collapse
Affiliation(s)
- Carlos Estevez-Fraga
- Huntington’s Disease Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Sarah J. Tabrizi
- Huntington’s Disease Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Edward J. Wild
- Huntington’s Disease Centre, UCL Queen Square Institute of Neurology, London, UK
| |
Collapse
|
42
|
Bogie JF, Guns J, Vanherle S. Lipid metabolism in neurodegenerative diseases. CELLULAR LIPID IN HEALTH AND DISEASE 2023:389-419. [DOI: 10.1016/b978-0-323-95582-9.00008-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
43
|
Bile acids and neurological disease. Pharmacol Ther 2022; 240:108311. [PMID: 36400238 DOI: 10.1016/j.pharmthera.2022.108311] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/29/2022] [Accepted: 11/14/2022] [Indexed: 11/17/2022]
Abstract
This review will focus on how bile acids are being used in clinical trials to treat neurological diseases due to their central involvement with the gut-liver-brain axis and their physiological and pathophysiological roles in both normal brain function and multiple neurological diseases. The synthesis of primary and secondary bile acids species and how the regulation of the bile acid pool may differ between the gut and brain is discussed. The expression of several bile acid receptors in brain and their currently known functions along with the tools available to manipulate them pharmacologically are examined, together with discussion of the interaction of bile acids with the gut microbiome and their lesser-known effects upon brain glucose and lipid metabolism. How dysregulation of the gut microbiome, aging and sex differences may lead to disruption of bile acid signalling and possible causal roles in a number of neurological disorders are also considered. Finally, we discuss how pharmacological treatments targeting bile acid receptors are currently being tested in an array of clinical trials for several different neurodegenerative diseases.
Collapse
|
44
|
Du J, Chen X, Zhao Y, Zhao T, Wang D, Chen Z, Wang C, Meng Q, Yao J, Sun H, Liu K, Wu J. Characterization of three naturally occurring lignans, sesamol, sesamolin, and sesamin, as potent inhibitors of human cytochrome P450 46A1: Implications for treating excitatory neurotoxicity. Front Pharmacol 2022; 13:1046814. [PMID: 36483743 PMCID: PMC9722955 DOI: 10.3389/fphar.2022.1046814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 10/31/2022] [Indexed: 08/22/2023] Open
Abstract
CYP46A1 is a brain-specific enzyme responsible for cholesterol homeostasis. Inhibition of CYP46A1 activity serves as a therapeutic target for excitatory neurotoxicity. Sesame is a common medicine and food resource; its component lignans possess various pharmacological activities. In this study, the inhibitory effects of sesame lignans on CYP46A1 activity were investigated. Inhibition kinetics analyses revealed that sesamin and sesamolin produce mixed partial competitive inhibition of CYP46A1, while sesamol produces non-competitive inhibition. Notably, molecular simulations revealed that the sesame lignans have excellent orientations within the active cavity of CYP46A1. Importantly, the sesame lignans had high permeability coefficients and low efflux ratios. Furthermore, sesamin significantly reduced the levels of 24S-hydroxycholesterol in rat plasma and brain tissues, and down-regulated the protein expressions of CYP46A1, NMDAR2A, NMDAR2B, and HMGCR. Collectively, sesame lignans exhibit significant inhibitory effects on CYP46A1 activity, highlighting their potential therapeutic role in treating excitatory neurotoxicity.
