1
|
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
|
2
|
Gao S, Wang Y, Wang J, Dong Y. Leveraging explainable deep learning methodologies to elucidate the biological underpinnings of Huntington's disease using single-cell RNA sequencing data. BMC Genomics 2024; 25:930. [PMID: 39367331 PMCID: PMC11451194 DOI: 10.1186/s12864-024-10855-5] [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: 05/10/2024] [Accepted: 09/30/2024] [Indexed: 10/06/2024] Open
Abstract
BACKGROUND Huntington's disease (HD) is a hereditary neurological disorder caused by mutations in HTT, leading to neuronal degeneration. Traditionally, HD is associated with the misfolding and aggregation of mutant huntingtin due to an extended polyglutamine domain encoded by an expanded CAG tract. However, recent research has also highlighted the role of global transcriptional dysregulation in HD pathology. However, understanding the intricate relationship between mRNA expression and HD at the cellular level remains challenging. Our study aimed to elucidate the underlying mechanisms of HD pathology using single-cell sequencing data. RESULTS We used single-cell RNA sequencing analysis to determine differential gene expression patterns between healthy and HD cells. HD cells were effectively modeled using a residual neural network (ResNet), which outperformed traditional and convolutional neural networks. Despite the efficacy of our approach, the F1 score for the test set was 96.53%. Using the SHapley Additive exPlanations (SHAP) algorithm, we identified genes influencing HD prediction and revealed their roles in HD pathobiology, such as in the regulation of cellular iron metabolism and mitochondrial function. SHAP analysis also revealed low-abundance genes that were overlooked by traditional differential expression analysis, emphasizing its effectiveness in identifying biologically relevant genes for distinguishing between healthy and HD cells. Overall, the integration of single-cell RNA sequencing data and deep learning models provides valuable insights into HD pathology. CONCLUSION We developed the model capable of analyzing HD at single-cell transcriptomic level.
Collapse
Affiliation(s)
- Shichen Gao
- School of Life Sciences, Anhui University, Hefei, 230601, China
- College of Biology and Food Engineering, Chuzhou University, Chuzhou, 239000, China
| | - Yadong Wang
- School of Life Sciences, Anhui University, Hefei, 230601, China
- College of Biology and Food Engineering, Chuzhou University, Chuzhou, 239000, China
| | - Jiajia Wang
- College of Biology and Food Engineering, Chuzhou University, Chuzhou, 239000, China
| | - Yan Dong
- College of Biology and Food Engineering, Chuzhou University, Chuzhou, 239000, China.
| |
Collapse
|
3
|
Cho K, Kim GW. Decreased SREBP2 of the striatal cell relates to disrupted protein degradation in Huntington's disease. Brain Res 2024; 1846:149250. [PMID: 39313167 DOI: 10.1016/j.brainres.2024.149250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 09/14/2024] [Accepted: 09/20/2024] [Indexed: 09/25/2024]
Abstract
This study delineated the intricate relation between cholesterol metabolism, protein degradation mechanisms, and the pathogenesis of Huntington's disease (HD). Through investigations using both animal models and cellular systems, we have observed significant alterations in cholesterol levels, particularly in the striatum, which is the primary lesion site in HD. Our findings indicate the dysregulation of cholesterol metabolism-related factors, such as LDLR and SREBP2, in HD, which may contribute to disease progression. Additionally, we uncovered disruptions in protein degradation pathways, including decreased neddylated proteins and dysregulated autophagy, which further exacerbated HD pathology. Moreover, our study highlighted the potential therapeutic implications of targeting these pathways. By restoring cholesterol levels and modulating protein degradation mechanisms, particularly through interventions, such as MLN4924, we observed potential improvements in cellular function, as indicated by the increased BDNF levels. These insights underscore the importance of simultaneously addressing cholesterol metabolism and protein degradation to alleviate HD pathology. Collectively, this study provides a basic understanding of the interplay between the decrease of SREBP2 and the dysfunctional protein degradation system derived from disrupted cholesterol metabolism in HD and HD cells.
