1
|
Wang NQ, Sun PX, Shen QQ, Deng MY. Cholesterol Metabolism in CNS Diseases: The Potential of SREBP2 and LXR as Therapeutic Targets. Mol Neurobiol 2025; 62:6283-6307. [PMID: 39775479 DOI: 10.1007/s12035-024-04672-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025]
Abstract
The brain is the organ with the highest cholesterol content in the body. Cholesterol in the brain plays a crucial role in maintaining the integrity of synapses and myelin sheaths to ensure normal brain function. Disruptions in cholesterol metabolism are closely associated with various central nervous system (CNS) diseases, including Alzheimer's disease (AD), Huntington's disease (HD), and multiple sclerosis (MS). In this review, we explore the synthesis, regulation, transport, and functional roles of cholesterol in the CNS. We discuss in detail the associations between cholesterol homeostasis imbalance and CNS diseases including AD, HD, and MS, highlighting the significant role of cholesterol metabolism abnormalities in the development of these diseases. Sterol regulatory element binding protein-2 (SREBP2) and liver X receptor (LXR) are two critical transcription factors that play central roles in cholesterol synthesis and reverse transport, respectively. Their cooperative interaction finely tunes the balance of brain cholesterol metabolism, presenting potential therapeutic value for preventing and treating CNS diseases. We particularly emphasize the alterations in SREBP2 and LXR under pathological conditions and their impacts on disease progression. This review summarizes current therapeutic agents targeting these two pathways, with the hope of broadening the perspectives of CNS drug developers and encouraging further study into SREBP2 and LXR-related therapies for CNS diseases.
Collapse
Affiliation(s)
- Ning-Qi Wang
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Institute of Clinical Medicine, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, 450001, China
| | - Pei-Xiang Sun
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Institute of Clinical Medicine, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, 450001, China
| | - Qi-Qi Shen
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Institute of Clinical Medicine, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, 450001, China
| | - Meng-Yan Deng
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
2
|
Di Sarno A, Romano F, Arianna R, Serpico D, Lavorgna M, Savastano S, Colao A, Di Somma C. Lipid Metabolism and Statin Therapy in Neurodegenerative Diseases: An Endocrine View. Metabolites 2025; 15:282. [PMID: 40278411 DOI: 10.3390/metabo15040282] [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: 01/31/2025] [Revised: 03/25/2025] [Accepted: 04/02/2025] [Indexed: 04/26/2025] Open
Abstract
Background/aim: A growing body of evidence suggests a link between dyslipidemias and neurodegenerative diseases, highlighting the crucial role of lipid metabolism in the health of the central nervous system. The aim of our work was to provide an update on this topic, with a focus on clinical practice from an endocrinological point of view. Endocrinologists, being experts in the management of dyslipidemias, can play a key role in the prevention and treatment of neurodegenerative conditions, through precocious and effective lipid profile optimization. Methods: The literature was scanned to identify clinical trials and correlation studies on the association between dyslipidemia, statin therapy, and the following neurodegenerative diseases: Alzheimer's disease (AD), Parkisons's disease (PD), Multiple sclerosis (MS), and Amyotrophic lateral sclerosis (ALS). Results: Impaired lipid homeostasis, such as that frequently observed in patients affected by obesity and diabetes, is related to neurodegenerative diseases, such as AD, PD, and other cognitive deficits related to aging. AD and related dementias are now a real priority health problem. In the United States, there are approximately 7 million subjects aged 65 and older living with AD and related dementias, and this number is projected to grow to 12 million in the coming decades. Lipid-lowering therapy with statins is an effective strategy in reducing serum low-density lipoprotein cholesterol to normal range concentrations and, therefore, cardiovascular disease risk; moreover, statins have been reported to have a positive effect on neurodegenerative diseases. Conclusions: Several pieces of research have found inconsistent information following our review. There was no association between statin use and ALS incidence. More positive evidence has emerged regarding statin use and AD/PD. However, further large-scale prospective randomized control trials are required to properly understand this issue.
Collapse
Affiliation(s)
- Antonella Di Sarno
- Section of Endocrinology, Endocrinology Diabetology and Andrology Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Via Sergio Pansini 5, 80138 Naples, Italy
| | - Fiammetta Romano
- Section of Endocrinology, Endocrinology Diabetology and Andrology Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Via Sergio Pansini 5, 80138 Naples, Italy
| | - Rossana Arianna
- Section of Endocrinology, Endocrinology Diabetology and Andrology Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Via Sergio Pansini 5, 80138 Naples, Italy
| | - Domenico Serpico
- Section of Endocrinology, Endocrinology Diabetology and Andrology Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Via Sergio Pansini 5, 80138 Naples, Italy
| | - Mariarosaria Lavorgna
- Section of Endocrinology, Endocrinology Diabetology and Andrology Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Via Sergio Pansini 5, 80138 Naples, Italy
| | - Silvia Savastano
- Section of Endocrinology, Endocrinology Diabetology and Andrology Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Via Sergio Pansini 5, 80138 Naples, Italy
| | - Annamaria Colao
- Section of Endocrinology, Endocrinology Diabetology and Andrology Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Via Sergio Pansini 5, 80138 Naples, Italy
- UNESCO Chair "Education for Health and Sustainable Development", University of Naples Federico II, 80138 Naples, Italy
| | - Carolina Di Somma
- Section of Endocrinology, Endocrinology Diabetology and Andrology Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Via Sergio Pansini 5, 80138 Naples, Italy
- UNESCO Chair "Education for Health and Sustainable Development", University of Naples Federico II, 80138 Naples, Italy
| |
Collapse
|
3
|
Azizi L, Hausman H, Meyer AK, Wong M, Pajonk F. The Mevalonate Pathway in the Radiation Response of Cancer. Int J Radiat Oncol Biol Phys 2025:S0360-3016(25)00278-0. [PMID: 40194746 DOI: 10.1016/j.ijrobp.2025.03.059] [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: 10/23/2024] [Revised: 02/14/2025] [Accepted: 03/19/2025] [Indexed: 04/09/2025]
Abstract
The mevalonate (MVA) pathway plays a critical role in cholesterol biosynthesis, protein prenylation, and metabolic reprogramming, all of which contribute to cancer progression and therapy resistance. Targeting the MVA pathway with statins and other inhibitors has shown promise in preclinical studies; however, clinical outcomes remain controversial, raising concerns about translating these findings into effective treatments. Additionally, the interaction between the MVA pathway and radiation therapy (RT) is not yet fully understood, as RT upregulates the pathway, which can enhance tumor cell survival. This review summarizes the current literature on MVA pathway inhibition in cancer therapy, focusing on its potential to enhance the efficacy of RT. A better understanding of the pathway's role in radiation responses will be essential to translate combination therapies that target this pathway.
Collapse
Affiliation(s)
- Linda Azizi
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA.
| | - Hannah Hausman
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA
| | - Alexandra K Meyer
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA
| | - Matthew Wong
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA
| | - Frank Pajonk
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA; Department of Neurosurgery, David Geffen School of Medicine at UCLA; Jonsson Comprehensive Cancer Center at UCLA
| |
Collapse
|
4
|
Vanherle S, Loix M, Miron VE, Hendriks JJA, Bogie JFJ. Lipid metabolism, remodelling and intercellular transfer in the CNS. Nat Rev Neurosci 2025; 26:214-231. [PMID: 39972160 DOI: 10.1038/s41583-025-00908-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2025] [Indexed: 02/21/2025]
Abstract
Lipid metabolism encompasses the catabolism and anabolism of lipids, and is fundamental for the maintenance of cellular homeostasis, particularly within the lipid-rich CNS. Increasing evidence further underscores the importance of lipid remodelling and transfer within and between glial cells and neurons as key orchestrators of CNS lipid homeostasis. In this Review, we summarize and discuss the complex landscape of processes involved in lipid metabolism, remodelling and intercellular transfer in the CNS. Highlighted are key pathways, including those mediating lipid (and lipid droplet) biogenesis and breakdown, lipid oxidation and phospholipid metabolism, as well as cell-cell lipid transfer mediated via lipoproteins, extracellular vesicles and tunnelling nanotubes. We further explore how the dysregulation of these pathways contributes to the onset and progression of neurodegenerative diseases, and examine the homeostatic and pathogenic impacts of environment, diet and lifestyle on CNS lipid metabolism.
Collapse
Affiliation(s)
- Sam Vanherle
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Centre, Hasselt University, Hasselt, Belgium
| | - Melanie Loix
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Centre, Hasselt University, Hasselt, Belgium
| | - Veronique E Miron
- Keenan Research Centre for Biomedical Science and Barlo Multiple Sclerosis Centre, St Michael's Hospital, Toronto, Ontario, Canada
- Department of Immunology, The University of Toronto, Toronto, Ontario, Canada
- UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK
| | - Jerome J A Hendriks
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Centre, Hasselt University, Hasselt, Belgium
| | - Jeroen F J Bogie
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium.
- University MS Centre, Hasselt University, Hasselt, Belgium.
| |
Collapse
|
5
|
Zhang L, Wei J, Liu X, Li D, Pang X, Chen F, Cao H, Lei P. Gut microbiota-astrocyte axis: new insights into age-related cognitive decline. Neural Regen Res 2025; 20:990-1008. [PMID: 38989933 PMCID: PMC11438350 DOI: 10.4103/nrr.nrr-d-23-01776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 03/04/2024] [Indexed: 07/12/2024] Open
Abstract
With the rapidly aging human population, age-related cognitive decline and dementia are becoming increasingly prevalent worldwide. Aging is considered the main risk factor for cognitive decline and acts through alterations in the composition of the gut microbiota, microbial metabolites, and the functions of astrocytes. The microbiota-gut-brain axis has been the focus of multiple studies and is closely associated with cognitive function. This article provides a comprehensive review of the specific changes that occur in the composition of the gut microbiota and microbial metabolites in older individuals and discusses how the aging of astrocytes and reactive astrocytosis are closely related to age-related cognitive decline and neurodegenerative diseases. This article also summarizes the gut microbiota components that affect astrocyte function, mainly through the vagus nerve, immune responses, circadian rhythms, and microbial metabolites. Finally, this article summarizes the mechanism by which the gut microbiota-astrocyte axis plays a role in Alzheimer's and Parkinson's diseases. Our findings have revealed the critical role of the microbiota-astrocyte axis in age-related cognitive decline, aiding in a deeper understanding of potential gut microbiome-based adjuvant therapy strategies for this condition.
Collapse
Affiliation(s)
- Lan Zhang
- Haihe Laboratory of Cell Ecosystem, Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Jingge Wei
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Xilei Liu
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Dai Li
- Haihe Laboratory of Cell Ecosystem, Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaoqi Pang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Fanglian Chen
- Tianjin Neurological Institution, Tianjin Medical University General Hospital, Tianjin, China
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Ping Lei
- Haihe Laboratory of Cell Ecosystem, Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
6
|
Yoon H, Triplet EM, Wurtz L, Simon WL, Choi CI, Scarisbrick IA. Regulation of CNS Lipids by Protease Activated Receptor 1. J Neurochem 2025; 169:e70047. [PMID: 40123504 PMCID: PMC11968084 DOI: 10.1111/jnc.70047] [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: 08/16/2024] [Revised: 02/22/2025] [Accepted: 02/28/2025] [Indexed: 03/25/2025]
Abstract
Disruptions in the metabolism of cholesterol and other lipids are strongly implicated in the pathogenesis of neurological disease. The CNS is highly enriched in cholesterol, which is primarily synthesized de novo. Cholesterol synthesis is also rate limiting for myelin regeneration. Given that knockout of the thrombin receptor (Protease Activated Receptor 1 (PAR1)) accelerates myelin regeneration, here we sought to determine the potential regulatory actions of PAR1 in CNS cholesterol and lipid metabolism in the intact adult CNS and during myelin regeneration. We present quantitative PCR and RNAseq evidence from murine spinal cords at the peak of myelination and in adulthood showing PAR1 knockout is associated with increased gene expression for cholesterol biosynthesis (Hmgcs1, Hmgcr, Sqle, and Dhcr7), lipid transport (ApoE, Abca1, and Ldlr), and intracellular processing (Lcat, Npc1, and Npc2) at one or more time points examined. An upregulation of genes involved in the synthesis of other lipids enriched in the myelin membrane, specifically Fa2h, Ugt8a, and Gal3st1, was also observed in PAR1 knockouts. Transcription factors essential for lipid and cholesterol production (Srebf1 and Srebf2) were also increased in PAR1 knockout spinal cords at the postnatal day 21 peak of myelination and at day 45. GC-MS and LC-MS quantification of lipids demonstrated coordinate increases in the abundance of select cholesterol and lipid species in the spinal cords of PAR1 knockout mice, including enrichment of esterified cholesterol, together with sphingomyelins and sphingolipids. Co-localization of the SREBP1 and SREBP2 transcription factors, as well as HMGCS1, a rate-limiting enzyme in cholesterol biosynthesis, to glia during remyelination post-lysolecithin or cuprizone-mediated demyelination showed a prominent regulatory role for PAR1 in Olig2+ oligodendrocytes. PAR1 knockouts also demonstrated elevated levels of SREBP2 in more mature GST3+ oligodendrocytes and SREBP1 in GFAP+ astrocytes during remyelination post-lysolecithin. These findings demonstrate novel roles for PAR1 as a regulator of CNS cholesterol and lipid metabolism and its potential as a therapeutic target to increase cholesterol availability to improve myelin regeneration.
Collapse
Affiliation(s)
- Hyesook Yoon
- Department of Physical Medicine and Rehabilitation, Center for Regenerative Biotherapeutics, Rochester, MN 55905
| | - Erin M. Triplet
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic Alix School of Medicine, and the Mayo Clinic Medical Scientist Training Program Sciences Rochester, Rochester, MN 55905
| | - Lincoln Wurtz
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic Alix School of Medicine, and the Mayo Clinic Medical Scientist Training Program Sciences Rochester, Rochester, MN 55905
| | - Whitney L. Simon
- Department of Physical Medicine and Rehabilitation, Center for Regenerative Biotherapeutics, Rochester, MN 55905
| | - Chan-Il Choi
- Department of Physical Medicine and Rehabilitation, Center for Regenerative Biotherapeutics, Rochester, MN 55905
| | - Isobel A. Scarisbrick
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic Alix School of Medicine, and the Mayo Clinic Medical Scientist Training Program Sciences Rochester, Rochester, MN 55905
- Department of Physical Medicine and Rehabilitation, Center for Regenerative Biotherapeutics, Rochester, MN 55905
- Department of Physiology and Biomedical Engineering, Rochester, MN 55905
| |
Collapse
|
7
|
Puljko B, Štracak M, Kalanj-Bognar S, Todorić Laidlaw I, Mlinac-Jerkovic K. Gangliosides and Cholesterol: Dual Regulators of Neuronal Membrane Framework in Autism Spectrum Disorder. Int J Mol Sci 2025; 26:1322. [PMID: 39941090 PMCID: PMC11818915 DOI: 10.3390/ijms26031322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 01/30/2025] [Accepted: 02/01/2025] [Indexed: 02/16/2025] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder with heterogeneous clinical presentation. Diagnosing ASD is complex, and the criteria for diagnosis, as well as the term ASD, have changed during the last decades. Diagnosis is made based on observation and accomplishment of specific diagnostic criteria, while a particular biomarker of ASD does not yet exist. However, studies universally report a disequilibrium in membrane lipid content, pointing to a unique neurolipid signature of ASD. This review sheds light on the possible role of cholesterol and gangliosides, complex membrane glycosphingolipids, in the development of ASD. In addition to maintaining membrane integrity, neuronal signaling, and synaptic plasticity, these lipids play a role in neurotransmitter release and calcium signaling. Evidence linking ASD to lipidome changes includes low cholesterol levels, unusual ganglioside levels, and unique metabolic profiles. ASD symptoms may be mitigated with therapeutic interventions targeting the lipid composition of membranes. However, restoring membrane equilibrium in the central nervous system remains a challenge. This review underscores the need for comprehensive research into lipid metabolism to uncover practical insights into ASD etiology and treatment as lipidomics emerges as a major area in ASD research.