Collapse
Affiliation(s)
- Jie Du
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Liaoning Dalian Medical University, Dalian, China
| | - Xiaodong Chen
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Liaoning Dalian Medical University, Dalian, China
| | - Yongshun Zhao
- Department of Neurosurgery, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Tingting Zhao
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Liaoning Dalian Medical University, Dalian, China
| | - Dalong Wang
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Liaoning Dalian Medical University, Dalian, China
| | - Zujia Chen
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Liaoning Dalian Medical University, Dalian, China
| | - Changyuan Wang
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Liaoning Dalian Medical University, Dalian, China
| | - Qiang Meng
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Liaoning Dalian Medical University, Dalian, China
| | - Jialin Yao
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Liaoning Dalian Medical University, Dalian, China
| | - Huijun Sun
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Liaoning Dalian Medical University, Dalian, China
| | - Kexin Liu
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Liaoning Dalian Medical University, Dalian, China
| | - Jingjing Wu
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Liaoning Dalian Medical University, Dalian, China
| |
Collapse
|
45
|
Identification of potential inhibitors of brain-specific CYP46A1 from phytoconstituents in Indian traditional medicinal plants. JOURNAL OF PROTEINS AND PROTEOMICS 2022; 13:227-245. [PMCID: PMC9667835 DOI: 10.1007/s42485-022-00098-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/27/2022] [Accepted: 09/29/2022] [Indexed: 11/17/2022]
|
46
|
Zhou K, Han J, Wang Y, Zhang Y, Zhu C. Routes of administration for adeno-associated viruses carrying gene therapies for brain diseases. Front Mol Neurosci 2022; 15:988914. [PMID: 36385771 PMCID: PMC9643316 DOI: 10.3389/fnmol.2022.988914] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 10/03/2022] [Indexed: 08/27/2023] Open
Abstract
Gene therapy is a powerful tool to treat various central nervous system (CNS) diseases ranging from monogenetic diseases to neurodegenerative disorders. Adeno-associated viruses (AAVs) have been widely used as the delivery vehicles for CNS gene therapies due to their safety, CNS tropism, and long-term therapeutic effect. However, several factors, including their ability to cross the blood-brain barrier, the efficiency of transduction, their immunotoxicity, loading capacity, the choice of serotype, and peripheral off-target effects should be carefully considered when designing an optimal AAV delivery strategy for a specific disease. In addition, distinct routes of administration may affect the efficiency and safety of AAV-delivered gene therapies. In this review, we summarize different administration routes of gene therapies delivered by AAVs to the brain in mice and rats. Updated knowledge regarding AAV-delivered gene therapies may facilitate the selection from various administration routes for specific disease models in future research.
Collapse
Affiliation(s)
- Kai Zhou
- Henan Neurodevelopment Engineering Research Center for Children, Zhengzhou Key Laboratory of Pediatric Neurobehavior, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Jinming Han
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yafeng Wang
- Henan Neurodevelopment Engineering Research Center for Children, Zhengzhou Key Laboratory of Pediatric Neurobehavior, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou, China
- Department of Hematology and Oncology, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Yaodong Zhang
- Henan Neurodevelopment Engineering Research Center for Children, Zhengzhou Key Laboratory of Pediatric Neurobehavior, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, The Third Affiliated Hospital and Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
- Centre for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
47
|
Haider A, Zhao C, Wang L, Xiao Z, Rong J, Xia X, Chen Z, Pfister SK, Mast N, Yutuc E, Chen J, Li Y, Shao T, Warnock GI, Dawoud A, Connors TR, Oakley DH, Wei H, Wang J, Zheng Z, Xu H, Davenport AT, Daunais JB, Van RS, Shao Y, Wang Y, Zhang MR, Gebhard C, Pikuleva I, Levey AI, Griffiths WJ, Liang SH. Assessment of cholesterol homeostasis in the living human brain. Sci Transl Med 2022; 14:eadc9967. [PMID: 36197966 PMCID: PMC9581941 DOI: 10.1126/scitranslmed.adc9967] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Alterations in brain cholesterol homeostasis have been broadly implicated in neurological disorders. Notwithstanding the complexity by which cholesterol biology is governed in the mammalian brain, excess neuronal cholesterol is primarily eliminated by metabolic clearance via cytochrome P450 46A1 (CYP46A1). No methods are currently available for visualizing cholesterol metabolism in the living human brain; therefore, a noninvasive technology that quantitatively measures the extent of brain cholesterol metabolism via CYP46A1 could broadly affect disease diagnosis and treatment options using targeted therapies. Here, we describe the development and testing of a CYP46A1-targeted positron emission tomography (PET) tracer, 18F-CHL-2205 (18F-Cholestify). Our data show that PET imaging readouts correlate with CYP46A1 protein expression and with the extent to which cholesterol is metabolized in the brain, as assessed by cross-species postmortem analyses of specimens from rodents, nonhuman primates, and humans. Proof of concept of in vivo efficacy is provided in the well-established 3xTg-AD murine model of Alzheimer's disease (AD), where we show that the probe is sensitive to differences in brain cholesterol metabolism between 3xTg-AD mice and control animals. Furthermore, our clinical observations point toward a considerably higher baseline brain cholesterol clearance via CYP46A1 in women, as compared to age-matched men. These findings illustrate the vast potential of assessing brain cholesterol metabolism using PET and establish PET as a sensitive tool for noninvasive assessment of brain cholesterol homeostasis in the clinic.