Collapse
Affiliation(s)
- Kyoungjoo Cho
- Department of Life Science, Kyonggi University, Suwon, South Korea
| | - Gyung Whan Kim
- Department of Neurology, College of Medicine, Yonsei University, Seoul, South Korea.
| |
Collapse
|
4
|
Scolz A, Vezzoli E, Villa M, Talpo F, Cazzola J, Raffin F, Cordiglieri C, Falqui A, Pepe G, Maglione V, Besusso D, Biella G, Zuccato C. Neuroprotection by ADAM10 inhibition requires TrkB signaling in the Huntington's disease hippocampus. Cell Mol Life Sci 2024; 81:333. [PMID: 39112663 PMCID: PMC11335257 DOI: 10.1007/s00018-024-05382-1] [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: 01/30/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/22/2024]
Abstract
Synaptic dysfunction is an early pathogenic event leading to cognitive decline in Huntington's disease (HD). We previously reported that the active ADAM10 level is increased in the HD cortex and striatum, causing excessive proteolysis of the synaptic cell adhesion protein N-Cadherin. Conversely, ADAM10 inhibition is neuroprotective and prevents cognitive decline in HD mice. Although the breakdown of cortico-striatal connection has been historically linked to cognitive deterioration in HD, dendritic spine loss and long-term potentiation (LTP) defects identified in the HD hippocampus are also thought to contribute to the cognitive symptoms of the disease. The aim of this study is to investigate the contribution of ADAM10 to spine pathology and LTP defects of the HD hippocampus. We provide evidence that active ADAM10 is increased in the hippocampus of two mouse models of HD, leading to extensive proteolysis of N-Cadherin, which has a widely recognized role in spine morphology and synaptic plasticity. Importantly, the conditional heterozygous deletion of ADAM10 in the forebrain of HD mice resulted in the recovery of spine loss and ultrastructural synaptic defects in CA1 pyramidal neurons. Meanwhile, normalization of the active ADAM10 level increased the pool of synaptic BDNF protein and activated ERK neuroprotective signaling in the HD hippocampus. We also show that the ADAM10 inhibitor GI254023X restored LTP defects and increased the density of mushroom spines enriched with GluA1-AMPA receptors in HD hippocampal neurons. Notably, we report that administration of the TrkB antagonist ANA12 to HD hippocampal neurons reduced the beneficial effect of GI254023X, indicating that the BDNF receptor TrkB contributes to mediate the neuroprotective activity exerted by ADAM10 inhibition in HD. Collectively, these findings indicate that ADAM10 inhibition coupled with TrkB signaling represents an efficacious strategy to prevent hippocampal synaptic plasticity defects and cognitive dysfunction in HD.
Collapse
Affiliation(s)
- Andrea Scolz
- Department of Biosciences, University of Milan, Milan, Italy
- Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Elena Vezzoli
- Department of Biosciences, University of Milan, Milan, Italy
- Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
- Advanced Light and Electron Microscopy BioImaging Centre (ALEMBIC), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Michela Villa
- Department of Biosciences, University of Milan, Milan, Italy
- Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Francesca Talpo
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| | - Jessica Cazzola
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| | - Francesca Raffin
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| | - Chiara Cordiglieri
- Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Andrea Falqui
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (CIMaINa), Department of Physics, University of Milan, Milan, Italy
| | | | | | - Dario Besusso
- Department of Biosciences, University of Milan, Milan, Italy
- Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Gerardo Biella
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| | - Chiara Zuccato
- Department of Biosciences, University of Milan, Milan, Italy.
- Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy.
| |
Collapse
|
5
|
Li L, Zhuang L, Xu Z, Jiang L, Zhai Y, Liu D, Wu Q. U-shaped relationship between non-high-density lipoprotein cholesterol and cognitive impairment in Chinese middle-aged and elderly: a cross-sectional study. BMC Public Health 2024; 24:1624. [PMID: 38890653 PMCID: PMC11186169 DOI: 10.1186/s12889-024-19164-8] [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: 10/20/2023] [Accepted: 06/14/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND The relationship between blood lipids and cognitive function has long been a subject of interest, and the association between serum non-high-density lipoprotein cholesterol (non-HDL-C) levels and cognitive impairment remains contentious. METHODS We utilized data from the 2011 CHARLS national baseline survey, which after screening, included a final sample of 10,982 participants. Cognitive function was assessed using tests of episodic memory and cognitive intactness. We used multiple logistic regression models to estimate the relationship between non-HDL-C and cognitive impairment. Subsequently, utilizing regression analysis results from fully adjusted models, we explored the nonlinear relationship between non-HDL-C as well as cognitive impairment using smooth curve fitting and sought potential inflection points through saturation threshold effect analysis. RESULTS The results showed that each unit increase in non-HDL-C levels was associated with a 5.5% reduction in the odds of cognitive impairment (OR = 0.945, 95% CI: 0.897-0.996; p < 0.05). When non-HDL-C was used as a categorical variable, the results showed that or each unit increase in non-HDL-C levels, the odds of cognitive impairment were reduced by 14.2%, 20.9%, and 24% in the Q2, Q3, and Q4 groups, respectively, compared with Q1. In addition, in the fully adjusted model, analysis of the potential nonlinear relationship by smoothed curve fitting and saturation threshold effects revealed a U-shaped relationship between non-HDL-C and the risk of cognitive impairment, with an inflection point of 4.83. Before the inflection point, each unit increase in non-HDL-C levels was associated with a 12.3% decrease in the odds of cognitive impairment. After the tipping point, each unit increase in non-HDL-C levels was associated with an 18.8% increase in the odds of cognitive impairment (All p < 0.05). CONCLUSION There exists a U-shaped relationship between non-HDL-C and the risk of cognitive impairment in Chinese middle-aged and elderly individuals, with statistical significance on both sides of the turning points. This suggests that both lower and higher levels of serum non-high-density lipoprotein cholesterol increase the risk of cognitive impairment in middle-aged and elderly individuals.
