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
|
Herrmann C, Uzelac Z, Michels S, Weber A, Richter L, Elmas Z, Jagodzinski L, Wurster C, Schuster J, Dreyhaupt J, Dorst J. Alterations of Fat and Ketone Body Metabolism in ALS and SMA-A Prospective Observational Study. Eur J Neurol 2025; 32:e70132. [PMID: 40200760 PMCID: PMC11979353 DOI: 10.1111/ene.70132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 03/02/2025] [Accepted: 03/24/2025] [Indexed: 04/10/2025]
Abstract
BACKGROUND Amyotrophic lateral sclerdosis (ALS) and spinal muscular atrophy (SMA) are motor neuron diseases associated with distinct metabolic alterations. ALS patients feature an increased resting energy expenditure (REE) causing weight loss and cachexia. In SMA, a disturbed utilization of free fatty acids has been described. These metabolic alterations negatively affect prognosis in both diseases. The objective of this study was to further characterize these changes to identify potential therapeutic targets. METHODS Between 11/2020 and 08/2022, 112 ALS patients, 77 SMA patients, and 50 controls were recruited in the Department of Neurology of Ulm University. Standardized blood and urinary samples were collected to analyze fat and ketone metabolism. RESULTS Ketone body levels were higher in ALS and SMA compared to controls. In both diseases, patients with higher BMI featured higher ketone bodies and free fatty acids compared to those with lower BMI, while in controls we found the opposite phenomenon. In SMA, more severe disease types were associated with higher ketone body levels. Compared to ALS, SMA patients featured higher ketone body and free fatty acid levels. CONCLUSIONS Our data suggest that already during early disease stages, ALS patients produce ketone bodies to compensate for the energy deficit. In SMA, on the other hand, the persistence of ketogenesis may indicate an upregulation of all available metabolic pathways for energy production due to the disturbance of fatty acid utilization. Therefore, the application of additional sources of energy, such as ketone bodies, might constitute a promising therapeutic option in both diseases.
Collapse
Affiliation(s)
- C. Herrmann
- Department of NeurologyUniversity of UlmUlmGermany
| | - Z. Uzelac
- Department of NeurologyUniversity of UlmUlmGermany
| | - S. Michels
- Department of NeurologyUniversity of UlmUlmGermany
| | - A. Weber
- Department of NeurologyUniversity of UlmUlmGermany
| | - L. Richter
- Department of NeurologyUniversity of UlmUlmGermany
| | - Z. Elmas
- Department of NeurologyUniversity of UlmUlmGermany
| | | | - C. Wurster
- Department of NeurologyUniversity of UlmUlmGermany
| | - J. Schuster
- Department of NeurologyUniversity of UlmUlmGermany
| | - J. Dreyhaupt
- Institute for Epidemiology and Medical BiometryUlmGermany
| | - J. Dorst
- Department of NeurologyUniversity of UlmUlmGermany
- German Center for Neurodegenerative Diseases (DZNE)UlmGermany
| |
Collapse
|
3
|
Majewski S, Klein P, Boillée S, Clarke BE, Patani R. Towards an integrated approach for understanding glia in Amyotrophic Lateral Sclerosis. Glia 2025; 73:591-607. [PMID: 39318236 PMCID: PMC11784848 DOI: 10.1002/glia.24622] [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: 04/17/2024] [Revised: 09/03/2024] [Accepted: 09/15/2024] [Indexed: 09/26/2024]
Abstract
Substantial advances in technology are permitting a high resolution understanding of the salience of glia, and have helped us to transcend decades of predominantly neuron-centric research. In particular, recent advances in 'omic' technologies have enabled unique insights into glial biology, shedding light on the cellular and molecular aspects of neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). Here, we review studies using omic techniques to attempt to understand the role of glia in ALS across different model systems and post mortem tissue. We also address caveats that should be considered when interpreting such studies, and how some of these may be mitigated through either using a multi-omic approach and/or careful low throughput, high fidelity orthogonal validation with particular emphasis on functional validation. Finally, we consider emerging technologies and their potential relevance in deepening our understanding of glia in ALS.
Collapse
Affiliation(s)
- Stanislaw Majewski
- Department of Neuromuscular Diseases, Queen Square Institute of NeurologyUniversity College LondonLondonUK
- The Francis Crick InstituteLondonUK
| | - Pierre Klein
- Department of Neuromuscular Diseases, Queen Square Institute of NeurologyUniversity College LondonLondonUK
- The Francis Crick InstituteLondonUK
| | - Séverine Boillée
- Sorbonne Université, Institut du Cerveau—Paris Brain Institute—ICM, Inserm, CNRS, APHPHôpital de la Pitié‐SalpêtrièreParisFrance
| | - Benjamin E. Clarke
- Department of Neuromuscular Diseases, Queen Square Institute of NeurologyUniversity College LondonLondonUK
- The Francis Crick InstituteLondonUK
| | - Rickie Patani
- Department of Neuromuscular Diseases, Queen Square Institute of NeurologyUniversity College LondonLondonUK
- The Francis Crick InstituteLondonUK
| |
Collapse
|
4
|
Wang Z, Yin Z, Sun G, Zhang D, Zhang J. Genetic evidence for the liver-brain axis: lipid metabolism and neurodegenerative disease risk. Lipids Health Dis 2025; 24:41. [PMID: 39923073 PMCID: PMC11806572 DOI: 10.1186/s12944-025-02455-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 01/29/2025] [Indexed: 02/10/2025] Open
Abstract
BACKGROUND The liver‒brain axis is critical in neurodegenerative diseases (NDs), with lipid metabolism influencing neuroinflammation and microglial function. A systematic investigation of the genetic relationship between lipid metabolism abnormalities and ND, namely, Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), and amyotrophic lateral sclerosis (ALS), is lacking. To assess potential causal links between ND and six lipid parameters, two-sample Mendelian randomization (MR) was used. METHODS Large-scale European ancestry GWAS data for lipid parameters and ND (AD, ALS, PD, and MS) were used. Genetic variants demonstrating significant correlations (P < 5 × 10-8) with lipid metabolism parameters were identified and employed as instrumental variables (IVs) after proper validation. The research incorporated UK Biobank genomic data to examine associations between genetic variants and lipid metabolism parameters. The analysis included primary MR, sensitivity analyses, and multivariable MR, which considered potential mediators. RESULTS MR via the inverse-variance weighted method revealed causal effects of cholesterol (CHOL, OR = 1.10, 95% CI: 1.03-1.18, P = 4.23 × 10⁻3) and low-density lipoprotein cholesterol (LDLC, OR = 1.10, 95% CI: 1.03-1.17, P = 3.28 × 10⁻3) on the risk of ALS, which were validated across multiple methods. Potential correlations were observed between ApoB and ALS and inversely correlated with AD, whereas no significant associations were found for PD or MS. CHOL and LDLC associations with ALS demonstrated no significant heterogeneity or pleiotropy, supporting their reliability. CONCLUSIONS Higher CHOL and LDLC levels were associated with increased ALS risk, suggesting a potential causal link, and supporting the liver‒brain axis hypothesis in ND. Current genetic evidence does not support a significant role for lipid metabolism in PD and MS etiology, suggesting the relationship between lipid metabolism and other NDs may be more complex and warrants further investigation.
Collapse
Affiliation(s)
- Zeyu Wang
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Zixiao Yin
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Neurostimulation, Beijing, 100070, China
| | - Guangyong Sun
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Dong Zhang
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China.