Collapse
Affiliation(s)
- Borna Puljko
- Laboratory for Molecular Neurobiology and Neurochemistry, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (B.P.); (S.K.-B.)
- Department of Chemistry and Biochemistry, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | | | - Svjetlana Kalanj-Bognar
- Laboratory for Molecular Neurobiology and Neurochemistry, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (B.P.); (S.K.-B.)
- Department of Chemistry and Biochemistry, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Ivana Todorić Laidlaw
- Department for Forensic Psychiatry, University Psychiatric Hospital Vrapče, 10090 Zagreb, Croatia
| | - Kristina Mlinac-Jerkovic
- Laboratory for Molecular Neurobiology and Neurochemistry, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (B.P.); (S.K.-B.)
- Department of Chemistry and Biochemistry, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| |
Collapse
|
8
|
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
|
9
|
Ueno H, Takahashi Y, Mori S, Kitano E, Murakami S, Wani K, Miyazaki T, Matsumoto Y, Okamoto M, Ishihara T. Age-related behavioural abnormalities in C57BL/6.KOR- Apoe shl mice. Transl Neurosci 2025; 16:20220363. [PMID: 40026711 PMCID: PMC11868718 DOI: 10.1515/tnsci-2022-0363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 12/13/2024] [Accepted: 01/13/2025] [Indexed: 03/05/2025] Open
Abstract
Spontaneously hyperlipidaemic (Apoeshl) mice were discovered in 1999 as mice lacking apolipoprotein E (ApoE) owing to a mutation in the Apoe gene. However, age-related behavioural changes in commercially available Apoeshl mice have not yet been clarified. The behavioural abnormalities of ApoE-deficient mice, which are genetically modified mice artificially deficient in ApoE, have been investigated in detail, and it has been reported that they can serve as a model of Alzheimer's disease (AD). To understand whether Apoeshl mice can also serve as a murine model of AD, it is necessary to investigate age-related behavioural abnormalities in Apoeshl mice. In this study, we conducted a series of behavioural experiments on 7- and 11-month-old Apoeshl mice to investigate the behavioural abnormalities associated with ageing in Apoeshl mice. In this study, 7-month-old Apoeshl mice showed decreased body weight and grip strength compared to age-matched wild-type mice. In the open field test, 7-month-old Apoeshl mice showed increased anxiety-like behaviour compared to wild-type mice, whereas 11-month-old Apoeshl mice showed decreased anxiety-like behaviour. Moreover, Apoeshl mice aged 7 and 11 months had increased serum cholesterol levels. These results indicate that the behaviour of Apoeshl mice changes with age. However, 11-month-old Apoeshl mice did not show a decline in cognitive function or memory ability similar to murine models of AD. Our findings indicate that Apoeshl mice can be used to investigate the function of ApoE in the central nervous system.
Collapse
Affiliation(s)
- Hiroshi Ueno
- Department of Medical Technology, Kawasaki University of Medical Welfare,
Okayama, 701-0193, Japan
| | - Yu Takahashi
- Department of Psychiatry, Kawasaki Medical School,
Kurashiki, 701-0192, Japan
| | - Sachiko Mori
- Department of Psychiatry, Kawasaki Medical School,
Kurashiki, 701-0192, Japan
| | - Eriko Kitano
- Department of Psychiatry, Kawasaki Medical School,
Kurashiki, 701-0192, Japan
| | - Shinji Murakami
- Department of Psychiatry, Kawasaki Medical School,
Kurashiki, 701-0192, Japan
| | - Kenta Wani
- Department of Psychiatry, Kawasaki Medical School,
Kurashiki, 701-0192, Japan
| | - Tetsuji Miyazaki
- Department of Psychiatry, Kawasaki Medical School,
Kurashiki, 701-0192, Japan
| | - Yosuke Matsumoto
- Department of Neuropsychiatry, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University,
Okayama, 700-8558, Japan
| | - Motoi Okamoto
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University,
Okayama, 700-8558, Japan
| | - Takeshi Ishihara
- Department of Psychiatry, Kawasaki Medical School,
Kurashiki, 701-0192, Japan
| |
Collapse
|
10
|
Mirnics K. Fetal Fentanyl Syndrome - Only the "tip of the iceberg"? Eur Neuropsychopharmacol 2025; 90:69-70. [PMID: 39520878 DOI: 10.1016/j.euroneuro.2024.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 10/17/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Affiliation(s)
- Károly Mirnics
- Munroe-Meyer Institute for Genetics and Rehabilitation, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
11
|
Darmanto AG, Yen TL, Jan JS, Linh TTD, Taliyan R, Yang CH, Sheu JR. Beyond metabolic messengers: Bile acids and TGR5 as pharmacotherapeutic intervention for psychiatric disorders. Pharmacol Res 2025; 211:107564. [PMID: 39733841 DOI: 10.1016/j.phrs.2024.107564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/05/2024] [Accepted: 12/23/2024] [Indexed: 12/31/2024]
Abstract
Psychiatric disorders pose a significant global health challenge, exacerbated by the COVID-19 pandemic and insufficiently addressed by the current treatments. This review explores the emerging role of bile acids and the TGR5 receptor in the pathophysiology of psychiatric conditions, emphasizing their signaling within the gut-brain axis. We detail the synthesis and systemic functions of bile acids, their transformation by gut microbiota, and their impact across various neuropsychiatric disorders, including major depressive disorder, general anxiety disorder, schizophrenia, autism spectrum disorder, and bipolar disorder. The review highlights how dysbiosis and altered bile acid metabolism contribute to the development and exacerbation of these neuropsychiatric disorders through mechanisms involving inflammation, oxidative stress, and neurotransmitter dysregulation. Importantly, we detail both pharmacological and non-pharmacological interventions that modulate TGR5 signaling, offering potential breakthroughs in treatment strategies. These include dietary adjustments to enhance beneficial bile acids production and the use of specific TGR5 agonists that have shown promise in preclinical and clinical settings for their regulatory effects on critical pathways such as cAMP-PKA, NRF2-mediated antioxidant responses, and neuroinflammation. By integrating findings from the dynamics of gut microbiota, bile acids metabolism, and TGR5 receptor related signaling events, this review underscores cutting-edge therapeutic approaches poised to revolutionize the management and treatment of psychiatric disorders.
Collapse
Affiliation(s)
- Arief Gunawan Darmanto
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan, ROC; School of Medicine, Universitas Ciputra, Surabaya 60219, Indonesia
| | - Ting-Lin Yen
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan, ROC; Department of Medical Research, Cathay General Hospital, Taipei 22174, Taiwan, ROC
| | - Jing-Shiun Jan
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan, ROC
| | - Tran Thanh Duy Linh
- Family Medicine Training Center, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City 700000, Viet Nam
| | - Rajeev Taliyan
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Pilani Campus, Pilani, Rajasthan, India
| | - Chih-Hao Yang
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan, ROC; Research Center for Neuroscience, Taipei Medical University, Taipei, Taiwan, ROC.
| | - Joen-Rong Sheu
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan, ROC; Research Center for Neuroscience, Taipei Medical University, Taipei, Taiwan, ROC; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan, ROC.
| |
Collapse
|
12
|
Testa G, Giannelli S, Staurenghi E, Cecci R, Floro L, Gamba P, Sottero B, Leonarduzzi G. The Emerging Role of PCSK9 in the Pathogenesis of Alzheimer's Disease: A Possible Target for the Disease Treatment. Int J Mol Sci 2024; 25:13637. [PMID: 39769398 PMCID: PMC11727734 DOI: 10.3390/ijms252413637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/13/2024] [Accepted: 12/17/2024] [Indexed: 01/12/2025] Open
Abstract
Alzheimer's disease (AD) is a multifactorial neurodegenerative disease mainly caused by β-amyloid (Aβ) accumulation in the brain. Among the several factors that may concur to AD development, elevated cholesterol levels and brain cholesterol dyshomeostasis have been recognized to play a relevant role. Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a protein primarily known to regulate plasma low-density lipoproteins (LDLs) rich in cholesterol and to be one of the main causes of familial hypercholesterolemia. In addition to that, PCSK9 is also recognized to carry out diverse important activities in the brain, including control of neuronal differentiation, apoptosis, and, importantly, LDL receptors functionality. Moreover, PCSK9 appeared to be directly involved in some of the principal processes responsible for AD development, such as inflammation, oxidative stress, and Aβ deposition. On these bases, PCSK9 management might represent a promising approach for AD treatment. The purpose of this review is to elucidate the role of PCSK9, whether or not cholesterol-related, in AD pathogenesis and to give an updated overview of the most innovative therapeutic strategies developed so far to counteract the pleiotropic activities of both humoral and brain PCSK9, focusing in particular on their potentiality for AD management.
Collapse
Affiliation(s)
- Gabriella Testa
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (G.T.); (S.G.); (E.S.); (R.C.); (L.F.); (P.G.); (G.L.)
| | - Serena Giannelli
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (G.T.); (S.G.); (E.S.); (R.C.); (L.F.); (P.G.); (G.L.)
- Division of Neurology Vand Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Erica Staurenghi
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (G.T.); (S.G.); (E.S.); (R.C.); (L.F.); (P.G.); (G.L.)
| | - Rebecca Cecci
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (G.T.); (S.G.); (E.S.); (R.C.); (L.F.); (P.G.); (G.L.)
| | - Lucrezia Floro
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (G.T.); (S.G.); (E.S.); (R.C.); (L.F.); (P.G.); (G.L.)
| | - Paola Gamba
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (G.T.); (S.G.); (E.S.); (R.C.); (L.F.); (P.G.); (G.L.)
| | - Barbara Sottero
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (G.T.); (S.G.); (E.S.); (R.C.); (L.F.); (P.G.); (G.L.)
| | - Gabriella Leonarduzzi
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (G.T.); (S.G.); (E.S.); (R.C.); (L.F.); (P.G.); (G.L.)
| |
Collapse
|
13
|
Korade Z, Anderson AC, Sharma K, Tallman KA, Kim HYH, Porter NA, Gripp KW, Mirnics K. Inhibition of post-lanosterol biosynthesis by fentanyl: potential implications for Fetal Fentanyl Syndrome (FFS). Mol Psychiatry 2024; 29:3942-3949. [PMID: 38844533 DOI: 10.1038/s41380-024-02622-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/15/2024] [Accepted: 05/20/2024] [Indexed: 12/05/2024]
Abstract
A recent study discovered a novel, complex developmental disability syndrome, most likely caused by maternal fentanyl use disorder. This Fetal Fentanyl Syndrome (FFS) is biochemically characterized by elevated 7-dehydrocholesterol (7-DHC) levels in neonates, raising the question if fentanyl inhibition of the dehydrocholesterol reductase 7 (DHCR7) enzyme is causal for the emergence of the pathophysiology and phenotypic features of FFS. To test this hypothesis, we undertook a series of experiments on Neuro2a cells, primary mouse neuronal and astrocytic cultures, and human dermal fibroblasts (HDFs) with DHCR7+/+ and DHCR7+/- genotype. Our results revealed that in vitro exposure to fentanyl disrupted sterol biosynthesis across all four in vitro models. The sterol biosynthesis disruption by fentanyl was complex, and encompassed the majority of post-lanosterol intermediates, including elevated 7-DHC and decreased desmosterol (DES) levels across all investigated models. The overall findings suggested that maternal fentanyl use in the context of an opioid use disorder leads to FFS in the developing fetus through a strong disruption of the whole post-lanosterol pathway that is more complex than a simple DHCR7 inhibition. In follow-up experiments we found that heterozygous DHCR7+/- HDFs were significantly more susceptible to the sterol biosynthesis inhibitory effects of fentanyl than wild-type DHCR7+/+ fibroblasts. These data suggest that DHCR7+/- heterozygosity of mother and/or developing child (and potentially other sterol biosynthesis genes), when combined with maternal fentanyl use disorder, might be a significant contributory factor to the emergence of FFS in the exposed offspring. In a broader context, we believe that evaluation of new and existing medications for their effects on sterol biosynthesis should be an essential consideration during drug safety determinations, especially in pregnancy.
Collapse
Affiliation(s)
- Zeljka Korade
- Department of Pediatrics, Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Allison C Anderson
- Munroe-Meyer Institute for Genetics and Rehabilitation, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Kanika Sharma
- Mass Spectrometry Core, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Keri A Tallman
- Department of Chemistry, Vanderbilt Institute of Chemical Biology and Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN, 37240, USA
| | - Hye-Young H Kim
- Department of Chemistry, Vanderbilt Institute of Chemical Biology and Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN, 37240, USA
| | - Ned A Porter
- Department of Chemistry, Vanderbilt Institute of Chemical Biology and Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN, 37240, USA
| | - Karen W Gripp
- Division of Medical Genetics, Nemours Children's Hospital, Wilmington, DE, 19803, USA
| | - Karoly Mirnics
- Munroe-Meyer Institute for Genetics and Rehabilitation, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
14
|
He K, Zhao Z, Zhang J, Li D, Wang S, Liu Q. Cholesterol Metabolism in Neurodegenerative Diseases. Antioxid Redox Signal 2024; 41:1051-1072. [PMID: 38842175 DOI: 10.1089/ars.2024.0674] [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] [Indexed: 06/07/2024]
Abstract
Significance: Cholesterol plays a crucial role in the brain, where it is highly concentrated and tightly regulated to support normal brain functions. It serves as a vital component of cell membranes, ensuring their integrity, and acts as a key regulator of various brain processes. Dysregulation of cholesterol metabolism in the brain has been linked to impaired brain function and the onset of neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease, and Huntington's disease. Recent Advances: A significant advancement has been the identification of astrocyte-derived apoliprotein E as a key regulator of de novo cholesterol biosynthesis in neurons, providing insights into how extracellular signals influence neuronal cholesterol levels. In addition, the development of antibody-based therapies, particularly for AD, presents promising opportunities for therapeutic interventions. Critical Issues: Despite significant research, the association between cholesterol and neurodegenerative diseases remains inconclusive. It is crucial to distinguish between plasma cholesterol and brain cholesterol, as these pools are relatively independent. This differentiation should be considered when evaluating statin-based treatment approaches. Furthermore, assessing not only the total cholesterol content in the brain but also its distribution among different types of brain cells is essential. Future Direction: Establishing a causal link between changes in brain/plasma cholesterol levels and the onset of brain dysfunction/neurodegenerative diseases remains a key objective. In addition, conducting cell-specific analyses of cholesterol homeostasis in various types of brain cells under pathological conditions will enhance our understanding of cholesterol metabolism in neurodegenerative diseases. Manipulating cholesterol levels to restore homeostasis may represent a novel approach for alleviating neurological symptoms. Antioxid. Redox Signal. 41, 1051-1072.