Collapse
Affiliation(s)
- Ahmed Haider
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
- Emory University, Department of Radiology and Imaging Sciences, 1364 Clifton Rd, Atlanta, GA 30322, USA
| | - Chunyu Zhao
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
- Emory University, Department of Radiology and Imaging Sciences, 1364 Clifton Rd, Atlanta, GA 30322, USA
| | - Lu Wang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Zhiwei Xiao
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
- Emory University, Department of Radiology and Imaging Sciences, 1364 Clifton Rd, Atlanta, GA 30322, USA
| | - Jian Rong
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
- Emory University, Department of Radiology and Imaging Sciences, 1364 Clifton Rd, Atlanta, GA 30322, USA
| | - Xiaotian Xia
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, China
| | - Zhen Chen
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
| | - Stefanie K. Pfister
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
| | - Natalia Mast
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Eylan Yutuc
- Institute of Life Science, Swansea University Medical School, SA2 8PP Swansea, Wales, United Kingdom
| | - Jiahui Chen
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
- Emory University, Department of Radiology and Imaging Sciences, 1364 Clifton Rd, Atlanta, GA 30322, USA
| | - Yinlong Li
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
- Emory University, Department of Radiology and Imaging Sciences, 1364 Clifton Rd, Atlanta, GA 30322, USA
| | - Tuo Shao
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
| | - Geoffrey I. Warnock
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Alyaa Dawoud
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, 11835, Cairo, Egypt
| | - Theresa R. Connors
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Massachusetts Alzheimer’s Disease Research Center, Boston, MA 02129, USA
| | - Derek H. Oakley
- Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114-2696, USA
- C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital, Boston, MA 02114, USA
- Massachusetts Alzheimer’s Disease Research Center, Charlestown, MA 02129, USA
| | - Huiyi Wei
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Jinghao Wang
- Department of Pharmacy, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Zhihua Zheng
- Guangdong Province Pharmaceutical Association, Guangzhou 510080, China
| | - Hao Xu
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - April T. Davenport
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston Salem, NC, 27157, USA
| | - James B. Daunais
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston Salem, NC, 27157, USA
| | - Richard S. Van
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
| | - Yihan Shao
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
| | - Yuqin Wang
- Institute of Life Science, Swansea University Medical School, SA2 8PP Swansea, Wales, United Kingdom
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Catherine Gebhard
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Irina Pikuleva
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Allan I. Levey
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - William J. Griffiths
- Institute of Life Science, Swansea University Medical School, SA2 8PP Swansea, Wales, United Kingdom
| | - Steven H. Liang
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
- Emory University, Department of Radiology and Imaging Sciences, 1364 Clifton Rd, Atlanta, GA 30322, USA
| |
Collapse
|
48
|
Surdyka M, Jesion E, Niewiadomska-Cimicka A, Trottier Y, Kalinowska-Pośka Ż, Figiel M. Selective transduction of cerebellar Purkinje and granule neurons using delivery of AAV-PHP.eB and AAVrh10 vectors at axonal terminal locations. Front Mol Neurosci 2022; 15:947490. [PMID: 36176957 PMCID: PMC9513253 DOI: 10.3389/fnmol.2022.947490] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 07/25/2022] [Indexed: 11/25/2022] Open
Abstract
Adeno-associated virus (AAV)-based brain gene therapies require precision without off-targeting of unaffected neurons to avoid side effects. The cerebellum and its cell populations, including granule and Purkinje cells, are vulnerable to neurodegeneration; hence, conditions to deliver the therapy to specific cell populations selectively remain challenging. We have investigated a system consisting of the AAV serotypes, targeted injections, and transduction modes (direct or retrograde) for targeted delivery of AAV to cerebellar cell populations. We selected the AAV-PHP.eB and AAVrh10 serotypes valued for their retrograde features, and we thoroughly examined their cerebellar transduction pattern when injected into lobules and deep cerebellar nuclei. We found that AAVrh10 is suitable for the transduction of neurons in the mode highly dependent on placing the virus at axonal terminals. The strategy secures selective transduction for granule cells. The AAV-PHP.eB can transduce Purkinje cells and is very selective for the cell type when injected into the DCN at axonal PC terminals. Therefore, both serotypes can be used in a retrograde mode for selective transduction of major neuronal types in the cerebellum. Moreover, our in vivo transduction strategies are suitable for pre-clinical protocol development for gene delivery to granule cells by AAVrh10 and Purkinje cells by AAV-PHPeB.