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China.
| | - Jianguo Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Neurostimulation, Beijing, 100070, China.
| |
Collapse
|
5
|
Horiuchi M, Watanabe S, Komine O, Takahashi E, Kaneko K, Itohara S, Shimada M, Ogi T, Yamanaka K. ALS-linked mutant TDP-43 in oligodendrocytes induces oligodendrocyte damage and exacerbates motor dysfunction in mice. Acta Neuropathol Commun 2024; 12:184. [PMID: 39605053 PMCID: PMC11603663 DOI: 10.1186/s40478-024-01893-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: 07/01/2024] [Accepted: 11/17/2024] [Indexed: 11/29/2024] Open
Abstract
Nuclear clearance and cytoplasmic aggregation of TAR DNA-binding protein of 43 kDa (TDP-43) are pathological hallmarks of amyotrophic lateral sclerosis (ALS) and its pathogenic mechanism is mediated by both loss-of-function and gain-of-toxicity of TDP-43. However, the role of TDP-43 gain-of-toxicity in oligodendrocytes remains unclear. To investigate the impact of excess TDP-43 on oligodendrocytes, we established transgenic mice overexpressing the ALS-linked mutant TDP-43M337V in oligodendrocytes through crossbreeding with Mbp-Cre mice. Two-step crossbreeding of floxed TDP-43M337V and Mbp-Cre mice resulted in the heterozygous low-level systemic expression of TDP-43M337V with (Cre-positive) or without (Cre-negative) oligodendrocyte-specific overexpression of TDP-43M337V. Although Cre-negative mice also exhibit subtle motor dysfunction, TDP-43M337V overexpression in oligodendrocytes aggravated clasping signs and gait disturbance accompanied by myelin pallor in the corpus callosum and white matter of the lumbar spinal cord in Cre-positive mice. RNA sequencing analysis of oligodendrocyte lineage cells isolated from whole brains of 12-month-old transgenic mice revealed downregulation of myelinating oligodendrocyte marker genes and cholesterol-related genes crucial for myelination, along with marked upregulation of apoptotic pathway genes. Immunofluorescence staining showed cleaved caspase 3-positive apoptotic oligodendrocytes surrounded by activated microglia and astrocytes in aged transgenic mice. Collectively, our findings demonstrate that an excess amount of ALS-linked mutant TDP-43 expression in oligodendrocytes exacerbates motor dysfunction in mice, likely through oligodendrocyte dysfunction and neuroinflammation. Therefore, targeting oligodendrocyte protection, particularly through ameliorating TDP-43 pathology, could represent a potential therapeutic approach for ALS.
Collapse
Affiliation(s)
- Mai Horiuchi
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Chikusa-Ku, Nagoya, Aichi, 464-8601, Japan
- Department of Neuroscience and Pathobiology, Graduate School of Medicine, Nagoya University, Nagoya, Aichi, 466-8550, Japan
| | - Seiji Watanabe
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Chikusa-Ku, Nagoya, Aichi, 464-8601, Japan
- Department of Neuroscience and Pathobiology, Graduate School of Medicine, Nagoya University, Nagoya, Aichi, 466-8550, Japan
| | - Okiru Komine
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Chikusa-Ku, Nagoya, Aichi, 464-8601, Japan
- Department of Neuroscience and Pathobiology, Graduate School of Medicine, Nagoya University, Nagoya, Aichi, 466-8550, Japan
| | - Eiki Takahashi
- Department of Biomedicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Kumi Kaneko
- Support Unit for Bio-Material Analysis, Research Resources Division, RIKEN Center for Brain Science, Saitama, 351-0198, Japan
| | - Shigeyoshi Itohara
- Laboratory of Behavioral Genetics, RIKEN Center for Brain Science, Saitama, 351-0198, Japan
| | - Mayuko Shimada
- Department of Genetics, Research Institute of Environmental Medicine, Nagoya University, Aichi, 464-8601, Japan
- Department of Human Genetics and Molecular Biology, Nagoya University Graduate School of Medicine, Aichi, 466-8550, Japan
| | - Tomoo Ogi
- Department of Genetics, Research Institute of Environmental Medicine, Nagoya University, Aichi, 464-8601, Japan
- Department of Human Genetics and Molecular Biology, Nagoya University Graduate School of Medicine, Aichi, 466-8550, Japan
| | - Koji Yamanaka
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Chikusa-Ku, Nagoya, Aichi, 464-8601, Japan.
- Department of Neuroscience and Pathobiology, Graduate School of Medicine, Nagoya University, Nagoya, Aichi, 466-8550, Japan.
- Institute for Glyco-Core Research (iGCORE), Nagoya University, Aichi, Japan.
- Center for One Medicine Innovative Translational Research (COMIT), Nagoya University, Aichi, Japan.
- Research Institute for Quantum and Chemical Innovation, Institutes of Innovation for Future Society, Nagoya University, Aichi, Japan.
| |
Collapse
|
6
|
Zhu L, Bai D, Wang X, Ou K, Li B, Jia Q, Tan Z, Liang J, He D, Yan S, Wang L, Li S, Li X, Yin P. Pathologic TDP-43 downregulates myelin gene expression in the monkey brain. Brain Pathol 2024; 34:e13277. [PMID: 38779803 PMCID: PMC11483520 DOI: 10.1111/bpa.13277] [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: 09/28/2023] [Accepted: 05/02/2024] [Indexed: 05/25/2024] Open
Abstract
Growing evidence indicates that non-neuronal oligodendrocyte plays an important role in Amyotrophic lateral sclerosis (ALS) and other neurodegenerative diseases. In patient's brain, the impaired myelin structure is a pathological feature with the observation of TDP-43 in cytoplasm of oligodendrocyte. However, the mechanism underlying the gain of function by TDP-43 in oligodendrocytes, which are vital for the axonal integrity, remains unclear. Recently, we found that the primate-specific cleavage of truncated TDP-43 fragments occurred in cytoplasm of monkey neural cells. This finding opened up the avenue to investigate the myelin integrity affected by pathogenic TDP-43 in oligodendrocytes. In current study, we demonstrated that the truncated TDP-35 in oligodendrocytes specifically, could lead to the dysfunctional demyelination in corpus callosum of monkey. As a consequence of the interaction of myelin regulatory factor with the accumulated TDP-35 in cytoplasm, the downstream myelin-associated genes expression was downregulated at the transcriptional level. Our study aims to investigate the potential effect on myelin structure injury, affected by the truncated TDP-43 in oligodendrocyte, which provided the additional clues on the gain of function during the progressive pathogenesis and symptoms in TDP-43 related diseases.
Collapse
Affiliation(s)
- Longhong Zhu
- Guangdong Key Laboratory of Non‐human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouChina
| | - Dazhang Bai
- Guangdong Key Laboratory of Non‐human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouChina
- Department of Neurology, Affiliated Hospital of North Sichuan Medical CollegeInstitute of Neurological Diseases, North Sichuan Medical CollegeNanchongChina
| | - Xiang Wang
- Guangdong Key Laboratory of Non‐human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouChina
| | - Kaili Ou
- Guangdong Key Laboratory of Non‐human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouChina
| | - Bang Li
- Guangdong Key Laboratory of Non‐human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouChina
| | - Qingqing Jia
- Guangdong Key Laboratory of Non‐human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouChina
| | - Zhiqiang Tan
- Department of Medical Imaging, First Affiliated HospitalJinan UniversityGuangzhouChina
| | - Jiahui Liang
- Department of Medical Imaging, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Dajian He
- Guangdong Key Laboratory of Non‐human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouChina
| | - Sen Yan
- Guangdong Key Laboratory of Non‐human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouChina
| | - Lu Wang
- Department of Medical Imaging, First Affiliated HospitalJinan UniversityGuangzhouChina
| | - Shihua Li
- Guangdong Key Laboratory of Non‐human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouChina
| | - Xiao‐Jiang Li
- Guangdong Key Laboratory of Non‐human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouChina
| | - Peng Yin
- Guangdong Key Laboratory of Non‐human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouChina
| |
Collapse
|
7
|
Jiang Z, Gu XJ, Su WM, Duan QQ, Yin KF, Ren YL, Wang Y, Cao B, Chen YP. Discovery and Exploration of Lipid-Modifying Drug Targets for ALS by Mendelian Randomization. Mol Neurobiol 2024; 61:6572-6583. [PMID: 38324182 DOI: 10.1007/s12035-024-04007-9] [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: 10/08/2023] [Accepted: 01/30/2024] [Indexed: 02/08/2024]
Abstract
Observational studies have faced challenges in identifying replicable causes for amyotrophic lateral sclerosis (ALS). To address this, we employed an unbiased and data-driven approach to discover and explore potential causal exposures using two-sample Mendelian randomization (MR) analyses. In the phenotype discovery stage, we assessed 3948 environmental exposures from the UK Biobank and utilized ALS summary statistics (Europeans, 20,806 cases, 59,804 controls) as the outcome within a phenome-wide MR pipeline. Through a range of sensitivity analyses, two medication traits were identified to be protective for ALS. In the target exploration stage, we further conducted drug target MR analyses using the latest and trans-ethnic summary data on lipid-related traits and ALS (Europeans, 27,205 cases, 110,881 controls; East Asians, 1234 cases, 2850 controls). Our aim was to explore potential causal drug targets through six lipid-modifying effects. These comprehensive analyses revealed significant findings. Specifically, "cholesterol-lowering medication" and "atorvastatin" survived predefined criteria in the phenotype discovery stage and exhibited a protective effect on ALS. Further in the target exploration stage, we demonstrated that the therapeutic effect of APOB through LDL-lowering was associated with reduced ALS liability in Europeans (OR = 0.835, P = 5.61E - 5). Additionally, the therapeutic effect of APOA1 and LDLR through TC-lowering was associated with reduced ALS liability in East Asians (APOA1, OR = 0.859, P = 5.38E - 4; LDLR, OR = 0.910, P = 2.73E - 5). Overall, we propose potential protective effects of cholesterol-lowering drugs or statins on ALS risk from thousands of exposures. Our research also suggests APOB, APOA1, and LDLR as novel therapeutic targets for ALS and supports their potential protective mechanisms may be mediated by LDL-lowering or TC-lowering effects.