Collapse
Affiliation(s)
- Keqiang He
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Zhiwei Zhao
- Department of Cardiovascular Surgery, the First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China
| | - Juan Zhang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China
- CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Dingfeng Li
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China
- CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Sheng Wang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Qiang Liu
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China
- CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
- Neurodegenerative Disorder Research Center, Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
- Key Laboratory of Immune Response and Immunotherapy, University of Science and Technology of China, Hefei, China
| |
Collapse
|
15
|
Semreen AM, Alsoud LO, Semreen MH, Ahmed M, Al-Hroub HM, El-Awady R, Ramadan WS, Abuhelwa A, Bustanji Y, Soares NC, Alzoubi KH. Multi-omics analysis revealed significant metabolic changes in brain cancer cells treated with paclitaxel and/or topotecan. Heliyon 2024; 10:e39420. [PMID: 39524752 PMCID: PMC11550653 DOI: 10.1016/j.heliyon.2024.e39420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 10/14/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
Glioblastoma (GBM) stands as the most common primary malignant brain tumor. Despite the best standard therapies, GBM survivors have a brief survival time, about 24 months on average. The treatment is troublesome because the cancer cells may not respond well to specific therapies as they grow within an extensive network of blood vessels. Our study aims to evaluate the impact of paclitaxel 5.3 μg/mL and topotecan 0.26 μM solely and in pairwise combination on the resultant metabolic and proteomic signatures of the U87 cell line while using the precise ultra-high-performance liquid chromatography quadrupole time-of-flight mass spectrometry (UHPLC-Q-TOF) analytical technology. The U87 cells wear treated with DMSO, paclitaxel 5.3 μM, topotecan 0.26 μM, and their combinations. Using One-way ANOVA, we observed 14 significantly altered metabolites compared to those cells treated with DMSO. For combination treatment (paclitaxel and topotecan), 11 metabolites were significantly dysregulated. Sparse partial least squares-discriminant analysis (sPLS-DA) revealed minimal overlap, highlighting distinctions among the four groups. While for proteomics, a total of 79 proteins were significantly dysregulated among the groups. These findings can aid in identifying new biomarkers associated with the utilized drugs and creating a map for targeted therapy. EIF3F, GNB2L1, HINT2, and RPA3 were shown to be significantly upregulated in the combination group relative to the control. Moreover, ribosome, apoptosis, HIF-1 signaling, arginine and proline, glutathione, purine metabolism, apelin signaling pathway, and glycolysis were significantly altered in the combination group. Overall, this study underscores the effectiveness of multi-omics approaches in revealing the molecular mechanisms driving chemotherapy responses in cancer cells. Additionally, this work generates a comprehensive list of molecular alterations that can serve as a foundation for further investigations and inform personalized healthcare strategies to enhance patient outcomes.
Collapse
Affiliation(s)
- Ahlam M. Semreen
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Leen Oyoun Alsoud
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Mohammad H. Semreen
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Munazza Ahmed
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Hamza M. Al-Hroub
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Raafat El-Awady
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Wafaa S. Ramadan
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Ahmad Abuhelwa
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Yasser Bustanji
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
- School of Pharmacy, The University of Jordan, Amman, 11942, Jordan
- Department of Basic and Clinical Pharmacology, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Nelson C. Soares
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
- Center for Applied and Translational Genomics (CATG), Mohamed Bin Rashid University of Medicine and Health Sciences (MBRU), Dubai Health, Dubai, United Arab Emirates
- College of Medicine, Mohamed Bin Rashid University of Medicine and Health Sciences (MBRU), Dubai Health, Dubai, United Arab Emirates
| | - Karem H. Alzoubi
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates
| |
Collapse
|
16
|
Watanabe Y, Obama T, Makiyama T, Itabe H. Oxysterols Suppress Release of DNA from Granulocytes into Extracellular Space After Stimulation with Phorbol Myristate Acetate. Biomedicines 2024; 12:2535. [PMID: 39595101 PMCID: PMC11592087 DOI: 10.3390/biomedicines12112535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/26/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Neutrophils eject their DNA strings and cellular proteins into the extracellular space upon treatment with various stimulants. In the present study, we examined the effects of four major oxidized cholesterol metabolites on DNA release from granulocytes. METHODS AND RESULTS When oxysterols were added to HL-60-derived granulocytes stimulated with phorbol 12-myristate 13-acetate (PMA), they suppressed the release of DNA and myeloperoxidase from the cells. Among the four oxysterols tested, 7-ketocholesterol was the most effective. Addition of the same concentration of 7-ketocholesterol did not induce any cytotoxic effects, as evaluated based on the release of lactate dehydrogenase and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazoliumbromide (MTT) assays. DNA release from human peripheral blood neutrophils after PMA stimulation was also suppressed by 7-ketocholesterol. Liquid chromatography with tandem mass spectrometry (LC-MS/MS) analysis was used to quantify sterol content in the cells. The addition of oxysterols increased the cellular content of the corresponding compounds by more than 10-fold compared to those at baseline. Treatment of HL-60-derived granulocytes with methyl-β-cyclodextrin that removes sterol compounds from the membranes increased DNA release from the cells in a dose-dependent manner. CONCLUSIONS These results suggest that oxysterols have suppressive effects on DNA release from granulocytes stimulated with PMA.
Collapse
Affiliation(s)
| | | | | | - Hiroyuki Itabe
- Division of Biological Chemistry, Department of Pharmaceutical Sciences, Showa University Graduate School of Pharmacy, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan; (Y.W.); (T.O.); (T.M.)
| |
Collapse
|
17
|
Langlois J, Lange S, Ebeling M, Macnair W, Schmucki R, Li C, DeGeer J, Sudharshan TJJ, Yong VW, Shen YA, Harp C, Collin L, Keaney J. Fenebrutinib, a Bruton's tyrosine kinase inhibitor, blocks distinct human microglial signaling pathways. J Neuroinflammation 2024; 21:276. [PMID: 39465429 PMCID: PMC11514909 DOI: 10.1186/s12974-024-03267-5] [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: 08/01/2024] [Accepted: 10/18/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND Bruton's tyrosine kinase (BTK) is an intracellular signaling enzyme that regulates B-lymphocyte and myeloid cell functions. Due to its involvement in both innate and adaptive immune compartments, BTK inhibitors have emerged as a therapeutic option in autoimmune disorders such as multiple sclerosis (MS). Brain-penetrant, small-molecule BTK inhibitors may also address compartmentalized neuroinflammation, which is proposed to underlie MS disease progression. BTK is expressed by microglia, which are the resident innate immune cells of the brain; however, the precise roles of microglial BTK and impact of BTK inhibitors on microglial functions are still being elucidated. Research on the effects of BTK inhibitors has been limited to rodent disease models. This is the first study reporting effects in human microglia. METHODS Here we characterize the pharmacological and functional properties of fenebrutinib, a potent, highly selective, noncovalent, reversible, brain-penetrant BTK inhibitor, in human microglia and complex human brain cell systems, including brain organoids. RESULTS We find that fenebrutinib blocks the deleterious effects of microglial Fc gamma receptor (FcγR) activation, including cytokine and chemokine release, microglial clustering and neurite damage in diverse human brain cell systems. Gene expression analyses identified pathways linked to inflammation, matrix metalloproteinase production and cholesterol metabolism that were modulated by fenebrutinib treatment. In contrast, fenebrutinib had no significant impact on human microglial pathways linked to Toll-like receptor 4 (TLR4) and NACHT, LRR and PYD domains-containing protein 3 (NLRP3) signaling or myelin phagocytosis. CONCLUSIONS Our study enhances the understanding of BTK functions in human microglial signaling that are relevant to MS pathogenesis and suggests that fenebrutinib could attenuate detrimental microglial activity associated with FcγR activation in people with MS.
Collapse
Affiliation(s)
- Julie Langlois
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Simona Lange
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Martin Ebeling
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Will Macnair
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Roland Schmucki
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Cenxiao Li
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary, 3330 Hospital Dr NW, Calgary, AB, Canada
| | - Jonathan DeGeer
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Tania J J Sudharshan
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - V Wee Yong
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary, 3330 Hospital Dr NW, Calgary, AB, Canada
| | - Yun-An Shen
- Genentech, Inc., 1 DNA Way, South San Francisco, CA, USA
| | | | - Ludovic Collin
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - James Keaney
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland.
| |
Collapse
|
18
|
Mbah NE, Myers AL, Sajjakulnukit P, Chung C, Thompson JK, Hong HS, Giza H, Dang D, Nwosu ZC, Shan M, Sweha SR, Maydan DD, Chen B, Zhang L, Magnuson B, Zhu Z, Radyk M, Lavoie B, Yadav VN, Koo I, Patterson AD, Wahl DR, Franchi L, Agnihotri S, Koschmann CJ, Venneti S, Lyssiotis CA. Therapeutic targeting of differentiation-state dependent metabolic vulnerabilities in diffuse midline glioma. Nat Commun 2024; 15:8983. [PMID: 39419964 PMCID: PMC11487135 DOI: 10.1038/s41467-024-52973-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 09/27/2024] [Indexed: 10/19/2024] Open
Abstract
H3K27M diffuse midline gliomas (DMG), including diffuse intrinsic pontine gliomas (DIPG), exhibit cellular heterogeneity comprising less-differentiated oligodendrocyte precursors (OPC)-like stem cells and more differentiated astrocyte (AC)-like cells. Here, we establish in vitro models that recapitulate DMG-OPC-like and AC-like phenotypes and perform transcriptomics, metabolomics, and bioenergetic profiling to identify metabolic programs in the different cellular states. We then define strategies to target metabolic vulnerabilities within specific tumor populations. We show that AC-like cells exhibit a mesenchymal phenotype and are sensitized to ferroptotic cell death. In contrast, OPC-like cells upregulate cholesterol biosynthesis, have diminished mitochondrial oxidative phosphorylation (OXPHOS), and are accordingly more sensitive to statins and OXPHOS inhibitors. Additionally, statins and OXPHOS inhibitors show efficacy and extend survival in preclinical orthotopic models established with stem-like H3K27M DMG cells. Together, this study demonstrates that cellular subtypes within DMGs harbor distinct metabolic vulnerabilities that can be uniquely and selectively targeted for therapeutic gain.
Collapse
Affiliation(s)
- Nneka E Mbah
- Chad Carr Pediatric Brain Tumor Center, University of Michigan, Ann Arbor, USA
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, USA
| | - Amy L Myers
- Chad Carr Pediatric Brain Tumor Center, University of Michigan, Ann Arbor, USA
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, USA
| | - Peter Sajjakulnukit
- Chad Carr Pediatric Brain Tumor Center, University of Michigan, Ann Arbor, USA
- Graduate Program in Cancer Biology, University of Michigan, Ann Arbor, USA
| | - Chan Chung
- Chad Carr Pediatric Brain Tumor Center, University of Michigan, Ann Arbor, USA
- Laboratory of Brain Tumor Metabolism and Epigenetics, Department of Pathology, University of Michigan Medical School, Ann Arbor, USA
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, South Korea
| | | | - Hanna S Hong
- Graduate Program in Immunology, University of Michigan, Ann Arbor, USA
| | - Heather Giza
- Graduate Program in Cancer Biology, University of Michigan, Ann Arbor, USA
| | - Derek Dang
- Chad Carr Pediatric Brain Tumor Center, University of Michigan, Ann Arbor, USA
- Laboratory of Brain Tumor Metabolism and Epigenetics, Department of Pathology, University of Michigan Medical School, Ann Arbor, USA
- Graduate Program in Molecular & Cellular Pathology, University of Michigan, Ann Arbor, USA
| | - Zeribe C Nwosu
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, USA
| | - Mengrou Shan
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, USA
| | - Stefan R Sweha
- Chad Carr Pediatric Brain Tumor Center, University of Michigan, Ann Arbor, USA
- Laboratory of Brain Tumor Metabolism and Epigenetics, Department of Pathology, University of Michigan Medical School, Ann Arbor, USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, USA
| | - Daniella D Maydan
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, USA
| | - Brandon Chen
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, USA
- Graduate Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, USA
| | - Li Zhang
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, USA
| | - Brian Magnuson
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, USA
| | - Zirui Zhu
- Graduate Program in Chemical Biology, University of Michigan, Ann Arbor, USA
| | - Megan Radyk
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, USA
| | - Brooke Lavoie
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, USA
| | - Viveka Nand Yadav
- The Department of Pediatrics, Children's Mercy Research Institute (CMRI), Kansas, USA
| | - Imhoi Koo
- Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, USA
| | - Andrew D Patterson
- Department of Biochemistry and Molecular Biology and Department of Veterinary and Biomedical Sciences, the Pennsylvania State University, University Park, USA
| | - Daniel R Wahl
- Chad Carr Pediatric Brain Tumor Center, University of Michigan, Ann Arbor, USA
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, USA
| | - Luigi Franchi
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, USA
| | | | - Carl J Koschmann
- Chad Carr Pediatric Brain Tumor Center, University of Michigan, Ann Arbor, USA
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, USA
| | - Sriram Venneti
- Chad Carr Pediatric Brain Tumor Center, University of Michigan, Ann Arbor, USA.
- Laboratory of Brain Tumor Metabolism and Epigenetics, Department of Pathology, University of Michigan Medical School, Ann Arbor, USA.
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, USA.
- Department of Pathology, University of Michigan Medical School, Ann Arbor, USA.
| | - Costas A Lyssiotis
- Chad Carr Pediatric Brain Tumor Center, University of Michigan, Ann Arbor, USA.
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, USA.
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, USA.
- Rogel Cancer Center, University of Michigan, Ann Arbor, USA.
| |
Collapse
|
19
|
Fanlo-Ucar H, Picón-Pagès P, Herrera-Fernández V, ILL-Raga G, Muñoz FJ. The Dual Role of Amyloid Beta-Peptide in Oxidative Stress and Inflammation: Unveiling Their Connections in Alzheimer's Disease Etiopathology. Antioxidants (Basel) 2024; 13:1208. [PMID: 39456461 PMCID: PMC11505517 DOI: 10.3390/antiox13101208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 10/03/2024] [Accepted: 10/05/2024] [Indexed: 10/28/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease, and it is currently the seventh leading cause of death worldwide. It is characterized by the extracellular aggregation of the amyloid β-peptide (Aβ) into oligomers and fibrils that cause synaptotoxicity and neuronal death. Aβ exhibits a dual role in promoting oxidative stress and inflammation. This review aims to unravel the intricate connection between these processes and their contribution to AD progression. The review delves into oxidative stress in AD, focusing on the involvement of metals, mitochondrial dysfunction, and biomolecule oxidation. The distinct yet overlapping concept of nitro-oxidative stress is also discussed, detailing the roles of nitric oxide, mitochondrial perturbations, and their cumulative impact on Aβ production and neurotoxicity. Inflammation is examined through astroglia and microglia function, elucidating their response to Aβ and their contribution to oxidative stress within the AD brain. The blood-brain barrier and oligodendrocytes are also considered in the context of AD pathophysiology. We also review current diagnostic methodologies and emerging therapeutic strategies aimed at mitigating oxidative stress and inflammation, thereby offering potential treatments for halting or slowing AD progression. This comprehensive synthesis underscores the pivotal role of Aβ in bridging oxidative stress and inflammation, advancing our understanding of AD and informing future research and treatment paradigms.
Collapse
Affiliation(s)
- Hugo Fanlo-Ucar
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (H.F.-U.); (P.P.-P.); (V.H.-F.); (G.I.-R.)
| | - Pol Picón-Pagès
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (H.F.-U.); (P.P.-P.); (V.H.-F.); (G.I.-R.)