Collapse
Affiliation(s)
- Lei Li
- Clinical Laboratory, The First Affiliated Hospital of Wannan Medical College, No. 2, West Zheshan Road, Wuhu, Anhui, 241001, China
| | - Lingdan Zhuang
- Clinical Laboratory, The First Affiliated Hospital of Wannan Medical College, No. 2, West Zheshan Road, Wuhu, Anhui, 241001, China
| | - Zichen Xu
- Clinical Laboratory, The First Affiliated Hospital of Wannan Medical College, No. 2, West Zheshan Road, Wuhu, Anhui, 241001, China
| | - Luqing Jiang
- Clinical Laboratory, The First Affiliated Hospital of Wannan Medical College, No. 2, West Zheshan Road, Wuhu, Anhui, 241001, China
| | - Ying Zhai
- Clinical Laboratory, The First Affiliated Hospital of Wannan Medical College, No. 2, West Zheshan Road, Wuhu, Anhui, 241001, China
| | - Daoqin Liu
- Department of Kidney Medicine, The First Affiliated Hospital of Wannan Medical College, No. 2, West Zheshan Road, Wuhu, Anhui, 241001, China.
| | - Qiwen Wu
- Clinical Laboratory, The First Affiliated Hospital of Wannan Medical College, No. 2, West Zheshan Road, Wuhu, Anhui, 241001, China.
| |
Collapse
|
6
|
D’Ambrosio A, Bressan D, Ferracci E, Carbone F, Mulè P, Rossi F, Barbieri C, Sorrenti E, Fiaccadori G, Detone T, Vezzoli E, Bianchi S, Sartori C, Corso S, Fukuda A, Bertalot G, Falqui A, Barbareschi M, Romanel A, Pasini D, Chiacchiera F. Increased genomic instability and reshaping of tissue microenvironment underlie oncogenic properties of Arid1a mutations. SCIENCE ADVANCES 2024; 10:eadh4435. [PMID: 38489371 PMCID: PMC10942108 DOI: 10.1126/sciadv.adh4435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 02/12/2024] [Indexed: 03/17/2024]
Abstract
Oncogenic mutations accumulating in many chromatin-associated proteins have been identified in different tumor types. With a mutation rate from 10 to 57%, ARID1A has been widely considered a tumor suppressor gene. However, whether this role is mainly due to its transcriptional-related activities or its ability to preserve genome integrity is still a matter of intense debate. Here, we show that ARID1A is largely dispensable for preserving enhancer-dependent transcriptional regulation, being ARID1B sufficient and required to compensate for ARID1A loss. We provide in vivo evidence that ARID1A is mainly required to preserve genomic integrity in adult tissues. ARID1A loss primarily results in DNA damage accumulation, interferon type I response activation, and chronic inflammation leading to tumor formation. Our data suggest that in healthy tissues, the increased genomic instability that follows ARID1A mutations and the selective pressure imposed by the microenvironment might result in the emergence of aggressive, possibly immune-resistant, tumors.