Collapse
Affiliation(s)
- Zheng Jiang
- Department of Neurology, Centre for Rare Diseases, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xiao-Jing Gu
- Mental Health Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Wei-Ming Su
- Department of Neurology, Centre for Rare Diseases, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Qing-Qing Duan
- Department of Neurology, Centre for Rare Diseases, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Kang-Fu Yin
- Department of Neurology, Centre for Rare Diseases, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yan-Lin Ren
- Department of Pathophysiology, West China College of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yi Wang
- Department of Pathophysiology, West China College of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Bei Cao
- Department of Neurology, Centre for Rare Diseases, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yong-Ping Chen
- Department of Neurology, Centre for Rare Diseases, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
8
|
Rezaee Semnani M, Mirzaasgari Z, Ariaei A, Haghi Ashtiani B. Evaluation of carotid Intima-Media Thickness (IMT) in amyotrophic lateral sclerosis disease using ultrasonography. J Clin Neurosci 2024; 124:67-72. [PMID: 38657488 DOI: 10.1016/j.jocn.2024.04.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/22/2024] [Accepted: 04/19/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND Amyotrophic Lateral Sclerosis (ALS) is a progressive neurodegenerative disease with multi-mechanisms as; inflammation, oxidative stress, glutamate excitotoxicity, protein aggregation, etc. This study aimed to evaluate the carotid Intima-Media Thickness (IMT) in ALS and healthy groups, as a possible indicator of these mechanisms. METHODS 42 patients with ALS along with 53 normal age and body mass index (BMI) matched participants were recruited from the Firoozgar hospital. Carotid IMT values of the participants were measured using B-mode ultrasonography. Using Pearson correlation and logistic regression adjusting with age, BMI, and gender, the IMT values were assessed. RESULTS The mean right and left carotid IMT values of the ALS patients (0.66 ± 0.09) were significantly higher than normal participants (0.45 ± 0.10) (p < 0.001). In addition, the IMT values were highly correlated with the age (r = 0.632; p < 0.001) and the age of ALS onset (r = 0.595; p < 0.001), in contrast to the BMI. Moreover, the higher value of IMT was associated with an increasing risk of ALS with an odd ratio (OR) of 1.483 (95 % Confidence interval [1.026-2.144]). Eventually, evaluating IMT by classifying ALS patients based on the ALS Health State Scale (ALSHSS) from early to late stage revealed a non-linear increase in the OR (1.372, 1.898, 2.172, and 3.403). CONCLUSION The increased value of the carotid IMT independent of BMI in ALS could be assessed through ultrasonography as a convenient tool to evaluate the disease severity or possible systemic inflammation.
Collapse
Affiliation(s)
- Maryam Rezaee Semnani
- Department of Neurology, School of Medicine, Iran University of Medical Sciences, Iran
| | - Zahra Mirzaasgari
- Department of Neurology, School of Medicine, Iran University of Medical Sciences, Iran
| | - Armin Ariaei
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Bahram Haghi Ashtiani
- Department of Neurology, School of Medicine, Iran University of Medical Sciences, Iran.
| |
Collapse
|
9
|
Vaage AM, Benth JŠ, Meyer HE, Holmøy T, Nakken O. Premorbid lipid levels and long-term risk of ALS-a population-based cohort study. Amyotroph Lateral Scler Frontotemporal Degener 2024; 25:358-366. [PMID: 38117120 DOI: 10.1080/21678421.2023.2295455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 12/11/2023] [Indexed: 12/21/2023]
Abstract
OBJECTIVE To assess the temporal relationship between premorbid lipid levels and long-term amyotrophic lateral sclerosis (ALS) risk. METHODS From Norwegian cardiovascular health surveys (1974-2003), we collected information on total cholesterol (TC), triglycerides (TG), high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), glucose, and other cardiovascular risk factors. ALS incidence and mortality were identified through validated Norwegian health registries. The relation between premorbid lipid levels and ALS risk was assessed by Cox regression models. RESULTS Out of 640,066 study participants (51.5% females), 974 individuals (43.5% females) developed ALS. Mean follow-up time was 23.7 (SD 7.1) years among ALS cases. One mmol/l increase in LDL-C was associated with 6% increase in risk for ALS (hazard ratio 1.06 [95% CI: 1.01-1.09]). Higher levels of TC and TG were also associated with increased ALS risk, but only within the last 6-7 years prior to ALS diagnosis or death. No association between HDL-C and ALS risk was found. Adjusting for body mass index, birth cohort, smoking, and physical activity did not alter the results. CONCLUSIONS Higher levels of LDL-C are associated with increased ALS risk over 40 years later, compatible with a causal relationship. The temporal relationship between TG, TC, and ALS risk suggests that increased levels of these lipid biomarkers represent consequences of ALS.
Collapse
Affiliation(s)
- Anders Myhre Vaage
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Jūratė Šaltytė Benth
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Health Services Research Unit, Akershus University Hospital, Lørenskog, Norway
| | - Haakon E Meyer
- Department of Physical Health and Ageing, Norwegian Institute of Public Health, Oslo, Norway, and
- Department of Community Medicine and Global Health, University of Oslo, Oslo, Norway
| | - Trygve Holmøy
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ola Nakken
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway
| |
Collapse
|
10
|
Godoy-Corchuelo JM, Ali Z, Brito Armas JM, Martins-Bach AB, García-Toledo I, Fernández-Beltrán LC, López-Carbonero JI, Bascuñana P, Spring S, Jimenez-Coca I, Muñoz de Bustillo Alfaro RA, Sánchez-Barrena MJ, Nair RR, Nieman BJ, Lerch JP, Miller KL, Ozdinler HP, Fisher EMC, Cunningham TJ, Acevedo-Arozena A, Corrochano S. TDP-43-M323K causes abnormal brain development and progressive cognitive and motor deficits associated with mislocalised and increased levels of TDP-43. Neurobiol Dis 2024; 193:106437. [PMID: 38367882 PMCID: PMC10988218 DOI: 10.1016/j.nbd.2024.106437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/02/2024] [Accepted: 02/08/2024] [Indexed: 02/19/2024] Open
Abstract
TDP-43 pathology is found in several neurodegenerative disorders, collectively referred to as "TDP-43 proteinopathies". Aggregates of TDP-43 are present in the brains and spinal cords of >97% of amyotrophic lateral sclerosis (ALS), and in brains of ∼50% of frontotemporal dementia (FTD) patients. While mutations in the TDP-43 gene (TARDBP) are usually associated with ALS, many clinical reports have linked these mutations to cognitive impairments and/or FTD, but also to other neurodegenerative disorders including Parkinsonism (PD) or progressive supranuclear palsy (PSP). TDP-43 is a ubiquitously expressed, highly conserved RNA-binding protein that is involved in many cellular processes, mainly RNA metabolism. To investigate systemic pathological mechanisms in TDP-43 proteinopathies, aiming to capture the pleiotropic effects of TDP-43 mutations, we have further characterised a mouse model carrying a point mutation (M323K) within the endogenous Tardbp gene. Homozygous mutant mice developed cognitive and behavioural deficits as early as 3 months of age. This was coupled with significant brain structural abnormalities, mainly in the cortex, hippocampus, and white matter fibres, together with progressive cortical interneuron degeneration and neuroinflammation. At the motor level, progressive phenotypes appeared around 6 months of age. Thus, cognitive phenotypes appeared to be of a developmental origin with a mild associated progressive neurodegeneration, while the motor and neuromuscular phenotypes seemed neurodegenerative, underlined by a progressive loss of upper and lower motor neurons as well as distal denervation. This is accompanied by progressive elevated TDP-43 protein and mRNA levels in cortex and spinal cord of homozygous mutant mice from 3 months of age, together with increased cytoplasmic TDP-43 mislocalisation in cortex, hippocampus, hypothalamus, and spinal cord at 12 months of age. In conclusion, we find that Tardbp M323K homozygous mutant mice model many aspects of human TDP-43 proteinopathies, evidencing a dual role for TDP-43 in brain morphogenesis as well as in the maintenance of the motor system, making them an ideal in vivo model system to study the complex biology of TDP-43.