- Laboratory of Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), 08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 08028 Barcelona, Spain
| | - Víctor Herrera-Fernández
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (H.F.-U.); (P.P.-P.); (V.H.-F.); (G.I.-R.)
| | - Gerard ILL-Raga
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (H.F.-U.); (P.P.-P.); (V.H.-F.); (G.I.-R.)
| | - Francisco J. Muñoz
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (H.F.-U.); (P.P.-P.); (V.H.-F.); (G.I.-R.)
| |
Collapse
|
20
|
Chalitsios CV, Ley H, Gao J, Turner MR, Thompson AG. Apolipoproteins, lipids, lipid-lowering drugs and risk of amyotrophic lateral sclerosis and frontotemporal dementia: a meta-analysis and Mendelian randomisation study. J Neurol 2024; 271:6956-6969. [PMID: 39230722 PMCID: PMC11447100 DOI: 10.1007/s00415-024-12665-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/05/2024]
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) have clinical, pathological and genetic overlapping. Lipid pathways are implicated in ALS. This study examined the effect of blood lipid levels on ALS, FTD risk, and survival in ALS. METHODS A systematic review and meta-analysis of high and low-density lipoprotein cholesterol (HDL-c and LDL-c), total cholesterol, triglycerides, apolipoproteins B and A1 levels with ALS was performed. Two-sample Mendelian randomisation (MR) analysis sought the causal effects of these exposures on ALS, FTD, and survival in ALS. The effect of lipid-lowering drugs was also examined using genetic proxies for targets of lipid-lowering medications. RESULTS Three cohort studies met the inclusion criteria for meta-analysis. Meta-analysis indicated an association between higher LDL-c (HRper mmol/L = 1.07, 95%CI:1.02-1.12;I 2 =18%) and lower HDL-c (HRper mmol/L = 0.83, 95%CI:0.74-0.94;I 2 =0%) with an increased risk of ALS. MR suggested causal effects of higher LDL-c (ORIVW = 1.085, 95%:CI 1.008-1.168, pFDR = 0.0406), total cholesterol (ORIVW = 1.081, 95%:CI 1.013-1.154, pFDR = 0.0458) and apolipoprotein B (ORIVW = 1.104, 95%:CI 1.041-1.171, pFDR = 0.0061) increasing ALS risk, and higher apolipoprotein B level increasing FTD risk (ORIVW = 1.424, 95%CI 1.072-1.829, pFDR = 0.0382). Reducing LDL-c through APOB inhibition was associated with lower ALS (ORIVW = 0.84, 95%CI 0.759-0.929, pFDR = 0.00275) and FTD risk (ORIVW = 0.581, 95%CI 0.387-0.874, pFDR = 0.0362). CONCLUSION These data support the influence of LDL-c and total cholesterol on ALS risk and apolipoprotein B on the risk of ALS and FTD. Potential APOB inhibition might decrease the risk of sporadic ALS and FTD. Further work in monogenic forms of ALS and FTD is necessary to determine whether blood lipids influence penetrance and phenotype.
Collapse
Affiliation(s)
- Christos V Chalitsios
- Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Level 6, West Wing, Oxford, OX3 9DU, UK
| | - Harriet Ley
- Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Level 6, West Wing, Oxford, OX3 9DU, UK
| | - Jiali Gao
- Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Level 6, West Wing, Oxford, OX3 9DU, UK
| | - Martin R Turner
- Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Level 6, West Wing, Oxford, OX3 9DU, UK
| | - Alexander G Thompson
- Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Level 6, West Wing, Oxford, OX3 9DU, UK.
| |
Collapse
|
21
|
Wang Y, Zhai Y, Wang J. Insight into the early pathogenesis and therapeutic strategies of spinocerebellar ataxia type 3/machado-joseph disease from mouse models. Parkinsonism Relat Disord 2024; 126:106991. [PMID: 38749872 DOI: 10.1016/j.parkreldis.2024.106991] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/31/2024] [Accepted: 04/29/2024] [Indexed: 09/05/2024]
Abstract
Spinocerebellar ataxia type 3 (SCA3), also known as Machado-Joseph disease (MJD), is the most common subtype of hereditary ataxia (HA), which is characterized by motor deficits and a lack of effective treatments, and imposes a huge physical, mental, and financial burden on patients and their families. Therefore, it is important to study the early pathogenesis of spinal cerebellar ataxia type 3 based on a mouse model for subsequent preventive treatment and seeking new therapeutic targets.
Collapse
Affiliation(s)
- Ying Wang
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - YuYun Zhai
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Ju Wang
- Department of Rehabilitation, Traditional Chinese Hospital Medicine of Qing Yang District of Chengdu, Sichuan, China.
| |
Collapse
|
22
|
Romero-Ramírez L, Mey J. Emerging Roles of Bile Acids and TGR5 in the Central Nervous System: Molecular Functions and Therapeutic Implications. Int J Mol Sci 2024; 25:9279. [PMID: 39273226 PMCID: PMC11395147 DOI: 10.3390/ijms25179279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/15/2024] [Accepted: 08/17/2024] [Indexed: 09/15/2024] Open
Abstract
Bile acids (BAs) are cholesterol derivatives synthesized in the liver and released into the digestive tract to facilitate lipid uptake during the digestion process. Most of these BAs are reabsorbed and recycled back to the liver. Some of these BAs progress to other tissues through the bloodstream. The presence of BAs in the central nervous system (CNS) has been related to their capacity to cross the blood-brain barrier (BBB) from the systemic circulation. However, the expression of enzymes and receptors involved in their synthesis and signaling, respectively, support the hypothesis that there is an endogenous source of BAs with a specific function in the CNS. Over the last decades, BAs have been tested as treatments for many CNS pathologies, with beneficial effects. Although they were initially reported as neuroprotective substances, they are also known to reduce inflammatory processes. Most of these effects have been related to the activation of the Takeda G protein-coupled receptor 5 (TGR5). This review addresses the new challenges that face BA research for neuroscience, focusing on their molecular functions. We discuss their endogenous and exogenous sources in the CNS, their signaling through the TGR5 receptor, and their mechanisms of action as potential therapeutics for neuropathologies.
Collapse
Affiliation(s)
- Lorenzo Romero-Ramírez
- Laboratorio de Regeneración Neuronal, Hospital Nacional de Parapléjicos, Servicio de Salud de Castilla-La Mancha, 45071 Toledo, Spain
| | - Jörg Mey
- Laboratorio de Regeneración Neuronal, Hospital Nacional de Parapléjicos, Servicio de Salud de Castilla-La Mancha, 45071 Toledo, Spain
- EURON Graduate School of Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands;
| |
Collapse
|
23
|
Wu M, Wang T, Ji N, Lu T, Yuan R, Wu L, Zhang J, Li M, Cao P, Zhao J, Li G, Li J, Li Y, Tang Y, Gao Z, Wang X, Cheng W, Ge M, Cui G, Li R, Wu A, You Y, Zhang W, Wang Q, Chen J. Multi-omics and pharmacological characterization of patient-derived glioma cell lines. Nat Commun 2024; 15:6740. [PMID: 39112531 PMCID: PMC11306361 DOI: 10.1038/s41467-024-51214-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 07/31/2024] [Indexed: 08/10/2024] Open
Abstract
Glioblastoma (GBM) is the most common brain tumor and remains incurable. Primary GBM cultures are widely used tools for drug screening, but there is a lack of genomic and pharmacological characterization for these primary GBM cultures. Here, we collect 50 patient-derived glioma cell (PDGC) lines and characterize them by whole genome sequencing, RNA sequencing, and drug response screening. We identify three molecular subtypes among PDGCs: mesenchymal (MES), proneural (PN), and oxidative phosphorylation (OXPHOS). Drug response profiling reveals that PN subtype PDGCs are sensitive to tyrosine kinase inhibitors, whereas OXPHOS subtype PDGCs are sensitive to histone deacetylase inhibitors, oxidative phosphorylation inhibitors, and HMG-CoA reductase inhibitors. PN and OXPHOS subtype PDGCs stably form tumors in vivo upon intracranial transplantation into immunodeficient mice, whereas most MES subtype PDGCs fail to form tumors in vivo. In addition, PDGCs cultured by serum-free medium, especially long-passage PDGCs, carry MYC/MYCN amplification, which is rare in GBM patients. Our study provides a valuable resource for understanding primary glioma cell cultures and clinical translation and highlights the problems of serum-free PDGC culture systems that cannot be ignored.
Collapse
Affiliation(s)
- Min Wu
- Chinese Institute for Brain Research, Beijing, Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, Beijing, China
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, China
- Shanghai Immune Therapy Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Tingting Wang
- Chinese Institute for Brain Research, Beijing, Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, Beijing, China
- Changping Laboratory, Beijing, China
- Chinese Institute for Brain Research, Beijing, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Nan Ji
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ting Lu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ran Yuan
- Chinese Institute for Brain Research, Beijing, Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, Beijing, China
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, China
| | - Lingxiang Wu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Junxia Zhang
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Mengyuan Li
- Chinese Institute for Brain Research, Beijing, Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, Beijing, China
- Changping Laboratory, Beijing, China
| | - Penghui Cao
- Chinese Institute for Brain Research, Beijing, Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, Beijing, China
| | - Jiarui Zhao
- Chinese Institute for Brain Research, Beijing, Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, Beijing, China
| | - Guanzhang Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Jianyu Li
- Chinese Institute for Brain Research, Beijing, Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, Beijing, China
| | - Yu Li
- Chinese Institute for Brain Research, Beijing, Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, Beijing, China
| | - Yujie Tang
- Shanghai Key Laboratory of Reproductive Medicine, Department of Histoembryology, Genetics and Developmental Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhengliang Gao
- Fundamental Research Center, Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
- Department of Anesthesiology, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| | - Xiuxing Wang
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Wen Cheng
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ming Ge
- Department of Neurosurgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Gang Cui
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Rui Li
- Department of Neurosurgery, China-Japan Friendship Hospital, No. 2 Yinghua East Road, Chaoyang District, Beijing, China
| | - Anhua Wu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yongping You
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Wei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
| | - Qianghu Wang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China.
- Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China.
| | - Jian Chen
- Chinese Institute for Brain Research, Beijing, Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, Beijing, China.
- Changping Laboratory, Beijing, China.
- Chinese Institute for Brain Research, Beijing, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
24
|
Cleland NRW, Bruce KD. Fatty acid sensing in the brain: The role of glial-neuronal metabolic crosstalk and horizontal lipid flux. Biochimie 2024; 223:166-178. [PMID: 35998849 DOI: 10.1016/j.biochi.2022.08.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/01/2022] [Accepted: 08/17/2022] [Indexed: 11/15/2022]
Abstract
The central control of energy homeostasis is a regulatory axis that involves the sensing of nutrients, signaling molecules, adipokines, and neuropeptides by neurons in the metabolic centers of the hypothalamus. However, non-neuronal glial cells are also abundant in the hypothalamus and recent findings have underscored the importance of the metabolic crosstalk and horizontal lipid flux between glia and neurons to the downstream regulation of systemic metabolism. New transgenic models and high-resolution analyses of glial phenotype and function have revealed that glia sit at the nexus between lipid metabolism and neural function, and may markedly impact the brain's response to dietary lipids or the supply of brain-derived lipids. Glia comprise the main cellular compartment involved in lipid synthesis, lipoprotein production, and lipid processing in the brain. In brief, tanycytes provide an interface between peripheral lipids and neurons, astrocytes produce lipoproteins that transport lipids to neurons and other glia, oligodendrocytes use brain-derived and dietary lipids to myelinate axons and influence neuronal function, while microglia can remove unwanted lipids in the brain and contribute to lipid re-utilization through cholesterol efflux. Here, we review recent findings regarding glial-lipid transport and highlight the specific molecular factors necessary for lipid processing in the brain, and how dysregulation of glial-neuronal metabolic crosstalk contributes to imbalanced energy homeostasis. Furthering our understanding of glial lipid metabolism will guide the design of future studies that target horizontal lipid processing in the brain to ameliorate the risk of developing obesity and metabolic disease.
Collapse
Affiliation(s)
- Nicholas R W Cleland
- Division of Endocrinology Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Kimberley D Bruce
- Division of Endocrinology Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
25
|
Mistry H, Richardson CD, Higginbottom A, Ashford B, Ahamed SU, Moore Z, Matthews FE, Brayne C, Simpson JE, Wharton SB. Relationships of brain cholesterol and cholesterol biosynthetic enzymes to Alzheimer's pathology and dementia in the CFAS population-derived neuropathology cohort. Neurosci Res 2024; 204:22-33. [PMID: 38278219 PMCID: PMC11192635 DOI: 10.1016/j.neures.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 01/05/2024] [Accepted: 01/17/2024] [Indexed: 01/28/2024]
Abstract
Altered cholesterol metabolism is implicated in brain ageing and Alzheimer's disease. We examined whether key genes regulating cholesterol metabolism and levels of brain cholesterol are altered in dementia and Alzheimer's disease neuropathological change (ADNC). Temporal cortex (n = 99) was obtained from the Cognitive Function and Ageing Study. Expression of the cholesterol biosynthesis rate-limiting enzyme HMG-CoA reductase (HMGCR) and its regulator, SREBP2, were detected using immunohistochemistry. Expression of HMGCR, SREBP2, CYP46A1 and ABCA1 were quantified by qPCR in samples enriched for astrocyte and neuronal RNA following laser-capture microdissection. Total cortical cholesterol was measured using the Amplex Red assay. HMGCR and SREBP2 proteins were predominantly expressed in pyramidal neurones, and in glia. Neuronal HMGCR did not vary with ADNC, oxidative stress, neuroinflammation or dementia status. Expression of HMGCR neuronal mRNA decreased with ADNC (p = 0.022) and increased with neuronal DNA damage (p = 0.049), whilst SREBP2 increased with ADNC (p = 0.005). High or moderate tertiles for cholesterol levels were associated with increased dementia risk (OR 1.44, 1.58). APOE ε4 allele was not associated with cortical cholesterol levels. ADNC is associated with gene expression changes that may impair cholesterol biosynthesis in neurones but not astrocytes, whilst levels of cortical cholesterol show a weak relationship to dementia status.
Collapse
Affiliation(s)
- Hemant Mistry
- Sheffield Institute for Translational Neuroscience, and the Neuroscience Institute, the University of Sheffield, UK
| | | | - Adrian Higginbottom
- Sheffield Institute for Translational Neuroscience, and the Neuroscience Institute, the University of Sheffield, UK
| | - Bridget Ashford
- Sheffield Institute for Translational Neuroscience, and the Neuroscience Institute, the University of Sheffield, UK
| | - Saif U Ahamed
- Sheffield Institute for Translational Neuroscience, and the Neuroscience Institute, the University of Sheffield, UK
| | - Zoe Moore
- Sheffield Institute for Translational Neuroscience, and the Neuroscience Institute, the University of Sheffield, UK
| | | | - Carol Brayne
- Cambridge Public Health, University of Cambridge, UK
| | - Julie E Simpson
- Sheffield Institute for Translational Neuroscience, and the Neuroscience Institute, the University of Sheffield, UK
| | - Stephen B Wharton
- Sheffield Institute for Translational Neuroscience, and the Neuroscience Institute, the University of Sheffield, UK.
| |
Collapse
|
26
|
Chiappelli J, Savransky A, Ma Y, Gao S, Kvarta MD, Kochunov P, Slavich GM, Hong LE. Impact of lifetime stressor exposure on neuroenergetics in schizophrenia spectrum disorders. Schizophr Res 2024; 269:58-63. [PMID: 38733800 PMCID: PMC11180558 DOI: 10.1016/j.schres.2024.04.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 03/22/2024] [Accepted: 04/28/2024] [Indexed: 05/13/2024]
Abstract
N-acetylasparate and lactate are two prominent brain metabolites closely related to mitochondrial functioning. Prior research revealing lower levels of NAA and higher levels of lactate in the cerebral cortex of patients with schizophrenia suggest possible abnormalities in the energy supply pathway necessary for brain function. Given that stress and adversity are a strong risk factor for a variety of mental health problems, including psychotic disorders, we investigated the hypothesis that stress contributes to abnormal neuroenergetics in patients with schizophrenia. To test this hypothesis, we used the Stress and Adversity Inventory (STRAIN) to comprehensively assess the lifetime stressor exposure profiles of 35 patients with schizophrenia spectrum disorders and 33 healthy controls who were also assessed with proton magnetic resonance spectroscopy at the anterior cingulate cortex using 3 Tesla scanner. Consistent with the hypothesis, greater lifetime stressor exposure was significantly associated with lower levels of N-acetylasparate (β = -0.36, p = .005) and higher levels of lactate (β = 0.43, p = .001). Moreover, these results were driven by patients, as these associations were significant for the patient but not control group. Though preliminary, these findings suggest a possible role for stress processes in the pathophysiology of abnormal neuroenergetics in schizophrenia.