Collapse
Affiliation(s)
- Magdalena Surdyka
- Department of Molecular Neurobiology, Institute of Bioorganic Chemistry Polish Academy of Sciences, Poznań, Poland
| | - Ewelina Jesion
- Department of Molecular Neurobiology, Institute of Bioorganic Chemistry Polish Academy of Sciences, Poznań, Poland
| | - Anna Niewiadomska-Cimicka
- Institute of Genetics and Molecular and Cellular Biology, INSERM U1258, CNRS UMR7104, University of Strasbourg, Illkirch, France
| | - Yvon Trottier
- Institute of Genetics and Molecular and Cellular Biology, INSERM U1258, CNRS UMR7104, University of Strasbourg, Illkirch, France
| | - Żaneta Kalinowska-Pośka
- Department of Molecular Neurobiology, Institute of Bioorganic Chemistry Polish Academy of Sciences, Poznań, Poland
| | - Maciej Figiel
- Department of Molecular Neurobiology, Institute of Bioorganic Chemistry Polish Academy of Sciences, Poznań, Poland
- *Correspondence: Maciej Figiel
| |
Collapse
|
49
|
Cholesterol and its reciprocal association with prion infection. Cell Tissue Res 2022; 392:235-246. [PMID: 35821439 DOI: 10.1007/s00441-022-03669-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 07/04/2022] [Indexed: 11/02/2022]
Abstract
Prion diseases are incurable, infectious and fatal neurodegenerative diseases that affect both humans and animals. The pathogenesis of prion disease involves the misfolding of the cellular prion protein, PrPC, to a disease-causing conformation, PrPSc, in the brain. The exact mechanism of conversion of PrPC to PrPSc is not clear; however, there are numerous studies supporting that this process of misfolding requires the association of PrPC with lipid raft domains of the plasma membrane. An increase in the cellular cholesterol content with prion infection has been observed in both in vivo and in vitro studies. As cholesterol is critical for the formation of lipid rafts, on the one hand, this increase may be related to, or aiding in, the process of prion conversion. On the other hand, increased cholesterol levels may affect neuronal viability. Here, we discuss current literature on the underlying mechanisms and potential consequences of elevated neuronal cholesterol in prion infection and advancements in prion disease therapeutics targeting brain cholesterol homeostasis.
Collapse
|
50
|
Mast N, Li Y, Pikuleva IA. Increased Acetylcholine Levels and Other Brain Effects in 5XFAD Mice after Treatment with 8,14-Dihydroxy Metabolite of Efavirenz. Int J Mol Sci 2022; 23:ijms23147669. [PMID: 35887013 PMCID: PMC9317559 DOI: 10.3390/ijms23147669] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/06/2022] [Accepted: 07/09/2022] [Indexed: 02/08/2023] Open
Abstract
Efavirenz (EFV), an FDA-approved anti-HIV drug, has off-target binding to CYP46A1, the CNS enzyme which converts cholesterol to 24-hydroxycholesterol. At small doses, EFV allosterically activates CYP46A1 in mice and humans and mitigates some of the Alzheimer's disease manifestations in 5XFAD mice, an animal model. Notably, in vitro, all phase 1 EFV hydroxymetabolites activate CYP46A1 as well and bind either to the allosteric site for EFV, neurotransmitters or both. Herein, we treated 5XFAD mice with 8,14-dihydroxyEFV, the binder to the neurotransmitter allosteric site, which elicits the highest CYP46A1 activation in vitro. We found that treated animals of both sexes had activation of CYP46A1 and cholesterol turnover in the brain, decreased content of the amyloid beta 42 peptide, increased levels of acetyl-CoA and acetylcholine, and altered expression of the brain marker proteins. In addition, male mice had improved performance in the Barnes Maze test and increased expression of the acetylcholine-related genes. This work expands our knowledge of the beneficial CYP46A1 activation effects and demonstrates that 8,14-dihydroxyEFV crosses the blood-brain barrier and has therapeutic potential as a CYP46A1 activator.
Collapse
|