Collapse
Affiliation(s)
- Juan M Godoy-Corchuelo
- Neurological Disorders Group, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria Hospital Clínico San Carlos (IdiSSC), Madrid 28040, Spain
| | - Zeinab Ali
- Neurological Disorders Group, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria Hospital Clínico San Carlos (IdiSSC), Madrid 28040, Spain; MRC Harwell Institute, Oxfordshire, UK
| | - Jose M Brito Armas
- Unidad de Investigación, Hospital Universitario de Canarias, ITB-ULL and CIBERNED, La Laguna, Spain
| | | | - Irene García-Toledo
- Neurological Disorders Group, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria Hospital Clínico San Carlos (IdiSSC), Madrid 28040, Spain
| | - Luis C Fernández-Beltrán
- Neurological Disorders Group, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria Hospital Clínico San Carlos (IdiSSC), Madrid 28040, Spain; Department of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Juan I López-Carbonero
- Neurological Disorders Group, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria Hospital Clínico San Carlos (IdiSSC), Madrid 28040, Spain
| | - Pablo Bascuñana
- Brain Mapping Group, Hospital Clínico San Carlos, IdISSC, Madrid, Spain
| | - Shoshana Spring
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, ON, Canada
| | - Irene Jimenez-Coca
- Neurological Disorders Group, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria Hospital Clínico San Carlos (IdiSSC), Madrid 28040, Spain
| | | | - Maria J Sánchez-Barrena
- Department of Crystallography and Structural Biology, Institute of Physical Chemistry "Blas Cabrera", CSIC, Madrid, Spain
| | - Remya R Nair
- MRC Harwell Institute, Oxfordshire, UK; Nucleic Acid Therapy Accelerator (NATA), Harwell Campus, Oxfordshire, UK
| | - Brian J Nieman
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, ON, Canada
| | - Jason P Lerch
- Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, UK
| | - Karla L Miller
- Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, UK
| | - Hande P Ozdinler
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Elizabeth M C Fisher
- Department of Neuromuscular Diseases, and UCL Queen Square Motor Neuron Disease Centre, UCL, Institute of Neurology, London, UK
| | - Thomas J Cunningham
- MRC Harwell Institute, Oxfordshire, UK; MRC Prion Unit at UCL, UCL Institute of Prion Diseases, London, UK
| | - Abraham Acevedo-Arozena
- Unidad de Investigación, Hospital Universitario de Canarias, ITB-ULL and CIBERNED, La Laguna, Spain.
| | - Silvia Corrochano
- Neurological Disorders Group, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria Hospital Clínico San Carlos (IdiSSC), Madrid 28040, Spain; MRC Harwell Institute, Oxfordshire, UK.
| |
Collapse
|
11
|
Jamet M, Dupuis L, Gonzalez De Aguilar JL. Oligodendrocytes in amyotrophic lateral sclerosis and frontotemporal dementia: the new players on stage. Front Mol Neurosci 2024; 17:1375330. [PMID: 38585368 PMCID: PMC10995329 DOI: 10.3389/fnmol.2024.1375330] [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: 01/23/2024] [Accepted: 03/05/2024] [Indexed: 04/09/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are fatal adult-onset neurodegenerative disorders that share clinical, neuropathological and genetic features, which forms part of a multi-system disease spectrum. The pathological process leading to ALS and FTD is the result of the combination of multiple mechanisms that operate within specific populations of neurons and glial cells. The implication of oligodendrocytes has been the subject of a number of studies conducted on patients and related animal models. In this review we summarize our current knowledge on the alterations specific to myelin and the oligodendrocyte lineage occurring in ALS and FTD. We also consider different ways by which specific oligodendroglial alterations influence neurodegeneration and highlight the important role of oligodendrocytes in these two intrinsically associated neurodegenerative diseases.
Collapse
Affiliation(s)
| | | | - Jose-Luis Gonzalez De Aguilar
- Strasbourg Translational Neuroscience and Psychiatry, Institut National de la Santé et de la Recherche Médicale, Université de Strasbourg, Strasbourg, France
| |
Collapse
|
12
|
Zhu Y, Burg T, Neyrinck K, Vervliet T, Nami F, Vervoort E, Ahuja K, Sassano ML, Chai YC, Tharkeshwar AK, De Smedt J, Hu H, Bultynck G, Agostinis P, Swinnen JV, Van Den Bosch L, da Costa RFM, Verfaillie C. Disruption of MAM integrity in mutant FUS oligodendroglial progenitors from hiPSCs. Acta Neuropathol 2024; 147:6. [PMID: 38170217 PMCID: PMC10764485 DOI: 10.1007/s00401-023-02666-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: 07/26/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 01/05/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a rapidly progressive and fatal neurodegenerative disorder, characterized by selective loss of motor neurons (MNs). A number of causative genetic mutations underlie the disease, including mutations in the fused in sarcoma (FUS) gene, which can lead to both juvenile and late-onset ALS. Although ALS results from MN death, there is evidence that dysfunctional glial cells, including oligodendroglia, contribute to neurodegeneration. Here, we used human induced pluripotent stem cells (hiPSCs) with a R521H or a P525L mutation in FUS and their isogenic controls to generate oligodendrocyte progenitor cells (OPCs) by inducing SOX10 expression from a TET-On SOX10 cassette. Mutant and control iPSCs differentiated efficiently into OPCs. RNA sequencing identified a myelin sheath-related phenotype in mutant OPCs. Lipidomic studies demonstrated defects in myelin-related lipids, with a reduction of glycerophospholipids in mutant OPCs. Interestingly, FUSR521H OPCs displayed a decrease in the phosphatidylcholine/phosphatidylethanolamine ratio, known to be associated with maintaining membrane integrity. A proximity ligation assay further indicated that mitochondria-associated endoplasmic reticulum membranes (MAM) were diminished in both mutant FUS OPCs. Moreover, both mutant FUS OPCs displayed increased susceptibility to ER stress when exposed to thapsigargin, and exhibited impaired mitochondrial respiration and reduced Ca2+ signaling from ER Ca2+ stores. Taken together, these results demonstrate a pathological role of mutant FUS in OPCs, causing defects in lipid metabolism associated with MAM disruption manifested by impaired mitochondrial metabolism with increased susceptibility to ER stress and with suppressed physiological Ca2+ signaling. As such, further exploration of the role of oligodendrocyte dysfunction in the demise of MNs is crucial and will provide new insights into the complex cellular mechanisms underlying ALS.
Collapse
Affiliation(s)
- Yingli Zhu
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, 3000, Leuven, Belgium.