Collapse
Affiliation(s)
- Joshua Chiappelli
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Anya Savransky
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Yizhou Ma
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Si Gao
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mark D Kvarta
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Peter Kochunov
- Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - George M Slavich
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| | - L Elliot Hong
- Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
27
|
He L, Ioannidis A, Hoffman CJ, Arambula E, Joshi P, Whitelegge J, Liau LM, Kornblum HI, Pajonk F. Activation of the Mevalonate Pathway in Response to Anti-cancer Treatments Drives Glioblastoma Recurrences Through Activation of Rac-1. CANCER RESEARCH COMMUNICATIONS 2024; 4:1566-1580. [PMID: 38837899 PMCID: PMC11197925 DOI: 10.1158/2767-9764.crc-24-0049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/26/2024] [Accepted: 05/31/2024] [Indexed: 06/07/2024]
Abstract
Glioblastoma (GBM) is the deadliest adult brain cancer. Under the current standard of care, almost all patients succumb to the disease and novel treatments are urgently needed. Recognizing that GBMs are addicted to cholesterol, past clinical trials have repurposed statins against GBM but failed. The purpose of this study was to test whether treatments that upregulate the cholesterol biosynthesis pathway in GBM would generate a metabolic vulnerability that can be exploited using statins and to determine the underlying mechanisms.Effects of radiotherapy and temozolomide or dopamine receptor antagonists on the mevalonate pathway in GBM were assessed in vitro and in vivo. The impact of statins on self-renewal of glioma stem cells and median survival was studied. Branches of the mevalonate pathway were probed to identify relevant effector proteins.Cells surviving combination treatments that converge in activating the immediate early response, universally upregulated the mevalonate pathway and increased stemness of GBM cells through activation of the Rho-GTPase Rac-1. Activation of the mevalonate pathway and Rac-1 was inhibited by statins, which led to improved survival in mouse models of glioblastoma when combined with radiation and drugs that target the glioma stem cell pool and plasticity of glioma cells.We conclude that a combination of dopamine receptor antagonists and statins could potentially improve radiotherapy outcome and warrants further investigation. SIGNIFICANCE Combination therapies that activate the mevalonate pathway in GBM cells after sublethal treatment enhance self-renewal and migratory capacity through Rac-1 activation, which creates a metabolic vulnerability that can be further potentially exploited using statins.
Collapse
Affiliation(s)
- Ling He
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, Los Angeles, California
- Jonsson Comprehensive Cancer Center at UCLA, Los Angeles, California
| | - Angeliki Ioannidis
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Carter J. Hoffman
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Evelyn Arambula
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Purva Joshi
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Julian Whitelegge
- Jonsson Comprehensive Cancer Center at UCLA, Los Angeles, California
- Department of Psychiatry and Human Behavior, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Linda M. Liau
- Jonsson Comprehensive Cancer Center at UCLA, Los Angeles, California
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Harley I. Kornblum
- Jonsson Comprehensive Cancer Center at UCLA, Los Angeles, California
- Department of Psychiatry and Human Behavior, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Frank Pajonk
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, Los Angeles, California
- Jonsson Comprehensive Cancer Center at UCLA, Los Angeles, California
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, California
| |
Collapse
|
28
|
Yasamineh S, Mehrabani FJ, Derafsh E, Danihiel Cosimi R, Forood AMK, Soltani S, Hadi M, Gholizadeh O. Potential Use of the Cholesterol Transfer Inhibitor U18666A as a Potent Research Tool for the Study of Cholesterol Mechanisms in Neurodegenerative Disorders. Mol Neurobiol 2024; 61:3503-3527. [PMID: 37995080 DOI: 10.1007/s12035-023-03798-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 11/03/2023] [Indexed: 11/24/2023]
Abstract
Cholesterol is an essential component of mammalian cell membranes and a precursor for crucial signaling molecules. The brain contains the highest level of cholesterol in the body, and abnormal cholesterol metabolism links to many neurodegenerative disorders. The results indicate that faulty cholesterol metabolism is a common feature among people living with neurodegenerative conditions. The researchers suggest that restoring cholesterol levels may become a beneficial new strategy in treating certain neurodegenerative conditions. Several neurodegenerative disorders, such as Alzheimer's disease (AD), Niemann-Pick type C (NPC) disease, and Parkinson's disease (PD), have been connected to abnormalities in brain cholesterol metabolism. Consequently, using a lipid research tool is vital to study further and understand the effect of lipids in neurodegenerative disorders such as NPC, AD, PD, and Huntington's disease (HD). U18666A, also known as 3-(2-(diethylamino) ethoxy) androst-5-en-17-one, is a pharmaceutical drug that suppresses cholesterol trafficking and is a well-known class-2 amphiphile. U18666A has performed many functions, allowing for essential discoveries in lipid studies and shedding light on the pathophysiology of neurodegenerative disorders. Additionally, U18666A prevented the downregulation of low-density lipoprotein (LDL) receptors that are induced by LDL and led to the buildup of cholesterol in lysosomes. Numerous studies show that U18666A impacts the function of cholesterol trafficking to control the metabolism and transport of amyloid precursor proteins (APPs). Treating cortical neurons with U18666A may provide a new in vitro model system for studying the underlying molecular process of NPC, AD, HD, and PD. In this article, we review the mechanism and function of U18666A as a vital tool for studying cholesterol mechanisms in neurological diseases related to abnormal cholesterol metabolism, such as AD, NPC, HD, and PD.
Collapse
Affiliation(s)
| | | | - Ehsan Derafsh
- Windsor University School of Medicine, Cayon, Saint Kitts and Nevis
| | | | | | - Siamak Soltani
- Department of Forensic Medicine, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Meead Hadi
- Department Of Microbiology, Faculty of Basic Sciences, Tehran Central Branch, Islamic Azad University, Tehran, Iran
| | | |
Collapse
|
29
|
Son SM, Lee HS, Kim J, Kwon RJ. Expression and prognostic significance of microsomal triglyceride transfer protein in brain tumors: a retrospective cohort study. Transl Cancer Res 2024; 13:2282-2294. [PMID: 38881934 PMCID: PMC11170499 DOI: 10.21037/tcr-23-2286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 04/10/2024] [Indexed: 06/18/2024]
Abstract
Background Glioblastoma (GBM) is the most common malignant brain tumor and has poor survival. An elevated cholesterol level is involved occurrence and progression of brain tumors. Microsomal triglyceride transfer protein (MTTP) is a target for lowering lipids, and its inhibition helps to improve hyperlipidemia. However, whether the altered expression of MTTP affects the development and prognosis of brain tumors is currently unidentified. The purpose of this study is to determine MTTP as a prognostic marker for brain tumors. Methods Data for patients with brain cancers and control brain tissue were acquired from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO). The datasets were analyzed using Mann-Whitney U-test or t-test to compare the expression of MTTP in normal and brain tumor tissues. To examine whether MTTP affected the prognosis of patients with brain tumors, log-rank test and multivariable Cox proportional hazard regression were conducted. Results The expression of MTTP was significantly upregulated in brain tumors and was correlated with age, tumor stage, and isocitrate dehydrogenase (IDH) mutation. Importantly, increased MTTP expression in brain tumors is associated with poor patient survival. Conclusions High MTTP expression is associated with brain tumor development, tumor stage, and prognosis. Therefore, MTTP is an independent prognostic indicator for brain tumors, which can serve as one of the possible targets for adjuvant treatment of GBM.
Collapse
Affiliation(s)
- Soo Min Son
- Family Medicine Clinic and Research Institute of Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Korea
- Department of Family Medicine, Pusan National University School of Medicine, Yangsan, Korea
| | - Hye Sun Lee
- Family Medicine Clinic and Research Institute of Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Jeongsu Kim
- Division of Cardiology, Department of Internal Medicine, Pusan National University Yangsan Hospital, Yangsan, Korea
- Division of Cardiology, Department of Internal Medicine, Pusan National University School of Medicine, Yangsan, Korea
| | - Ryuk Jun Kwon
- Family Medicine Clinic and Research Institute of Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Korea
- Department of Family Medicine, Pusan National University School of Medicine, Yangsan, Korea
| |
Collapse
|
30
|
Shibuya T, Sato A, Nishimoto-Kusunose S, Yoshizawa K, Higashi T. Further evidence for blood-to-brain influx of unconjugated bile acids by passive diffusion: Determination of their brain-to-serum concentration ratios in rats by LC/MS/MS. Steroids 2024; 204:109397. [PMID: 38508483 DOI: 10.1016/j.steroids.2024.109397] [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: 01/28/2024] [Revised: 02/28/2024] [Accepted: 03/17/2024] [Indexed: 03/22/2024]
Abstract
Bile acids (BAs) reside in the brain and are probably involved in some neurological disorders. The view that most of unconjugated BAs in the brain are derived across the blood-brain barrier from the periphery by passive diffusion depending on their hydrophobicity is currently dominant, but some studies have made conflicting claims. In this study, the correlation analysis between the rat brain and serum levels of unconjugated BAs with a wider range of hydrophobicity was conducted to obtain further evidence about the blood-to-brain influx of unconjugated BAs by passive diffusion. We first developed the precise, accurate and matrix effect-free LC/ESI-MS/MS methods for quantifying eight major unconjugated BAs in the rat brain and serum. Derivatization was employed for increasing the assay sensitivity and specificity. The analysis using these methods reproduced the strong positive correlations between the brain and serum levels, and significant higher concentrations in the serum than in the brain for all the unconjugated BAs. The BA with the higher logPow (hydrophobicity) had the higher brain-to-serum concentration ratio (mono- > di- > trihydroxy BAs). Furthermore, the hydrophobicity was considered as the stronger factor for the blood-to-brain influx of the BAs than the serum protein binding ratio. Thus, this study provided further evidence supporting that passive diffusion is the major mechanism for the blood-to-brain influx of the unconjugated BAs.
Collapse
Affiliation(s)
- Toma Shibuya
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Anri Sato
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Shoich Nishimoto-Kusunose
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Kazumi Yoshizawa
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Tatsuya Higashi
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan.
| |
Collapse
|
31
|
Li Z, Yu S, Li L, Zhou C, Wang L, Tang S, Gu N, Zhang Z, Huang Z, Chen H, Tang W, Wang Y, Yang X, Sun X, Yan J. TREM2 alleviates white matter injury after traumatic brain injury in mice might be mediated by regulation of DHCR24/LXR pathway in microglia. Clin Transl Med 2024; 14:e1665. [PMID: 38649789 PMCID: PMC11035381 DOI: 10.1002/ctm2.1665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 03/26/2024] [Accepted: 04/04/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND White matter injury (WMI) is an important pathological process after traumatic brain injury (TBI). The correlation between white matter functions and the myeloid cells expressing triggering receptor-2 (TREM2) has been convincingly demonstrated. Moreover, a recent study revealed that microglial sterol metabolism is crucial for early remyelination after demyelinating diseases. However, the potential roles of TREM2 expression and microglial sterol metabolism in WMI after TBI have not yet been explored. METHODS Controlled cortical injury was induced in both wild-type (WT) and TREM2 depletion (TREM2 KO) mice to simulate clinical TBI. COG1410 was used to upregulate TREM2, while PLX5622 and GSK2033 were used to deplete microglia and inhibit the liver X receptor (LXR), respectively. Immunofluorescence, Luxol fast blue staining, magnetic resonance imaging, transmission electron microscopy, and oil red O staining were employed to assess WMI after TBI. Neurological behaviour tests and electrophysiological recordings were utilized to evaluate cognitive functions following TBI. Microglial cell sorting and transcriptomic sequencing were utilized to identify alterations in microglial sterol metabolism-related genes, while western blot was conducted to validate the findings. RESULTS TREM2 expressed highest at 3 days post-TBI and was predominantly localized to microglial cells within the white matter. Depletion of TREM2 worsened aberrant neurological behaviours, and this phenomenon was mediated by the exacerbation of WMI, reduced renewal of oligodendrocytes, and impaired phagocytosis ability of microglia after TBI. Subsequently, the upregulation of TREM2 alleviated WMI, promoted oligodendrocyte regeneration, and ultimately facilitated the recovery of neurological behaviours after TBI. Finally, the expression of DHCR24 increased in TREM2 KO mice after TBI. Interestingly, TREM2 inhibited DHCR24 and upregulated members of the LXR pathway. Moreover, LXR inhibition could partially reverse the effects of TREM2 upregulation on electrophysiological activities. CONCLUSIONS We demonstrate that TREM2 has the potential to alleviate WMI following TBI, possibly through the DHCR24/LXR pathway in microglia.
Collapse
Affiliation(s)
- Zhao Li
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Emergency DepartmentChengdu First People's HospitalChengduChina
| | - Shenghui Yu
- Emergency DepartmentChengdu First People's HospitalChengduChina
| | - Lin Li
- Department of NeurosurgeryChongqing University Cancer HospitalChongqingChina
| | - Chao Zhou
- Emergency DepartmentChengdu First People's HospitalChengduChina
| | - Lin Wang
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Department of NeurosurgeryNanchong Central HospitalThe Second Clinical Medical College of North Sichuan Medical CollegeNanchongChina
| | - Shuang Tang
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Department of NeurosurgerySuining Central HospitalSuiningChina
| | - Nina Gu
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Zhaosi Zhang
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Zhijian Huang
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Hong Chen
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Wei Tang
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Yingwen Wang
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Xiaomin Yang
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Xiaochuan Sun
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Jin Yan
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| |
Collapse
|
32
|
Fang XX, Wei P, Zhao K, Sheng ZC, Song BL, Yin L, Luo J. Fatty acid-binding proteins 3, 7, and 8 bind cholesterol and facilitate its egress from lysosomes. J Cell Biol 2024; 223:e202211062. [PMID: 38429999 PMCID: PMC10909654 DOI: 10.1083/jcb.202211062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 03/22/2023] [Accepted: 01/18/2024] [Indexed: 03/03/2024] Open
Abstract
Cholesterol from low-density lipoprotein (LDL) can be transported to many organelle membranes by non-vesicular mechanisms involving sterol transfer proteins (STPs). Fatty acid-binding protein (FABP) 7 was identified in our previous study searching for new regulators of intracellular cholesterol trafficking. Whether FABP7 is a bona fide STP remains unknown. Here, we found that FABP7 deficiency resulted in the accumulation of LDL-derived cholesterol in lysosomes and reduced cholesterol levels on the plasma membrane. A crystal structure of human FABP7 protein in complex with cholesterol was resolved at 2.7 Å resolution. In vitro, FABP7 efficiently transported the cholesterol analog dehydroergosterol between the liposomes. Further, the silencing of FABP3 and 8, which belong to the same family as FABP7, caused robust cholesterol accumulation in lysosomes. These two FABP proteins could transport dehydroergosterol in vitro as well. Collectively, our results suggest that FABP3, 7, and 8 are a new class of STPs mediating cholesterol egress from lysosomes.
Collapse
Affiliation(s)
- Xian-Xiu Fang
- The Institute for Advanced Studies, College of Life Sciences, Hubei Key Laboratory of Cell Homeostasis, Taikang Center for Life and Medical Sciences, Taikang Medical School, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Pengcheng Wei
- The Institute for Advanced Studies, College of Life Sciences, Hubei Key Laboratory of Cell Homeostasis, Taikang Center for Life and Medical Sciences, Taikang Medical School, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Kai Zhao
- The Institute for Advanced Studies, College of Life Sciences, Hubei Key Laboratory of Cell Homeostasis, Taikang Center for Life and Medical Sciences, Taikang Medical School, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Zhao-Chen Sheng
- The Institute for Advanced Studies, College of Life Sciences, Hubei Key Laboratory of Cell Homeostasis, Taikang Center for Life and Medical Sciences, Taikang Medical School, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Bao-Liang Song
- The Institute for Advanced Studies, College of Life Sciences, Hubei Key Laboratory of Cell Homeostasis, Taikang Center for Life and Medical Sciences, Taikang Medical School, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Lei Yin
- The Institute for Advanced Studies, College of Life Sciences, Hubei Key Laboratory of Cell Homeostasis, Taikang Center for Life and Medical Sciences, Taikang Medical School, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Jie Luo
- The Institute for Advanced Studies, College of Life Sciences, Hubei Key Laboratory of Cell Homeostasis, Taikang Center for Life and Medical Sciences, Taikang Medical School, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| |
Collapse
|
33
|
Xu N, He Y, Zhang C, Zhang Y, Cheng S, Deng L, Zhong Y, Liao B, Wei Y, Feng J. TGR5 signalling in heart and brain injuries: focus on metabolic and ischaemic mechanisms. Neurobiol Dis 2024; 192:106428. [PMID: 38307367 DOI: 10.1016/j.nbd.2024.106428] [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: 10/06/2023] [Revised: 01/28/2024] [Accepted: 01/31/2024] [Indexed: 02/04/2024] Open
Abstract
The heart and brain are the core organs of the circulation and central nervous system, respectively, and play an important role in maintaining normal physiological functions. Early neuronal and cardiac damage affects organ function. The relationship between the heart and brain is being continuously investigated. Evidence-based medicine has revealed the concept of the "heart- brain axis," which may provide new therapeutic strategies for certain diseases. Takeda protein-coupled receptor 5 (TGR5) is a metabolic regulator involved in energy homeostasis, bile acid homeostasis, and glucose and lipid metabolism. Inflammation is critical for the development and regeneration of the heart and brain during metabolic diseases. Herein, we discuss the role of TGR5 as a metabolic regulator of heart and brain development and injury to facilitate new therapeutic strategies for metabolic and ischemic diseases of the heart and brain.
Collapse
Affiliation(s)
- Nan Xu
- Department of Cardiology, The First People's Hospital of Neijiang, Neijiang, China
| | - Yufeng He
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Chunyu Zhang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Yongqiang Zhang
- Department of Cardiology, Hejiang County People's Hospital, Luzhou, China
| | - Shengjie Cheng
- Department of Cardiology, The First People's Hospital of Neijiang, Neijiang, China
| | - Li Deng
- Department of Rheumatology, The Afliated Hospital of Southwest Medical University, Luzhou, China
| | - Yi Zhong
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Bin Liao
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Luzhou, China
| | - Yan Wei
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China.
| | - Jian Feng
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China.
| |
Collapse
|
34
|
Yi W, Lv D, Sun Y, Mu J, Lu X. Role of APOE in glaucoma. Biochem Biophys Res Commun 2024; 694:149414. [PMID: 38145596 DOI: 10.1016/j.bbrc.2023.149414] [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: 10/27/2023] [Revised: 12/15/2023] [Accepted: 12/19/2023] [Indexed: 12/27/2023]
Abstract
Glaucoma is a chronic blinding eye disease caused by the progressive loss of retinal ganglion cells (RGCs). Currently, no clinically approved treatment can directly improve the survival rate of RGCs. The Apolipoprotein E (APOE) gene is closely related to the genetic risk of numerous neurodegenerative diseases and has become a hot topic in the field of neurodegenerative disease research in recent years. The optic nerve and retina are extensions of the brain's nervous system. The pathogenesis of retinal degenerative diseases is closely related to the degenerative diseases of the nerves in the brain. APOE consists of three alleles, ε4, ε3, and ε2, in a single locus. They have varying degrees of risk for glaucoma. APOE4 and the APOE gene deletion (APOE-/-) can reduce RGC loss. By contrast, APOE3 and the overall presence of APOE genes (APOE+/+) result in significant loss of RGC bodies and axons, increasing the risk of glaucoma RGCs death. Currently, there is no clear literature indicating that APOE2 is beneficial or harmful to glaucoma. This study summarises the mechanism of different APOE genes in glaucoma and speculates that APOE targeted intervention may be a promising method for protecting against RGCs loss in glaucoma.
Collapse
Affiliation(s)
- Wenhua Yi
- Eye School of Chengdu University of TCM, Chengdu City, Sichuan province, China.
| | - De Lv
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, China.
| | - Yue Sun
- Eye School of Chengdu University of TCM, Chengdu City, Sichuan province, China.
| | - Jingyu Mu
- Eye School of Chengdu University of TCM, Chengdu City, Sichuan province, China.
| | - Xuejing Lu
- Eye School of Chengdu University of TCM, Chengdu City, Sichuan province, China; Ineye Hospital of Chengdu University of TCM, Chengdu City, Sichuan province, China; Key Laboratory of Sichuan Province Ophthalmopathy Prevention & Cure and Visual Function Protection with TCM Laboratory, Chengdu City, Sichuan province, China; Retinal Image Technology and Chronic Vascular Disease Prevention&Control and Collaborative Innovation Center, Chengdu City, Sichuan province, China.
| |
Collapse
|
35
|
Darwish A, Pammer M, Gallyas F, Vígh L, Balogi Z, Juhász K. Emerging Lipid Targets in Glioblastoma. Cancers (Basel) 2024; 16:397. [PMID: 38254886 PMCID: PMC10814456 DOI: 10.3390/cancers16020397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/09/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
GBM accounts for most of the fatal brain cancer cases, making it one of the deadliest tumor types. GBM is characterized by severe progression and poor prognosis with a short survival upon conventional chemo- and radiotherapy. In order to improve therapeutic efficiency, considerable efforts have been made to target various features of GBM. One of the targetable features of GBM is the rewired lipid metabolism that contributes to the tumor's aggressive growth and penetration into the surrounding brain tissue. Lipid reprogramming allows GBM to acquire survival, proliferation, and invasion benefits as well as supportive modulation of the tumor microenvironment. Several attempts have been made to find novel therapeutic approaches by exploiting the lipid metabolic reprogramming in GBM. In recent studies, various components of de novo lipogenesis, fatty acid oxidation, lipid uptake, and prostaglandin synthesis have been considered promising targets in GBM. Emerging data also suggest a significant role hence therapeutic potential of the endocannabinoid metabolic pathway in GBM. Here we review the lipid-related GBM characteristics in detail and highlight specific targets with their potential therapeutic use in novel antitumor approaches.
Collapse
Affiliation(s)
- Ammar Darwish
- Institute of Biochemistry and Medical Chemistry, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Milán Pammer
- Institute of Biochemistry and Medical Chemistry, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Ferenc Gallyas
- Institute of Biochemistry and Medical Chemistry, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - László Vígh
- Institute of Biochemistry, HUN-REN Biological Research Center, 6726 Szeged, Hungary
| | - Zsolt Balogi
- Institute of Biochemistry and Medical Chemistry, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Kata Juhász
- Institute of Biochemistry and Medical Chemistry, Medical School, University of Pécs, 7624 Pécs, Hungary
| |
Collapse
|
36
|
Bao X, Liang Y, Chang H, Cai T, Feng B, Gordon K, Zhu Y, Shi H, He Y, Xie L. Targeting proprotein convertase subtilisin/kexin type 9 (PCSK9): from bench to bedside. Signal Transduct Target Ther 2024; 9:13. [PMID: 38185721 PMCID: PMC10772138 DOI: 10.1038/s41392-023-01690-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 09/27/2023] [Accepted: 10/27/2023] [Indexed: 01/09/2024] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) has evolved as a pivotal enzyme in lipid metabolism and a revolutionary therapeutic target for hypercholesterolemia and its related cardiovascular diseases (CVD). This comprehensive review delineates the intricate roles and wide-ranging implications of PCSK9, extending beyond CVD to emphasize its significance in diverse physiological and pathological states, including liver diseases, infectious diseases, autoimmune disorders, and notably, cancer. Our exploration offers insights into the interaction between PCSK9 and low-density lipoprotein receptors (LDLRs), elucidating its substantial impact on cholesterol homeostasis and cardiovascular health. It also details the evolution of PCSK9-targeted therapies, translating foundational bench discoveries into bedside applications for optimized patient care. The advent and clinical approval of innovative PCSK9 inhibitory therapies (PCSK9-iTs), including three monoclonal antibodies (Evolocumab, Alirocumab, and Tafolecimab) and one small interfering RNA (siRNA, Inclisiran), have marked a significant breakthrough in cardiovascular medicine. These therapies have demonstrated unparalleled efficacy in mitigating hypercholesterolemia, reducing cardiovascular risks, and have showcased profound value in clinical applications, offering novel therapeutic avenues and a promising future in personalized medicine for cardiovascular disorders. Furthermore, emerging research, inclusive of our findings, unveils PCSK9's potential role as a pivotal indicator for cancer prognosis and its prospective application as a transformative target for cancer treatment. This review also highlights PCSK9's aberrant expression in various cancer forms, its association with cancer prognosis, and its crucial roles in carcinogenesis and cancer immunity. In conclusion, this synthesized review integrates existing knowledge and novel insights on PCSK9, providing a holistic perspective on its transformative impact in reshaping therapeutic paradigms across various disorders. It emphasizes the clinical value and effect of PCSK9-iT, underscoring its potential in advancing the landscape of biomedical research and its capabilities in heralding new eras in personalized medicine.
Collapse
Affiliation(s)
- Xuhui Bao
- Institute of Therapeutic Cancer Vaccines, Fudan University Pudong Medical Center, Shanghai, China.
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China.
- Department of Oncology, Fudan University Pudong Medical Center, Shanghai, China.
- Center for Clinical Research, Fudan University Pudong Medical Center, Shanghai, China.
- Clinical Research Center for Cell-based Immunotherapy, Fudan University, Shanghai, China.
- Department of Pathology, Duke University Medical Center, Durham, NC, USA.
| | - Yongjun Liang
- Center for Medical Research and Innovation, Fudan University Pudong Medical Center, Shanghai, China
| | - Hanman Chang
- Institute for Food Safety and Health, Illinois Institute of Technology, Chicago, IL, USA
| | - Tianji Cai
- Department of Sociology, University of Macau, Taipa, Macau, China
| | - Baijie Feng
- Department of Oncology, Fudan University Pudong Medical Center, Shanghai, China
| | - Konstantin Gordon
- Medical Institute, Peoples' Friendship University of Russia, Moscow, Russia
- A. Tsyb Medical Radiological Research Center, Obninsk, Russia
| | - Yuekun Zhu
- Department of Colorectal Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Hailian Shi
- Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Zhangjiang Hi-tech Park, Shanghai, China
| | - Yundong He
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China.
| | - Liyi Xie
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
37
|
Jaunmuktane Z. Neuropathology of white matter disorders. HANDBOOK OF CLINICAL NEUROLOGY 2024; 204:3-20. [PMID: 39322386 DOI: 10.1016/b978-0-323-99209-1.00011-9] [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: 09/27/2024]
Abstract
The hallmark neuropathologic feature of all leukodystrophies is depletion or alteration of the white matter of the central nervous system; however increasing genetic discoveries highlight the genetic heterogeneity of white matter disorders. These discoveries have significantly helped to advance the understanding of the complexity of molecular mechanisms involved in the biogenesis and maintenance of healthy white matter. Accordingly, genetic discoveries and functional studies have enabled us to firmly establish that multiple distinct structural defects can lead to white matter pathology. Leukodystrophies can develop not only due to defects in proteins essential for myelin biogenesis and maintenance or oligodendrocyte function, but also due to mutations encoding myriad of proteins involved in the function of neurons, astrocytes, microglial cells as well as blood vessels. To a variable extent, some leukodystrophies also show gray matter, peripheral nervous system, or multisystem involvement. Depending on the genetic defect and its role in the formation or maintenance of the white matter, leukodystrophies can present either in early childhood or adulthood. In this chapter, the classification of leukodystrophies will be discussed from the cellular defect point of view, followed by a description of known neuropathologic alterations for all leukodystrophies.
Collapse
Affiliation(s)
- Zane Jaunmuktane
- Division of Neuropathology, National Hospital for Neurology and Neurosurgery, University College London Hospitals, London, United Kingdom; Queen Square Brain Bank for Neurological Disorders, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom.
| |
Collapse
|
38
|
Hu ZL, Yuan YQ, Tong Z, Liao MQ, Yuan SL, Jian Y, Yang JL, Liu WF. Reexamining the Causes and Effects of Cholesterol Deposition in the Brains of Patients with Alzheimer's Disease. Mol Neurobiol 2023; 60:6852-6868. [PMID: 37507575 DOI: 10.1007/s12035-023-03529-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 07/20/2023] [Indexed: 07/30/2023]
Abstract
Alzheimer's disease (AD) is a degenerative disease of the central nervous system. Numerous studies have shown that imbalances in cholesterol homeostasis in the brains of AD patients precede the onset of clinical symptoms. In addition, cholesterol deposition has been observed in the brains of AD patients even though peripheral cholesterol does not enter the brain through the blood‒brain barrier (BBB). Studies have demonstrated that cholesterol metabolism in the brain is associated with many pathological conditions, such as amyloid beta (Aβ) production, Tau protein phosphorylation, oxidative stress, and inflammation. In 2022, some scholars put forward a new hypothesis of AD: the disease involves lipid invasion and its exacerbation of the abnormal metabolism of cholesterol in the brain. In this review, by discussing the latest research progress, the causes and effects of cholesterol retention in the brains of AD patients are analyzed and discussed. Additionally, the possible mechanism through which AD may be improved by targeting cholesterol is described. Finally, we propose that improving the impairments in cholesterol removal observed in the brains of AD patients, instead of further reducing the already impaired cholesterol synthesis in the brain, may be the key to preventing cholesterol deposition and improving the corresponding pathological symptoms.
Collapse
Affiliation(s)
- Ze-Lin Hu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Yang-Qi Yuan
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Zhen Tong
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Mei-Qing Liao
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Shun-Ling Yuan
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Ye Jian
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Jia-Lun Yang
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Wen-Feng Liu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China.
- Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, Hunan Normal University, Changsha, 410081, China.
| |
Collapse
|
39
|
Vanherle S, Guns J, Loix M, Mingneau F, Dierckx T, Wouters F, Kuipers K, Vangansewinkel T, Wolfs E, Lins PP, Bronckaers A, Lambrichts I, Dehairs J, Swinnen JV, Verberk SGS, Haidar M, Hendriks JJA, Bogie JFJ. Extracellular vesicle-associated cholesterol supports the regenerative functions of macrophages in the brain. J Extracell Vesicles 2023; 12:e12394. [PMID: 38124258 PMCID: PMC10733568 DOI: 10.1002/jev2.12394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 11/15/2023] [Accepted: 11/23/2023] [Indexed: 12/23/2023] Open
Abstract
Macrophages play major roles in the pathophysiology of various neurological disorders, being involved in seemingly opposing processes such as lesion progression and resolution. Yet, the molecular mechanisms that drive their harmful and benign effector functions remain poorly understood. Here, we demonstrate that extracellular vesicles (EVs) secreted by repair-associated macrophages (RAMs) enhance remyelination ex vivo and in vivo by promoting the differentiation of oligodendrocyte precursor cells (OPCs). Guided by lipidomic analysis and applying cholesterol depletion and enrichment strategies, we find that EVs released by RAMs show markedly elevated cholesterol levels and that cholesterol abundance controls their reparative impact on OPC maturation and remyelination. Mechanistically, EV-associated cholesterol was found to promote OPC differentiation predominantly through direct membrane fusion. Collectively, our findings highlight that EVs are essential for cholesterol trafficking in the brain and that changes in cholesterol abundance support the reparative impact of EVs released by macrophages in the brain, potentially having broad implications for therapeutic strategies aimed at promoting repair in neurodegenerative disorders.
Collapse
Affiliation(s)
- Sam Vanherle
- Department of Immunology and Infection, Biomedical Research InstituteHasselt UniversityDiepenbeekBelgium
- University MS Center HasseltPeltBelgium
| | - Jeroen Guns
- Department of Immunology and Infection, Biomedical Research InstituteHasselt UniversityDiepenbeekBelgium
- University MS Center HasseltPeltBelgium
| | - Melanie Loix
- Department of Immunology and Infection, Biomedical Research InstituteHasselt UniversityDiepenbeekBelgium
- University MS Center HasseltPeltBelgium
| | - Fleur Mingneau
- Department of Immunology and Infection, Biomedical Research InstituteHasselt UniversityDiepenbeekBelgium
- University MS Center HasseltPeltBelgium
| | - Tess Dierckx
- Department of Immunology and Infection, Biomedical Research InstituteHasselt UniversityDiepenbeekBelgium
- University MS Center HasseltPeltBelgium
| | - Flore Wouters
- Department of Immunology and Infection, Biomedical Research InstituteHasselt UniversityDiepenbeekBelgium
- University MS Center HasseltPeltBelgium
| | - Koen Kuipers
- Department of Immunology and Infection, Biomedical Research InstituteHasselt UniversityDiepenbeekBelgium
- University MS Center HasseltPeltBelgium
| | - Tim Vangansewinkel
- Department of Cardio and Organs Systems, Biomedical Research InstituteHasselt UniversityDiepenbeekBelgium
- VIB, Center for Brain & Disease Research, Laboratory of NeurobiologyUniversity of LeuvenLeuvenBelgium
| | - Esther Wolfs
- Department of Cardio and Organs Systems, Biomedical Research InstituteHasselt UniversityDiepenbeekBelgium
| | - Paula Pincela Lins
- Department of Cardio and Organs Systems, Biomedical Research InstituteHasselt UniversityDiepenbeekBelgium
- Health DepartmentFlemish Institute for Technological ResearchMolBelgium
| | - Annelies Bronckaers
- Department of Cardio and Organs Systems, Biomedical Research InstituteHasselt UniversityDiepenbeekBelgium
| | - Ivo Lambrichts
- Department of Cardio and Organs Systems, Biomedical Research InstituteHasselt UniversityDiepenbeekBelgium
| | - Jonas Dehairs
- Department of Oncology, Laboratory of Lipid Metabolism and Cancer, Leuven Cancer InstituteUniversity of LeuvenLeuvenBelgium
| | - Johannes V. Swinnen
- Department of Oncology, Laboratory of Lipid Metabolism and Cancer, Leuven Cancer InstituteUniversity of LeuvenLeuvenBelgium
| | - Sanne G. S. Verberk
- Department of Immunology and Infection, Biomedical Research InstituteHasselt UniversityDiepenbeekBelgium
- University MS Center HasseltPeltBelgium
| | - Mansour Haidar
- Department of Immunology and Infection, Biomedical Research InstituteHasselt UniversityDiepenbeekBelgium
- University MS Center HasseltPeltBelgium
| | - Jerome J. A. Hendriks
- Department of Immunology and Infection, Biomedical Research InstituteHasselt UniversityDiepenbeekBelgium
- University MS Center HasseltPeltBelgium
| | - Jeroen F. J. Bogie
- Department of Immunology and Infection, Biomedical Research InstituteHasselt UniversityDiepenbeekBelgium
- University MS Center HasseltPeltBelgium
| |
Collapse
|
40
|
Gao Y, Ye S, Tang Y, Tong W, Sun S. Brain cholesterol homeostasis and its association with neurodegenerative diseases. Neurochem Int 2023; 171:105635. [PMID: 37949118 DOI: 10.1016/j.neuint.2023.105635] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/17/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023]
Abstract
The brain is the most cholesterol-rich organ in mammals. However, cholesterol metabolism in the brain is completely independent of other tissues due to the presence of the blood-brain barrier (BBB). Neurons, astrocytes and oligodendrocytes are the main cells responsible for cholesterol synthesis in the brain. The cholesterol content in the brain is maintained at a relatively constant level under strict regulation of synthesis, transport, and turnover, that is, brain cholesterol homeostasis. Once this balance is disrupted, neurodegenerative diseases including Alzheimer's disease (AD), Parkinson's disease (PD) and Huntington's disease (HD) ensue. This review summarizes the processes controlling cholesterol homeostasis with respect to the synthesis, transport and turnover of cholesterol in the brain. We further focus on how cholesterol imbalance contributes to neurodegenerative diseases to explore the possibilities to modulate the key steps involved, which will provide clues for the development of therapies for the treatment of central nervous system (CNS) diseases.
Collapse
Affiliation(s)
- Yi Gao
- Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Shiying Ye
- Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yuehong Tang
- Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Wenjuan Tong
- Department of Gynecology and Obstetrics, First Affiliated Hospital, University of South China, Hengyang, Hunan, 421001, China.
| | - Shaowei Sun
- Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China.
| |
Collapse
|
41
|
Harnisch LO, Neugebauer S, Mihaylov D, Eidizadeh A, Zechmeister B, Maier I, Moerer O. Quantification of Bile Acids in Cerebrospinal Fluid: Results of an Observational Trial. Biomedicines 2023; 11:2947. [PMID: 38001948 PMCID: PMC10669160 DOI: 10.3390/biomedicines11112947] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 10/26/2023] [Accepted: 10/30/2023] [Indexed: 11/26/2023] Open
Abstract
(1) Background: Bile acids, known as aids in intestinal fat digestion and as messenger molecules in serum, can be detected in cerebrospinal fluid (CSF), although the blood-brain barrier is generally an insurmountable obstacle for bile acids. The exact mechanisms of the occurrence, as well as possible functions of bile acids in the central nervous system, are not precisely understood. (2) Methods: We conducted a single-center observational trial. The concentrations of 15 individual bile acids were determined using an in-house LC-MS/MS method in 54 patients with various acute and severe disorders of the central nervous system. We analyzed CSF from ventricular drainage taken within 24 h after placement, and blood samples were drawn at the same time for the presence and quantifiability of 15 individual bile acids. (3) Results: At a median time of 19.75 h after a cerebral insult, the concentration of bile acids in the CSF was minute and almost negligible. The CSF concentrations of total bile acids (TBAs) were significantly lower compared to the serum concentrations (serum 0.37 µmol/L [0.24, 0.89] vs. 0.14 µmol/L [0.05, 0.43]; p = 0.033). The ratio of serum-to-CSF bile acid levels calculated from the respective total concentrations were 3.10 [0.94, 14.64] for total bile acids, 3.05 for taurocholic acid, 14.30 [1.11, 27.13] for glycocholic acid, 0.0 for chenodeoxycholic acid, 2.19 for taurochenodeoxycholic acid, 1.91 [0.68, 8.64] for glycochenodeoxycholic acid and 0.77 [0.0, 13.79] for deoxycholic acid; other bile acids were not detected in the CSF. The ratio of CSF-to-serum S100 concentration was 0.01 [0.0, 0.02]. Serum total and conjugated (but not unconjugated) bilirubin levels and serum TBA levels were significantly correlated (total bilirubin p = 0.031 [0.023, 0.579]; conjugated bilirubin p = 0.001 [0.193, 0.683]; unconjugated p = 0.387 [-0.181, 0.426]). No correlations were found between bile acid concentrations and age, delirium, intraventricular blood volume, or outcome measured on a modified Rankin scale. (4) Conclusions: The determination of individual bile acids is feasible using the current LC-MS/MS method. The results suggest an intact blood-brain barrier in the patients studied. However, bile acids were detected in the CSF, which could have been achieved by active transport across the blood-brain barrier.
Collapse
Affiliation(s)
- Lars-Olav Harnisch
- Department of Anaesthesiology, University Medical Center, University of Göttingen, Robert-Koch-Str. 40, D-37075 Göttingen, Germany;
| | - Sophie Neugebauer
- Institute of Clinical Chemistry and Laboratory Diagnostics, University Hospital Jena, Am Klinikum 1, D-07747 Jena, Germany; (S.N.); (D.M.)
| | - Diana Mihaylov
- Institute of Clinical Chemistry and Laboratory Diagnostics, University Hospital Jena, Am Klinikum 1, D-07747 Jena, Germany; (S.N.); (D.M.)
| | - Abass Eidizadeh
- Interdisciplinary UMG Laboratories, University Medical Center, University of Göttingen, Robert-Koch-Str. 40, D-37075 Göttingen, Germany; (A.E.); (B.Z.)
| | - Bozena Zechmeister
- Interdisciplinary UMG Laboratories, University Medical Center, University of Göttingen, Robert-Koch-Str. 40, D-37075 Göttingen, Germany; (A.E.); (B.Z.)
| | - Ilko Maier
- Department of Neurology, University Medical Center, University of Göttingen, Robert-Koch-Str. 40, D-37075 Göttingen, Germany;
| | - Onnen Moerer
- Department of Anaesthesiology, University Medical Center, University of Göttingen, Robert-Koch-Str. 40, D-37075 Göttingen, Germany;
| |
Collapse
|
42
|
Zakirjanova GF, Giniatullin AR, Gafurova CR, Malomouzh AI, Fedorov NS, Khaziev AN, Tsentsevitsky AN, Petrov AM. Effects of cholesterol oxidase on neurotransmission and acetylcholine levels at the mice neuromuscular junctions. Arch Biochem Biophys 2023; 749:109803. [PMID: 37955112 DOI: 10.1016/j.abb.2023.109803] [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: 07/17/2023] [Revised: 09/20/2023] [Accepted: 10/27/2023] [Indexed: 11/14/2023]
Abstract
Membrane cholesterol oxidation is a hallmark of redox and metabolic imbalance, and it may accompany neurodegenerative disorders. Using microelectrode recordings of postsynaptic responses as well as fluorescent dyes for monitoring synaptic vesicle cycling and membrane properties, the action of enzymatic cholesterol oxidation on neuromuscular transmission was studied in the mice diaphragms. Cholesterol oxidase (ChO) at low concentration disturbed lipid-ordering specifically in the synaptic membranes, but it did not change markedly spontaneous exocytosis and evoked release in response to single stimuli. At low external Ca2+ conditions, analysis of single exocytotic events revealed a decrease in minimal synaptic delay and the probability of exocytosis upon plasmalemmal cholesterol oxidation. At moderate- and high-frequency activity, ChO treatment enhanced both neurotransmitter and FM-dye release. Furthermore, it precluded a change in exocytotic mode from full-fusion to kiss-and-run during high-frequency stimulation. Accumulation of extracellular acetylcholine (without stimulation) dependent on vesamicol-sensitive transporters was suppressed by ChO. The effects of plasmalemmal cholesterol oxidation on both neurotransmitter/dye release at intense activity and external acetylcholine levels were reversed when synaptic vesicle membranes were also exposed to ChO (i.e., the enzyme treatment was combined with induction of exo-endocytotic cycling). Thus, we suggest that plasmalemmal cholesterol oxidation affects exocytotic machinery functioning, enhances synaptic vesicle recruitment to the exocytosis and decreases extracellular neurotransmitter levels at rest, whereas ChO acting on synaptic vesicle membranes suppresses the participation of the vesicles in the subsequent exocytosis and increases the neurotransmitter leakage. The mechanisms underlying ChO action can be related to the lipid raft disruption.
Collapse
Affiliation(s)
- Guzalia F Zakirjanova
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center of RAS, 2/31 Lobachevsky St, Kazan, 420111, RT, Russia; Kazan State Medical University, 49 Butlerova St., Kazan, 420012, RT, Russia
| | - Arthur R Giniatullin
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center of RAS, 2/31 Lobachevsky St, Kazan, 420111, RT, Russia; Kazan State Medical University, 49 Butlerova St., Kazan, 420012, RT, Russia
| | - Chulpan R Gafurova
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center of RAS, 2/31 Lobachevsky St, Kazan, 420111, RT, Russia; Kazan State Medical University, 49 Butlerova St., Kazan, 420012, RT, Russia
| | - Artem I Malomouzh
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center of RAS, 2/31 Lobachevsky St, Kazan, 420111, RT, Russia; Kazan National Research Technical University, 10, K. Marx Street, Kazan, 420111, Russia
| | - Nikita S Fedorov
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center of RAS, 2/31 Lobachevsky St, Kazan, 420111, RT, Russia
| | - Arthur N Khaziev
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center of RAS, 2/31 Lobachevsky St, Kazan, 420111, RT, Russia
| | - Andrei N Tsentsevitsky
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center of RAS, 2/31 Lobachevsky St, Kazan, 420111, RT, Russia
| | - Alexey M Petrov
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center of RAS, 2/31 Lobachevsky St, Kazan, 420111, RT, Russia; Kazan State Medical University, 49 Butlerova St., Kazan, 420012, RT, Russia; Kazan Federal University, 18 Kremlyovskaya Street, Kazan, 420008, Russia.
| |
Collapse
|
43
|
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
|
44
|
Benachenhou S, Laroui A, Dionne O, Rojas D, Toupin A, Çaku A. Cholesterol alterations in fragile X syndrome, autism spectrum disorders and other neurodevelopmental disorders. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 173:115-139. [PMID: 37993175 DOI: 10.1016/bs.irn.2023.08.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Abstract
Neurodevelopmental disorders (NDDs) are a group of etiologically diverse diseases primarily associated with abnormal brain development, impaired cognition, and various behavioral problems. The majority of NDDs present a wide range of clinical phenotypes while sharing distinct cellular and biochemical alterations. Low plasma cholesterol levels have been reported in a subset of NNDs including, autism spectrum disorder (ASD) and fragile X syndrome (FXS). The present review focuses on cholesterol metabolism and discusses the current evidence of lipid disruption in ASD, FXS, and other genetically related NDDs. The characterization of these common deficits might provide valuable insights into their underlying physiopathology and help identify potential therapeutic targets.