| | - Thibaut Burg
- Department of Neurosciences, Experimental Neurology, KU Leuven, Leuven Brain Institute (LBI), 3000, Leuven, Belgium
- Laboratory of Neurobiology, VIB, Center for Brain and Disease Research, 3000, Leuven, Belgium
| | - Katrien Neyrinck
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, 3000, Leuven, Belgium
| | - Tim Vervliet
- Laboratory of Molecular and Cellular Signalling, Department of Cellular and Molecular Medicine, KU Leuven, 3000, Leuven, Belgium
| | - Fatemeharefeh Nami
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, 3000, Leuven, Belgium
| | - Ellen Vervoort
- Laboratory of Cell Death Research and Therapy, Department of Cellular and Molecular Medicine, KU Leuven, 3000, Leuven, Belgium
- Center for Cancer Biology, VIB, 3000, Leuven, Belgium
| | - Karan Ahuja
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, 3000, Leuven, Belgium
- Animal Physiology and Neurobiology Section, Department of Biology, Neural Circuit Development and Regeneration Research Group, 3000, Leuven, Belgium
| | - Maria Livia Sassano
- Laboratory of Cell Death Research and Therapy, Department of Cellular and Molecular Medicine, KU Leuven, 3000, Leuven, Belgium
- Center for Cancer Biology, VIB, 3000, Leuven, Belgium
| | - Yoke Chin Chai
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, 3000, Leuven, Belgium
| | - Arun Kumar Tharkeshwar
- Department of Neurosciences, Experimental Neurology, KU Leuven, Leuven Brain Institute (LBI), 3000, Leuven, Belgium
- Laboratory of Neurobiology, VIB, Center for Brain and Disease Research, 3000, Leuven, Belgium
| | - Jonathan De Smedt
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, 3000, Leuven, Belgium
| | - Haibo Hu
- National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou, China
| | - Geert Bultynck
- Laboratory of Molecular and Cellular Signalling, Department of Cellular and Molecular Medicine, KU Leuven, 3000, Leuven, Belgium
| | - Patrizia Agostinis
- Laboratory of Cell Death Research and Therapy, Department of Cellular and Molecular Medicine, KU Leuven, 3000, Leuven, Belgium
- Center for Cancer Biology, VIB, 3000, Leuven, Belgium
| | - Johannes V Swinnen
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, KU Leuven, 3000, Leuven, Belgium
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology, KU Leuven, Leuven Brain Institute (LBI), 3000, Leuven, Belgium
- Laboratory of Neurobiology, VIB, Center for Brain and Disease Research, 3000, Leuven, Belgium
| | | | - Catherine Verfaillie
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, 3000, Leuven, Belgium
| |
Collapse
|
13
|
Yu W, Yu F, Li M, Yang F, Wang H, Song H, Huang X. Quantitative association between lead exposure and amyotrophic lateral sclerosis: a Bayesian network-based predictive study. Environ Health 2024; 23:2. [PMID: 38166850 PMCID: PMC10763408 DOI: 10.1186/s12940-023-01041-3] [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: 10/15/2023] [Accepted: 12/13/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Environmental lead (Pb) exposure have been suggested as a causative factor for amyotrophic lateral sclerosis (ALS). However, the role of Pb content of human body in ALS outcomes has not been quantified clearly. The purpose of this study was to apply Bayesian networks to forecast the risk of Pb exposure on the disease occurrence. METHODS We retrospectively collected medical records of ALS inpatients who underwent blood Pb testing, while matched controlled inpatients on age, gender, hospital ward and admission time according to the radio of 1:9. Tree Augmented Naïve Bayes (TAN), a semi-naïve Bayes classifier, was established to predict probability of ALS or controls with risk factors. RESULTS A total of 140 inpatients were included in this study. The whole blood Pb levels of ALS patients (57.00 μg/L) were more than twice as high as the controls (27.71 μg/L). Using the blood Pb concentrations to calculate probability of ALS, TAN produced the total coincidence rate of 90.00%. The specificity, sensitivity of Pb for ALS prediction was 0.79, or 0.74, respectively. CONCLUSION Therefore, these results provided quantitative evidence that Pb exposure may contribute to the development of ALS. Bayesian networks may be used to predict the ALS early onset with blood Pb levels.
Collapse
Affiliation(s)
- Wenxiu Yu
- Medical School of Chinese PLA, Beijing, 100853, China
- Neurological Department of the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Fangfang Yu
- Department of Medical Innovation Research, PLA General Hospital, Beijing, 100853, China
| | - Mao Li
- Neurological Department of the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Fei Yang
- Neurological Department of the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Hongfen Wang
- Medical School of Chinese PLA, Beijing, 100853, China
- Neurological Department of the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Han Song
- Department of Health Service, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Xusheng Huang
- Medical School of Chinese PLA, Beijing, 100853, China.
- Neurological Department of the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
14
|
Festa LK, Grinspan JB, Jordan-Sciutto KL. White matter injury across neurodegenerative disease. Trends Neurosci 2024; 47:47-57. [PMID: 38052682 PMCID: PMC10842057 DOI: 10.1016/j.tins.2023.11.003] [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: 07/09/2023] [Revised: 10/16/2023] [Accepted: 11/11/2023] [Indexed: 12/07/2023]
Abstract
Oligodendrocytes (OLs), the myelin-generating cells of the central nervous system (CNS), are active players in shaping neuronal circuitry and function. It has become increasingly apparent that injury to cells within the OL lineage plays a central role in neurodegeneration. In this review, we focus primarily on three degenerative disorders in which white matter loss is well documented: Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). We discuss clinical data implicating white matter injury as a key feature of these disorders, as well as shared and divergent phenotypes between them. We examine the cellular and molecular mechanisms underlying the alterations to OLs, including chronic neuroinflammation, aggregation of proteins, lipid dysregulation, and organellar stress. Last, we highlight prospects for therapeutic intervention targeting the OL lineage to restore function.
Collapse
Affiliation(s)
- Lindsay K Festa
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Judith B Grinspan
- Department of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kelly L Jordan-Sciutto
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
15
|
Okano H, Morimoto S, Kato C, Nakahara J, Takahashi S. Induced pluripotent stem cells-based disease modeling, drug screening, clinical trials, and reverse translational research for amyotrophic lateral sclerosis. J Neurochem 2023; 167:603-614. [PMID: 37952981 DOI: 10.1111/jnc.16005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/09/2023] [Accepted: 10/19/2023] [Indexed: 11/14/2023]
Abstract
It has been more than 10 years since the hopes for disease modeling and drug discovery using induced pluripotent stem cell (iPSC) technology boomed. Recently, clinical trials have been conducted with drugs identified using this technology, and some promising results have been reported. For amyotrophic lateral sclerosis (ALS), a devastating neurodegenerative disease, several groups have identified candidate drugs, ezogabine (retigabine), bosutinib, and ropinirole, using iPSCs-based drug discovery, and clinical trials using these drugs have been conducted, yielding interesting results. In our previous study, an iPSCs-based drug repurposing approach was utilized to show the potential of ropinirole hydrochloride (ROPI) in reducing ALS-specific pathological phenotypes. Recently, a phase 1/2a trial was conducted to investigate the effects of ropinirole on ALS further. This double-blind, randomized, placebo-controlled study confirmed the safety and tolerability of and provided evidence of its ability to delay disease progression and prolong the time to respiratory failure in ALS patients. Furthermore, in the reverse translational research, in vitro characterization of patient-derived iPSCs-motor neurons (MNs) mimicked the therapeutic effects of ROPI in vivo, suggesting the potential application of this technology to the precision medicine of ALS. Interestingly, RNA-seq data showed that ROPI treatment suppressed the sterol regulatory element-binding protein 2-dependent cholesterol biosynthesis pathway. Therefore, this pathway may be involved in the therapeutic effect of ROPI on ALS. The possibility that this pathway may be involved in the therapeutic effect of ALS was demonstrated. Finally, new future strategies for ALS using iPSCs technology will be discussed in this paper.
Collapse
Affiliation(s)
- Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Satoru Morimoto
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Chris Kato
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Jin Nakahara
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Shinichi Takahashi
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
- Department of Neurology and Stroke, Saitama Medical University International Medical Center, Saitama, Japan
| |
Collapse
|
16
|
Ke H, Liu K, Jiao B, Zhao L. Implications of TDP-43 in non-neuronal systems. Cell Commun Signal 2023; 21:338. [PMID: 37996849 PMCID: PMC10666381 DOI: 10.1186/s12964-023-01336-5] [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: 07/27/2023] [Accepted: 09/26/2023] [Indexed: 11/25/2023] Open
Abstract
TAR DNA-binding protein 43 (TDP-43) is a versatile RNA/DNA-binding protein with multifaceted processes. While TDP-43 has been extensively studied in the context of degenerative diseases, recent evidence has also highlighted its crucial involvement in diverse life processes beyond neurodegeneration. Here, we mainly reviewed the function of TDP-43 in non-neurodegenerative physiological and pathological processes, including spermatogenesis, embryonic development, mammary gland development, tumor formation, and viral infection, highlighting its importance as a key regulatory factor for the maintenance of normal functions throughout life. TDP-43 exhibits diverse and sometimes opposite functionality across different cell types through various mechanisms, and its roles can shift at distinct stages within the same biological system. Consequently, TDP-43 operates in both a context-dependent and a stage-specific manner in response to a variety of internal and external stimuli. Video Abstract.
Collapse
Affiliation(s)
- Hao Ke
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, 330031, China
| | - Kang Liu
- Ganzhou People's Hospital, Ganzhou, 341000, China
| | - Baowei Jiao
- National Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650201, China.
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China.
| | - Limin Zhao
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, 330031, China.
| |
Collapse
|
17
|
Morimoto S, Takahashi S, Ito D, Daté Y, Okada K, Kato C, Nakamura S, Ozawa F, Chyi CM, Nishiyama A, Suzuki N, Fujimori K, Kondo T, Takao M, Hirai M, Kabe Y, Suematsu M, Jinzaki M, Aoki M, Fujiki Y, Sato Y, Suzuki N, Nakahara J, Okano H. Phase 1/2a clinical trial in ALS with ropinirole, a drug candidate identified by iPSC drug discovery. Cell Stem Cell 2023; 30:766-780.e9. [PMID: 37267913 DOI: 10.1016/j.stem.2023.04.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 01/12/2023] [Accepted: 04/24/2023] [Indexed: 06/04/2023]
Abstract
iPSC-based drug discovery led to a phase 1/2a trial of ropinirole in ALS. 20 participants with sporadic ALS received ropinirole or placebo for 24 weeks in the double-blind period to evaluate safety, tolerability, and therapeutic effects. Adverse events were similar in both groups. During the double-blind period, muscle strength and daily activity were maintained, but a decline in the ALSFRS-R, which assesses the functional status of ALS patients, was not different from that in the placebo group. However, in the open-label extension period, the ropinirole group showed significant suppression of ALSFRS-R decline and an additional 27.9 weeks of disease-progression-free survival. iPSC-derived motor neurons from participants showed dopamine D2 receptor expression and a potential involvement of the SREBP2-cholesterol pathway in therapeutic effects. Lipid peroxide represents a clinical surrogate marker to assess disease progression and drug efficacy. Limitations include small sample sizes and high attrition rates in the open-label extension period, requiring further validation.
Collapse
Affiliation(s)
- Satoru Morimoto
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan; Department of Neurology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Shinichi Takahashi
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan; Department of Neurology, Keio University School of Medicine, Tokyo 160-8582, Japan; Department of Neurology and Stroke, Saitama Medical University International Medical Center, Saitama 350-1298, Japan
| | - Daisuke Ito
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan; Department of Neurology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Yugaku Daté
- Department of Neurology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Kensuke Okada
- Department of Neurology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Chris Kato
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Shiho Nakamura
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Fumiko Ozawa
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Chai Muh Chyi
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan; Keio University Global Research Institute, Tokyo 108-8345, Japan
| | - Ayumi Nishiyama
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Naoki Suzuki
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Koki Fujimori
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Tosho Kondo
- Research Center of Neurology, ONO Pharmaceutical Co., Ltd., Osaka 541-8564, Japan
| | - Masaki Takao
- Department of Clinical Laboratory, National Center of Neurology and Psychiatry (NCNP), Tokyo 187-0031, Japan; Department of Neurology, Mihara Memorial Hospital, Isesaki, Gunmma 372-0006, Japan
| | - Miwa Hirai
- Department of Biochemistry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Yasuaki Kabe
- Department of Biochemistry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Makoto Suematsu
- Department of Biochemistry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Masahiro Jinzaki
- Department of Radiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Masashi Aoki
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Yuto Fujiki
- Keio University Hospital Clinical and Translational Research Center, Tokyo 160-8582, Japan
| | - Yasunori Sato
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Norihiro Suzuki
- Department of Neurology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Jin Nakahara
- Department of Neurology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan.
| |
Collapse
|
18
|
Gao YH, Li X. Cholesterol metabolism: Towards a therapeutic approach for multiple sclerosis. Neurochem Int 2023; 164:105501. [PMID: 36803679 DOI: 10.1016/j.neuint.2023.105501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 01/26/2023] [Accepted: 01/30/2023] [Indexed: 02/17/2023]
Abstract
Growing evidence points to the importance of cholesterol in preserving brain homeostasis. Cholesterol makes up the main component of myelin in the brain, and myelin integrity is vital in demyelinating diseases such as multiple sclerosis. Because of the connection between myelin and cholesterol, the interest in cholesterol in the central nervous system increased during the last decade. In this review, we provide a detailed overview on brain cholesterol metabolism in multiple sclerosis and its role in promoting oligodendrocyte precursor cell differentiation and remyelination.
Collapse
Affiliation(s)
- Yu-Han Gao
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China
| | - Xing Li
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China.
| |
Collapse
|
19
|
Association of blood lipids with onset and prognosis of amyotrophic lateral sclerosis: results from the ALS Swabia registry. J Neurol 2023; 270:3082-3090. [PMID: 36853389 DOI: 10.1007/s00415-023-11630-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/15/2023] [Accepted: 02/16/2023] [Indexed: 03/01/2023]
Abstract
BACKGROUND To date, the role of blood lipid levels and their association with the onset and prognosis of ALS is controversial. We explored these associations in a large, population-based case-control study. METHODS Between October 2010 and June 2014, 336 ALS patients (mean age 65.7 ± 10.7; 57.7% male) and 487 sex- and age-matched controls from the same geographic region were recruited within the ALS registry in Southwest Germany. Triglycerides and cholesterol (high-density lipoprotein (HDL), low-density lipoprotein (LDL), total) were measured. The ALS cohort was followed up for vital status. Conditional logistic regression models were applied to calculate odds ratio (OR) for risk of ALS associated with serum lipid concentrations. In ALS patients only, survival models were used to appraise the prognostic value. RESULTS High concentration of total cholesterol (OR 1.60, 95% confidence interval (CI) 1.03-2.49, top vs. bottom quartile), but not HDL, LDL, LDL-HDL ratio, or triglycerides, was positively associated with the risk of ALS. During the median follow-up time of 88.9 months, 291 deaths occurred among 336 ALS patients. In the adjusted survival analysis, higher HDL (HR 1.72, 95% CI 1.19-2.50) and LDL cholesterol levels (HR 1.58, 95% CI 1.11-2.26) were associated with higher mortality in ALS patients. In contrast, higher triglyceride levels were associated with lower mortality (HR 0.68, 95% CI 0.48-0.96). CONCLUSION The results highlight the importance to distinguish cholesterol from triglycerides when considering the prognostic role of lipid metabolism in ALS. It further strengthens the rationale for a triglyceride-rich diet, while the negative impact of cholesterol must be further explored.
Collapse
|
20
|
Lépine S, Castellanos-Montiel MJ, Durcan TM. TDP-43 dysregulation and neuromuscular junction disruption in amyotrophic lateral sclerosis. Transl Neurodegener 2022; 11:56. [PMID: 36575535 PMCID: PMC9793560 DOI: 10.1186/s40035-022-00331-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/29/2022] [Indexed: 12/28/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a disease characterized by upper and lower motor neuron (MN) loss with a signature feature of cytoplasmic aggregates containing TDP-43, which are detected in nearly all patients. Mutations in the gene that encodes TDP-43 (TARBDP) are known to result in both familial and sporadic ALS. In ALS, disruption of neuromuscular junctions (NMJs) constitutes a critical event in disease pathogenesis, leading to denervation atrophy, motor impairments and disability. Morphological defects and impaired synaptic transmission at NMJs have been reported in several TDP-43 animal models and in vitro, linking TDP-43 dysregulation to the loss of NMJ integrity in ALS. Through the lens of the dying-back and dying-forward hypotheses of ALS, this review discusses the roles of TDP-43 related to synaptic function, with a focus on the potential molecular mechanisms occurring within MNs, skeletal muscles and glial cells that may contribute to NMJ disruption in ALS.
Collapse
Affiliation(s)
- Sarah Lépine
- grid.14709.3b0000 0004 1936 8649The Neuro’s Early Drug Discovery Unit (EDDU), Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, 3801 University Street, Montreal, QC H3A 2B4 Canada ,grid.14709.3b0000 0004 1936 8649Faculty of Medicine and Health Sciences, McGill University, 3605 De La Montagne, Montreal, QC H3G 2M1 Canada
| | - Maria José Castellanos-Montiel
- grid.14709.3b0000 0004 1936 8649The Neuro’s Early Drug Discovery Unit (EDDU), Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, 3801 University Street, Montreal, QC H3A 2B4 Canada
| | - Thomas Martin Durcan
- grid.14709.3b0000 0004 1936 8649The Neuro’s Early Drug Discovery Unit (EDDU), Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, 3801 University Street, Montreal, QC H3A 2B4 Canada
| |
Collapse
|
21
|
Hartmann H, Ho WY, Chang JC, Ling SC. Cholesterol dyshomeostasis in amyotrophic lateral sclerosis: cause, consequence, or epiphenomenon? FEBS J 2022; 289:7688-7709. [PMID: 34469619 DOI: 10.1111/febs.16175] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/10/2021] [Accepted: 08/31/2021] [Indexed: 01/14/2023]
Abstract
Amyotrophic lateral sclerosis (ALS), the most common adult-onset motor neuron disease, is characterized by the selective degeneration of motor neurons leading to paralysis and eventual death. Multiple pathogenic mechanisms, including systemic dysmetabolism, have been proposed to contribute to ALS. Among them, dyslipidemia, i.e., abnormal level of cholesterol and other lipids in the circulation and central nervous system (CNS), has been reported in ALS patients, but without a consensus. Cholesterol is a constituent of cellular membranes and a precursor of steroid hormones, oxysterols, and bile acids. Consequently, optimal cholesterol levels are essential for health. Due to the blood-brain barrier (BBB), cholesterol cannot move between the CNS and the rest of the body. As such, cholesterol metabolism in the CNS is proposed to operate autonomously. Despite its importance, it remains elusive how cholesterol dyshomeostasis may contribute to ALS. In this review, we aim to describe the current state of cholesterol metabolism research in ALS, identify unresolved issues, and provide potential directions.