Collapse
Affiliation(s)
- Sérine Benachenhou
- Biochemistry and Functional Genomic Department, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Asma Laroui
- Biochemistry and Functional Genomic Department, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Olivier Dionne
- Biochemistry and Functional Genomic Department, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Daniela Rojas
- Biochemistry and Functional Genomic Department, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Amanda Toupin
- Biochemistry and Functional Genomic Department, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Artuela Çaku
- Biochemistry and Functional Genomic Department, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada.
| |
Collapse
|
45
|
Rudajev V, Novotny J. Cholesterol-dependent amyloid β production: space for multifarious interactions between amyloid precursor protein, secretases, and cholesterol. Cell Biosci 2023; 13:171. [PMID: 37705117 PMCID: PMC10500844 DOI: 10.1186/s13578-023-01127-y] [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: 05/17/2023] [Accepted: 09/05/2023] [Indexed: 09/15/2023] Open
Abstract
Amyloid β is considered a key player in the development and progression of Alzheimer's disease (AD). Many studies investigating the effect of statins on lowering cholesterol suggest that there may be a link between cholesterol levels and AD pathology. Since cholesterol is one of the most abundant lipid molecules, especially in brain tissue, it affects most membrane-related processes, including the formation of the most dangerous form of amyloid β, Aβ42. The entire Aβ production system, which includes the amyloid precursor protein (APP), β-secretase, and the complex of γ-secretase, is highly dependent on membrane cholesterol content. Moreover, cholesterol can affect amyloidogenesis in many ways. Cholesterol influences the stability and activity of secretases, but also dictates their partitioning into specific cellular compartments and cholesterol-enriched lipid rafts, where the amyloidogenic machinery is predominantly localized. The most complicated relationships have been found in the interaction between cholesterol and APP, where cholesterol affects not only APP localization but also the precise character of APP dimerization and APP processing by γ-secretase, which is important for the production of Aβ of different lengths. In this review, we describe the intricate web of interdependence between cellular cholesterol levels, cholesterol membrane distribution, and cholesterol-dependent production of Aβ, the major player in AD.
Collapse
Affiliation(s)
- Vladimir Rudajev
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Jiri Novotny
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| |
Collapse
|
46
|
Jaafar AK, Techer R, Chemello K, Lambert G, Bourane S. PCSK9 and the nervous system: a no-brainer? J Lipid Res 2023; 64:100426. [PMID: 37586604 PMCID: PMC10491654 DOI: 10.1016/j.jlr.2023.100426] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/08/2023] [Accepted: 08/10/2023] [Indexed: 08/18/2023] Open
Abstract
In the past 20 years, PCSK9 has been shown to play a pivotal role in LDL cholesterol metabolism and cardiovascular health by inducing the lysosomal degradation of the LDL receptor. PCSK9 was discovered by the cloning of genes up-regulated after apoptosis induced by serum deprivation in primary cerebellar neurons, but despite its initial identification in the brain, the precise role of PCSK9 in the nervous system remains to be clearly established. The present article is a comprehensive review of studies published or in print before July 2023 that have investigated the expression pattern of PCSK9, its effects on lipid metabolism as well as its putative roles specifically in the central and peripheral nervous systems, with a special focus on cerebrovascular and neurodegenerative diseases.
Collapse
Affiliation(s)
- Ali K Jaafar
- Laboratoire Inserm UMR 1188 DéTROI, Saint-Pierre, La Réunion, France
| | - Romuald Techer
- Laboratoire Inserm UMR 1188 DéTROI, Saint-Pierre, La Réunion, France
| | - Kévin Chemello
- Laboratoire Inserm UMR 1188 DéTROI, Saint-Pierre, La Réunion, France
| | - Gilles Lambert
- Laboratoire Inserm UMR 1188 DéTROI, Saint-Pierre, La Réunion, France; Faculté de Médecine, Université de La Réunion, Saint-Pierre, La Réunion, France.
| | - Steeve Bourane
- Laboratoire Inserm UMR 1188 DéTROI, Saint-Pierre, La Réunion, France
| |
Collapse
|
47
|
Hansen SB, Wang H. The shared role of cholesterol in neuronal and peripheral inflammation. Pharmacol Ther 2023; 249:108486. [PMID: 37390970 DOI: 10.1016/j.pharmthera.2023.108486] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 06/22/2023] [Accepted: 06/27/2023] [Indexed: 07/02/2023]
Abstract
Neurodegeneration and its loss of cognitive function is associated with inflammation and an accumulation of lipids. In the periphery, cholesterol's uptake drives a major component of chronic inflammation. In this perspective, we describe the cellular and molecular roles of cholesterol in neuroinflammation and contrast them with those in the periphery. Incorporating shared mechanisms from the periphery, cholesterol emerges as a central signal originating in astrocytes and connecting inflammatory escalation in neurons and microglia. A cholesterol uptake pathway is proposed for neuroinflammation, and we speculate on the binding of cholesterol transport protein apolipoprotein E (apoE), including the Christchurch mutant (R136S), to cell surface receptors as a potential protective modality against uptake of astrocyte cholesterol and escalated neuroinflammation. Lastly, we discuss the molecular basis of cholesterol signaling through nanoscopic clustering and peripheral sources of cholesterol after opening of the blood brain barrier.
Collapse
Affiliation(s)
- Scott B Hansen
- Department of Molecular Medicine, UF Scripps, Jupiter, FL 33458, USA; Department of Neuroscience, UF Scripps, Jupiter, FL 33458, USA.
| | - Hao Wang
- The Scripps Research Institute, Jupiter, FL 33458, USA
| |
Collapse
|
48
|
Yuan R, Zhang W, You Y, Cui G, Gao Z, Wang X, Chen J. Vitamin D3 suppresses the cholesterol homeostasis pathway in patient-derived glioma cell lines. FEBS Open Bio 2023; 13:1789-1806. [PMID: 37489660 PMCID: PMC10476568 DOI: 10.1002/2211-5463.13679] [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: 02/03/2023] [Revised: 05/19/2023] [Accepted: 07/24/2023] [Indexed: 07/26/2023] Open
Abstract
Glioblastoma is one of the most common malignant brain tumors. Vitamin D, primarily its hormonally active form calcitriol, has been reported to have anti-cancer activity. In the present study, we used patient-derived glioma cell lines to examine the effect of vitamin D3 and calcitriol on glioblastoma. Surprisingly, vitamin D3 showed a more significant inhibitory effect than calcitriol on cell viability and proliferation. Vitamin D receptor (VDR) mediates most of the cellular effects of vitamin D, and thus we examined the expression level and function of VDR via gene silencing and gene knockout experiments. We observed that VDR does not affect the sensitivity of patient-derived glioma cell lines to vitamin D3, and the gene encoding VDR is not essential for growth of patient-derived glioma cell lines. RNA sequencing data analysis and sterolomics analysis revealed that vitamin D3 inhibits cholesterol synthesis and cholesterol homeostasis by inhibiting the expression level of 7-dehydrocholesterol reductase, which leads to the accumulation of 7-dehydrocholesterol and other sterol intermediates. In conclusion, our results suggest that vitamin D3, rather than calcitriol, inhibits growth of patient-derived glioma cell lines via inhibition of the cholesterol homeostasis pathway.
Collapse
Affiliation(s)
- Ran Yuan
- Institute of Functional Nano & Soft Materials (FUNSOM)Soochow UniversitySuzhouChina
- Chinese Institute for Brain ResearchBeijingChina
- Research Unit of Medical NeurobiologyChinese Academy of Medical SciencesBeijingChina
| | - Wei Zhang
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Neuropathology, Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
| | - Yong‐Ping You
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Institute for Brain Tumors, Jiangsu Collaborative Innovation Center for Cancer Personalized MedicineNanjing Medical UniversityChina
| | - Gang Cui
- Department of Neurosurgery & Brain and Nerve Research LaboratoryThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Zhengliang Gao
- Fundamental Research Center, Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of MedicineTongji UniversityShanghaiChina
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of MedicineShanghai UniversityNantongChina
| | - Xiuxing Wang
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical SciencesNanjing Medical UniversityChina
| | - Jian Chen
- Institute of Functional Nano & Soft Materials (FUNSOM)Soochow UniversitySuzhouChina
- Chinese Institute for Brain ResearchBeijingChina
- Research Unit of Medical NeurobiologyChinese Academy of Medical SciencesBeijingChina
| |
Collapse
|
49
|
Guardia-Escote L, Biosca-Brull J, Cabré M, Blanco J, Mladenova-Koleva M, Basaure P, Pérez-Fernández C, Sánchez-Santed F, Domingo JL, Colomina MT. Developmental brain lipidomics is influenced by postnatal chlorpyrifos exposure and APOE genetic background in mice. Arch Toxicol 2023; 97:2463-2475. [PMID: 37439814 PMCID: PMC10404178 DOI: 10.1007/s00204-023-03555-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 07/03/2023] [Indexed: 07/14/2023]
Abstract
Lipids are a major component of the brain, and are involved in structural and neurodevelopmental processes such as neurogenesis, synaptogenesis and signaling. Apolipoprotein E (apoE) is the main lipoprotein involved in lipid transport in the brain. The apoE isoforms can determine vulnerability to the toxic effects of the pesticide chlorpyrifos (CPF), which can interfere with normal neurodevelopment. We aimed to study the effects of postnatal exposure to CPF and of the APOE genotype on the lipid composition of the brain at early ages. For it, we used apoE3 and apoE4 targeted-replacement (TR) male mice, as well as wild-type C57BL/6. The mice were orally exposed to 1 mg/kg/day of CPF on postnatal days 10-15 and, four hours after the treatment, we obtained samples to assess the cerebral lipid composition. Differences between APOE genotypes were found in the cerebral lipid profile in the postnatal period. ApoE4-TR mice exhibited higher lipid concentrations compared to the other groups in most of the cases. CPF exposure led to a decrease in cholesteryl ester and triglyceride concentrations, while modulating the levels of phosphatidylcholine species based on the apoE isoform. Specifically, CPF treatment decreased the concentration of some species of this lipid (PC30:0, PC31:0, PC32:2, PC36:5, PC40:4 and PC40:5) in C57BL/6 mice exposed to CPF, increased (PC31:0 and PC37:6) in apoE3-TR exposed mice while exposed apoE4-TR mice remained unaltered. These results provide further insights into the lipid composition of the brain at early ages, and how it can be modulated by environmental and genetic factors.
Collapse
Affiliation(s)
- Laia Guardia-Escote
- Research Group in Neurobehavior and Health (NEUROLAB), Universitat Rovira i Virgili, Tarragona, Spain
- Department of Psychology and Research Center for Behavior Assessment (CRAMC), Universitat Rovira i Virgili, Tarragona, Spain
| | - Judit Biosca-Brull
- Research Group in Neurobehavior and Health (NEUROLAB), Universitat Rovira i Virgili, Tarragona, Spain
- Department of Psychology and Research Center for Behavior Assessment (CRAMC), Universitat Rovira i Virgili, Tarragona, Spain
- Laboratory of Toxicology and Environmental Health (TECNATOX), Universitat Rovira i Virgili, Reus, Spain
| | - Maria Cabré
- Research Group in Neurobehavior and Health (NEUROLAB), Universitat Rovira i Virgili, Tarragona, Spain
- Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, Tarragona, Spain
| | - Jordi Blanco
- Research Group in Neurobehavior and Health (NEUROLAB), Universitat Rovira i Virgili, Tarragona, Spain
- Laboratory of Toxicology and Environmental Health (TECNATOX), Universitat Rovira i Virgili, Reus, Spain
- Department of Basic Medical Sciences, Universitat Rovira i Virgili, Reus, Spain
| | - Mikaela Mladenova-Koleva
- Research Group in Neurobehavior and Health (NEUROLAB), Universitat Rovira i Virgili, Tarragona, Spain
| | - Pia Basaure
- Research Group in Neurobehavior and Health (NEUROLAB), Universitat Rovira i Virgili, Tarragona, Spain
| | | | - Fernando Sánchez-Santed
- Department of Psychology, Health Research Center (CEINSA), Almería University, Almería, Spain
| | - José L Domingo
- Laboratory of Toxicology and Environmental Health (TECNATOX), Universitat Rovira i Virgili, Reus, Spain
| | - Maria Teresa Colomina
- Research Group in Neurobehavior and Health (NEUROLAB), Universitat Rovira i Virgili, Tarragona, Spain.
- Department of Psychology and Research Center for Behavior Assessment (CRAMC), Universitat Rovira i Virgili, Tarragona, Spain.
- Laboratory of Toxicology and Environmental Health (TECNATOX), Universitat Rovira i Virgili, Reus, Spain.
| |
Collapse
|
50
|
Sodero AO, Castagna VC, Elorza SD, Gonzalez-Rodulfo SM, Paulazo MA, Ballestero JA, Martin MG, Gomez-Casati ME. Phytosterols reverse antiretroviral-induced hearing loss, with potential implications for cochlear aging. PLoS Biol 2023; 21:e3002257. [PMID: 37619212 PMCID: PMC10449472 DOI: 10.1371/journal.pbio.3002257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 07/18/2023] [Indexed: 08/26/2023] Open
Abstract
Cholesterol contributes to neuronal membrane integrity, supports membrane protein clustering and function, and facilitates proper signal transduction. Extensive evidence has shown that cholesterol imbalances in the central nervous system occur in aging and in the development of neurodegenerative diseases. In this work, we characterize cholesterol homeostasis in the inner ear of young and aged mice as a new unexplored possibility for the prevention and treatment of hearing loss. Our results show that cholesterol levels in the inner ear are reduced during aging, an effect that is associated with an increased expression of the cholesterol 24-hydroxylase (CYP46A1), the main enzyme responsible for cholesterol turnover in the brain. In addition, we show that pharmacological activation of CYP46A1 with the antiretroviral drug efavirenz reduces the cholesterol content in outer hair cells (OHCs), leading to a decrease in prestin immunolabeling and resulting in an increase in the distortion product otoacoustic emissions (DPOAEs) thresholds. Moreover, dietary supplementation with phytosterols, plant sterols with structure and function similar to cholesterol, was able to rescue the effect of efavirenz administration on the auditory function. Altogether, our findings point towards the importance of cholesterol homeostasis in the inner ear as an innovative therapeutic strategy in preventing and/or delaying hearing loss.
Collapse
Affiliation(s)
- Alejandro O. Sodero
- Instituto de Investigaciones Biomédicas, Pontificia Universidad Católica Argentina, Consejo Nacional de Investigaciones Científicas y Técnicas (BIOMED, UCA-CONICET), Buenos Aires, Argentina
| | - Valeria C. Castagna
- Instituto de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Dr. Héctor N. Torres, Consejo Nacional de Investigaciones Científicas y Técnicas (INGEBI-CONICET), Buenos Aires, Argentina
| | - Setiembre D. Elorza
- Laboratorio de Neurobiología, Instituto de Investigaciones Médicas Mercedes y Martín Ferreyra, Consejo Nacional de Investigaciones Científicas y Técnicas (INIMEC-CONICET-UNC), Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Sara M. Gonzalez-Rodulfo
- Instituto de Investigaciones Biomédicas, Pontificia Universidad Católica Argentina, Consejo Nacional de Investigaciones Científicas y Técnicas (BIOMED, UCA-CONICET), Buenos Aires, Argentina
| | - María A. Paulazo
- Instituto de Investigaciones Biomédicas, Pontificia Universidad Católica Argentina, Consejo Nacional de Investigaciones Científicas y Técnicas (BIOMED, UCA-CONICET), Buenos Aires, Argentina
| | - Jimena A. Ballestero
- Instituto de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Mauricio G. Martin
- Laboratorio de Neurobiología, Instituto de Investigaciones Médicas Mercedes y Martín Ferreyra, Consejo Nacional de Investigaciones Científicas y Técnicas (INIMEC-CONICET-UNC), Universidad Nacional de Córdoba, Córdoba, Argentina
| | - María Eugenia Gomez-Casati
- Instituto de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|