Collapse
Affiliation(s)
- Hannelore Hartmann
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Wan Yun Ho
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jer-Cherng Chang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Shuo-Chien Ling
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Healthy Longevity Translational Research Programme, National University Health System, Singapore, Singapore.,Program in Neuroscience and Behavior Disorders, Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
22
|
Katzeff JS, Lok HC, Bhatia S, Fu Y, Halliday GM, Kim WS. ATP-binding cassette transporter expression is widely dysregulated in frontotemporal dementia with TDP-43 inclusions. Front Mol Neurosci 2022; 15:1043127. [PMID: 36385764 PMCID: PMC9663841 DOI: 10.3389/fnmol.2022.1043127] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 10/11/2022] [Indexed: 10/17/2023] Open
Abstract
The human brain is highly enriched in lipids and increasing evidence indicates that dysregulation of lipids in the brain is associated with neurodegeneration. ATP-binding cassette subfamily A (ABCA) transporters control the movement of lipids across cellular membranes and are implicated in a number of neurodegenerative diseases. However, very little is known about the role of ABCA transporters in frontotemporal lobar degeneration with TDP-43 inclusions (FTLD-TDP), which is a common form of younger-onset dementia. We therefore undertook a comprehensive analysis of the expression of ABCA transporters (ABCA1-13) in five key brain regions (amygdala, inferior temporal cortex, superior frontal cortex, cerebellum and parietal cortex) in FTLD-TDP and controls. We found that the expression of ABCA2, ABCA3, ABCA4, ABCA7, ABCA9, ABCA10 and ABCA13 was significantly altered in FTLD-TDP in a region-specific manner. In addition, the expression of ABCA transporters correlated specifically to different neural markers and TARDBP. These results suggest substantial dysregulation of ABCA transporters and lipid metabolism in FTLD-TDP and these changes are associated with neuroinflammation.
Collapse
Affiliation(s)
| | | | | | | | | | - Woojin Scott Kim
- Brain and Mind Centre & School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
23
|
Afridi R, Rahman MH, Suk K. Implications of glial metabolic dysregulation in the pathophysiology of neurodegenerative diseases. Neurobiol Dis 2022; 174:105874. [PMID: 36154877 DOI: 10.1016/j.nbd.2022.105874] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 08/28/2022] [Accepted: 09/21/2022] [Indexed: 11/16/2022] Open
Abstract
Glial cells are the most abundant cells of the brain, outnumbering neurons. These multifunctional cells are crucial for maintaining brain homeostasis by providing trophic and nutritional support to neurons, sculpting synapses, and providing an immune defense. Glia are highly plastic and undergo both structural and functional alterations in response to changes in the brain microenvironment. Glial phenotypes are intimately regulated by underlying metabolic machinery, which dictates the effector functions of these cells. Altered brain energy metabolism and chronic neuroinflammation are common features of several neurodegenerative diseases. Microglia and astrocytes are the major glial cells fueling the ongoing neuroinflammatory process, exacerbating neurodegeneration. Distinct metabolic perturbations in microglia and astrocytes, including altered carbohydrate, lipid, and amino acid metabolism have been documented in neurodegenerative diseases. These disturbances aggravate the neurodegenerative process by potentiating the inflammatory activation of glial cells. This review covers the recent advances in the molecular aspects of glial metabolic changes in the pathophysiology of neurodegenerative diseases. Finally, we discuss studies exploiting glial metabolism as a potential therapeutic avenue in neurodegenerative diseases.
Collapse
Affiliation(s)
- Ruqayya Afridi
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Md Habibur Rahman
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Kyoungho Suk
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; Brain Science and Engineering Institute, Kyungpook National University, Daegu 41944, Republic of Korea.
| |
Collapse
|
24
|
Altered TDP-43 Structure and Function: Key Insights into Aberrant RNA, Mitochondrial, and Cellular and Systemic Metabolism in Amyotrophic Lateral Sclerosis. Metabolites 2022; 12:metabo12080709. [PMID: 36005581 PMCID: PMC9415507 DOI: 10.3390/metabo12080709] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 07/27/2022] [Accepted: 07/27/2022] [Indexed: 12/10/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive and fatal neuromuscular disorder with no cure available and limited treatment options. ALS is a highly heterogeneous disease, whereby patients present with vastly different phenotypes. Despite this heterogeneity, over 97% of patients will exhibit pathological TAR-DNA binding protein-43 (TDP-43) cytoplasmic inclusions. TDP-43 is a ubiquitously expressed RNA binding protein with the capacity to bind over 6000 RNA and DNA targets—particularly those involved in RNA, mitochondrial, and lipid metabolism. Here, we review the unique structure and function of TDP-43 and its role in affecting the aforementioned metabolic processes in ALS. Considering evidence published specifically in TDP-43-relevant in vitro, in vivo, and ex vivo models we posit that TDP-43 acts in a positive feedback loop with mRNA transcription/translation, stress granules, cytoplasmic aggregates, and mitochondrial proteins causing a relentless cycle of disease-like pathology eventuating in neuronal toxicity. Given its undeniable presence in ALS pathology, TDP-43 presents as a promising target for mechanistic disease modelling and future therapeutic investigations.
Collapse
|
25
|
Dermitzakis I, Manthou ME, Meditskou S, Miliaras D, Kesidou E, Boziki M, Petratos S, Grigoriadis N, Theotokis P. Developmental Cues and Molecular Drivers in Myelinogenesis: Revisiting Early Life to Re-Evaluate the Integrity of CNS Myelin. Curr Issues Mol Biol 2022; 44:3208-3237. [PMID: 35877446 PMCID: PMC9324160 DOI: 10.3390/cimb44070222] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/14/2022] [Accepted: 07/17/2022] [Indexed: 02/07/2023] Open
Abstract
The mammalian central nervous system (CNS) coordinates its communication through saltatory conduction, facilitated by myelin-forming oligodendrocytes (OLs). Despite the fact that neurogenesis from stem cell niches has caught the majority of attention in recent years, oligodendrogenesis and, more specifically, the molecular underpinnings behind OL-dependent myelinogenesis, remain largely unknown. In this comprehensive review, we determine the developmental cues and molecular drivers which regulate normal myelination both at the prenatal and postnatal periods. We have indexed the individual stages of myelinogenesis sequentially; from the initiation of oligodendrocyte precursor cells, including migration and proliferation, to first contact with the axon that enlists positive and negative regulators for myelination, until the ultimate maintenance of the axon ensheathment and myelin growth. Here, we highlight multiple developmental pathways that are key to successful myelin formation and define the molecular pathways that can potentially be targets for pharmacological interventions in a variety of neurological disorders that exhibit demyelination.
Collapse
Affiliation(s)
- Iasonas Dermitzakis
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
| | - Maria Eleni Manthou
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
| | - Soultana Meditskou
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
| | - Dimosthenis Miliaras
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
| | - Evangelia Kesidou
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, 54621 Thessaloniki, Greece; (E.K.); (M.B.); (N.G.)
| | - Marina Boziki
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, 54621 Thessaloniki, Greece; (E.K.); (M.B.); (N.G.)
| | - Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC 3004, Australia;
| | - Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, 54621 Thessaloniki, Greece; (E.K.); (M.B.); (N.G.)
| | - Paschalis Theotokis
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, 54621 Thessaloniki, Greece; (E.K.); (M.B.); (N.G.)
- Correspondence:
| |
Collapse
|
26
|
Ho WY, Hartmann H, Ling SC. Central nervous system cholesterol metabolism in health and disease. IUBMB Life 2022; 74:826-841. [PMID: 35836360 DOI: 10.1002/iub.2662] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 06/23/2022] [Indexed: 12/19/2022]
Abstract
Cholesterol is a ubiquitous and essential component of cellular membranes, as it regulates membrane structure and fluidity. Furthermore, cholesterol serves as a precursor for steroid hormones, oxysterol, and bile acids, that are essential for maintaining many of the body's metabolic processes. The biosynthesis and excretion of cholesterol is tightly regulated in order to maintain homeostasis. Although virtually all cells have the capacity to make cholesterol, the liver and brain are the two main organs producing cholesterol in mammals. Once produced, cholesterol is transported in the form of lipoprotein particles to other cell types and tissues. Upon formation of the blood-brain barrier (BBB) during embryonic development, lipoproteins cannot move between the central nervous system (CNS) and the rest of the body. As such, cholesterol biosynthesis and metabolism in the CNS operate autonomously without input from the circulation system in normal physiological conditions. Nevertheless, similar regulatory mechanisms for maintaining cholesterol homeostasis are utilized in both the CNS and peripheral systems. Here, we discuss the functions and metabolism of cholesterol in the CNS. We further focus on how different CNS cell types contribute to cholesterol metabolism, and how ApoE, the major CNS apolipoprotein, is involved in normal and pathophysiological functions. Understanding these basic mechanisms will aid our ability to elucidate how CNS cholesterol dysmetabolism contributes to neurogenerative diseases.
Collapse
Affiliation(s)
- Wan Y Ho
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Program in Neuroscience and Behavior Disorders, Duke-NUS Medical School, Singapore
| | - Hannelore Hartmann
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Shuo-Chien Ling
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Program in Neuroscience and Behavior Disorders, Duke-NUS Medical School, Singapore.,Healthy Longevity Translational Research Programme, National University Health System, Singapore
| |
Collapse
|
27
|
TDP-43 regulates cholesterol biosynthesis by inhibiting sterol regulatory element-binding protein 2. Sci Rep 2022; 12:7988. [PMID: 35568729 PMCID: PMC9107471 DOI: 10.1038/s41598-022-12133-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 04/22/2022] [Indexed: 12/29/2022] Open
Abstract
Dyslipidemia is considered an essential component of the pathological process of amyotrophic lateral sclerosis (ALS), a fatal motor neuron disease. Although TAR DNA Binding Protein 43 kDa (TDP-43) links both familial and sporadic forms of ALS and cytoplasmic aggregates are a hallmark of most cases of ALS, the molecular mechanism and the in vivo relation of ALS dyslipidemia with TDP-43 have been unclear. To analyze the dyslipidemia-related gene expression by TDP-43, we performed expression microarray and RNA deep sequencing (RNA-Seq) using cell lines expressing high levels of TDP-43 and identified 434 significantly altered genes including sterol regulatory element-binding protein 2 (SREBP2), a master regulator of cholesterol homeostasis and its downstream genes. Elevated TDP-43 impaired SREBP2 transcriptional activity, leading to inhibition of cholesterol biosynthesis. The amount of cholesterol was significantly decreased in the spinal cords of TDP-43-overexpressed ALS model mice and in the cerebrospinal fluids of ALS patients. These results suggested that TDP-43 could play an essential role in cholesterol biosynthesis in relation to ALS dyslipidemia.
Collapse
|
28
|
Bottero V, Santiago JA, Quinn JP, Potashkin JA. Key Disease Mechanisms Linked to Amyotrophic Lateral Sclerosis in Spinal Cord Motor Neurons. Front Mol Neurosci 2022; 15:825031. [PMID: 35370543 PMCID: PMC8965442 DOI: 10.3389/fnmol.2022.825031] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/15/2022] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease with no modifying treatments available. The molecular mechanisms underpinning disease pathogenesis are not fully understood. Recent studies have employed co-expression networks to identify key genes, known as “switch genes”, responsible for dramatic transcriptional changes in the blood of ALS patients. In this study, we directly investigate the root cause of ALS by examining the changes in gene expression in motor neurons that degenerate in patients. Co-expression networks identified in ALS patients’ spinal cord motor neurons revealed 610 switch genes in seven independent microarrays. Switch genes were enriched in several pathways, including viral carcinogenesis, PI3K-Akt, focal adhesion, proteoglycans in cancer, colorectal cancer, and thyroid hormone signaling. Transcription factors ELK1 and GATA2 were identified as key master regulators of the switch genes. Protein-chemical network analysis identified valproic acid, cyclosporine, estradiol, acetaminophen, quercetin, and carbamazepine as potential therapeutics for ALS. Furthermore, the chemical analysis identified metals and organic compounds including, arsenic, copper, nickel, and benzo(a)pyrene as possible mediators of neurodegeneration. The identification of switch genes provides insights into previously unknown biological pathways associated with ALS.
Collapse
Affiliation(s)
- Virginie Bottero
- Chicago Medical School, Rosalind Franklin University of Medicine and Science, Center for Neurodegenerative Diseases and Therapeutics, Discipline of Cellular and Molecular Pharmacology, North Chicago, IL, United States
| | | | | | - Judith A. Potashkin
- Chicago Medical School, Rosalind Franklin University of Medicine and Science, Center for Neurodegenerative Diseases and Therapeutics, Discipline of Cellular and Molecular Pharmacology, North Chicago, IL, United States
- *Correspondence: Judy A. Potashkin
| |
Collapse
|
29
|
Chang TY, Chang CCY, Harned TC, De La Torre AL, Lee J, Huynh TN, Gow JG. Blocking cholesterol storage to treat Alzheimer's disease. EXPLORATION OF NEUROPROTECTIVE THERAPY 2021; 1:173-184. [PMID: 35199105 PMCID: PMC8863366 DOI: 10.37349/ent.2021.00014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/07/2021] [Indexed: 12/11/2022]
Abstract
Cholesterol serves as an essential lipid molecule in various membrane organelles of mammalian cells. The metabolites of cholesterol also play important functions. Acyl-coenzyme A: cholesterol acyltransferase 1 (ACAT1), also named as sterol O-acyltransferase 1, is a membrane-bound enzyme residing at the endoplasmic reticulum (ER). It converts cholesterol to cholesteryl esters (CEs) for storage, and is expressed in all cells. CEs cannot partition in membranes; they can only coalesce as cytosolic lipid droplets. Excess CEs are found in the vulnerable region of the brains of patients with late-onset Alzheimer's disease (AD), and in cell and mouse models for AD. Reducing CE contents by genetic inactivation of ACAT1, or by pharmacological inhibition of ACAT is shown to reduce amyloidopathy and other hallmarks for AD. To account for the various beneficial actions of the ACAT1 blockade (A1B), a working hypothesis is proposed here: the increase in CE contents observed in the AD brain is caused by damages of cholesterol-rich lipid rafts that are known to occur in neurons affected by AD. These damages cause cholesterol to release from lipid rafts and move to the ER where it will be converted to CEs by ACAT1. In addition, the increase in CE contents may also be caused by overloading with cholesterol-rich substances, or through activation of ACAT1 gene expression by various proinflammatory agents. Both scenarios may occur in microglia of the chronically inflamed brain. A1B ameliorates AD by diverting the cholesterol pool destined for CE biosynthesis such that it can be utilized more efficiently to repair membrane damage in various organelles, and to exert regulatory actions more effectively to defend against AD. To test the validity of the A1B hypothesis in cell culture and in vivo, the current status of various anti-ACAT1 agents that could be further developed is briefly discussed.
Collapse
Affiliation(s)
- Ta Yuan Chang
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Catherine C. Y. Chang
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Taylor C. Harned
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Adrianna L. De La Torre
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Junghoon Lee
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Thao N. Huynh
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - James G. Gow
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| |
Collapse
|
30
|
Histone Deacetylase Inhibition Regulates Lipid Homeostasis in a Mouse Model of Amyotrophic Lateral Sclerosis. Int J Mol Sci 2021; 22:ijms222011224. [PMID: 34681883 PMCID: PMC8541517 DOI: 10.3390/ijms222011224] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/08/2021] [Accepted: 10/15/2021] [Indexed: 12/19/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an incurable and fatal neurodegenerative disorder of the motor system. While the etiology is still incompletely understood, defects in metabolism act as a major contributor to the disease progression. Recently, histone deacetylase (HDAC) inhibition using ACY-738 has been shown to restore metabolic alterations in the spinal cord of a FUS mouse model of ALS, which was accompanied by a beneficial effect on the motor phenotype and survival. In this study, we investigated the specific effects of HDAC inhibition on lipid metabolism using untargeted lipidomic analysis combined with transcriptomic analysis in the spinal cord of FUS mice. We discovered that symptomatic FUS mice recapitulate lipid alterations found in ALS patients and in the SOD1 mouse model. Glycerophospholipids, sphingolipids, and cholesterol esters were most affected. Strikingly, HDAC inhibition mitigated lipid homeostasis defects by selectively targeting glycerophospholipid metabolism and reducing cholesteryl esters accumulation. Therefore, our data suggest that HDAC inhibition is a potential new therapeutic strategy to modulate lipid metabolism defects in ALS and potentially other neurodegenerative diseases.
Collapse
|