Collapse
Affiliation(s)
- Alessandro D’Ambrosio
- Laboratory of stem cells and cancer genomics, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy
- SEMM, University of Milan, 20142 Milan, Italy
| | - Davide Bressan
- Laboratory of stem cells and cancer genomics, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy
| | - Elisa Ferracci
- Laboratory of stem cells and cancer genomics, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy
| | - Francesco Carbone
- Unità Operativa Multizonale di Anatomia Patologica, APSS, 38122 Trento, Italy
| | - Patrizia Mulè
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, 20139 Milan, Italy
| | - Federico Rossi
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, 20139 Milan, Italy
| | - Caterina Barbieri
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, 20139 Milan, Italy
| | - Elisa Sorrenti
- Laboratory of stem cells and cancer genomics, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy
| | - Gaia Fiaccadori
- Laboratory of stem cells and cancer genomics, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy
| | - Thomas Detone
- Unità Operativa Multizonale di Anatomia Patologica, APSS, 38122 Trento, Italy
| | - Elena Vezzoli
- Department of Biomedical sciences for Health, University of Milan, 20133 Milan, Italy
| | - Salvatore Bianchi
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia (IIT), 20139 Milan, Italy
| | - Chiara Sartori
- Unità Operativa Multizonale di Anatomia Patologica, APSS, 38122 Trento, Italy
| | - Simona Corso
- Department of Oncology, University of Torino, 10060 Candiolo, Italy
- Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy
| | - Akihisa Fukuda
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Giovanni Bertalot
- Unità Operativa Multizonale di Anatomia Patologica, APSS, 38122 Trento, Italy
- Centre for Medical Sciences–CISMed, University of Trento, 38122 Trento, Italy
| | - Andrea Falqui
- Department of Physics, University of Milan, 20133 Milan, Italy
| | - Mattia Barbareschi
- Unità Operativa Multizonale di Anatomia Patologica, APSS, 38122 Trento, Italy
- Centre for Medical Sciences–CISMed, University of Trento, 38122 Trento, Italy
| | - Alessandro Romanel
- Laboratory of Bioinformatics and Computational Genomics, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy
| | - Diego Pasini
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, 20139 Milan, Italy
- Department of Health Sciences, University of Milan, 20142 Milan, Italy
| | - Fulvio Chiacchiera
- Laboratory of stem cells and cancer genomics, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy
| |
Collapse
|
7
|
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
|
8
|
Birolini G, Valenza M, Ottonelli I, Talpo F, Minoli L, Cappelleri A, Bombaci M, Caccia C, Canevari C, Trucco A, Leoni V, Passoni A, Favagrossa M, Nucera MR, Colombo L, Paltrinieri S, Bagnati R, Duskey JT, Caraffi R, Vandelli MA, Taroni F, Salmona M, Scanziani E, Biella G, Ruozi B, Tosi G, Cattaneo E. Chronic cholesterol administration to the brain supports complete and long-lasting cognitive and motor amelioration in Huntington's disease. Pharmacol Res 2023; 194:106823. [PMID: 37336430 PMCID: PMC10463277 DOI: 10.1016/j.phrs.2023.106823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/14/2023] [Accepted: 06/16/2023] [Indexed: 06/21/2023]
Abstract
Evidence that Huntington's disease (HD) is characterized by impaired cholesterol biosynthesis in the brain has led to strategies to increase its level in the brain of the rapidly progressing R6/2 mouse model, with a positive therapeutic outcome. Here we tested the long-term efficacy of chronic administration of cholesterol to the brain of the slowly progressing zQ175DN knock-in HD mice in preventing ("early treatment") or reversing ("late treatment") HD symptoms. To do this we used the most advanced formulation of cholesterol loaded brain-permeable nanoparticles (NPs), termed hybrid-g7-NPs-chol, which were injected intraperitoneally. We show that one cycle of treatment with hybrid-g7-NPs-chol, administered in the presymptomatic ("early treatment") or symptomatic ("late treatment") stages is sufficient to normalize cognitive defects up to 5 months, as well as to improve other behavioral and neuropathological parameters. A multiple cycle treatment combining both early and late treatments ("2 cycle treatment") lasting 6 months generates therapeutic effects for more than 11 months, without severe adverse reactions. Sustained cholesterol delivery to the brain of zQ175DN mice also reduces mutant Huntingtin aggregates in both the striatum and cortex and completely normalizes synaptic communication in the striatal medium spiny neurons compared to saline-treated HD mice. Furthermore, through a meta-analysis of published and current data, we demonstrated the power of hybrid-g7-NPs-chol and other strategies able to increase brain cholesterol biosynthesis, to reverse cognitive decline and counteract the formation of mutant Huntingtin aggregates. These results demonstrate that cholesterol delivery via brain-permeable NPs is a therapeutic option to sustainably reverse HD-related behavioral decline and neuropathological signs over time, highlighting the therapeutic potential of cholesterol-based strategies in HD patients. DATA AVAILABILITY: This study does not include data deposited in public repositories. Data are available on request to the corresponding authors.
Collapse
Affiliation(s)
- Giulia Birolini
- Department of Biosciences, University of Milan, 20133 Milan, Italy; Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi", 20122 Milan, Italy
| | - Marta Valenza
- Department of Biosciences, University of Milan, 20133 Milan, Italy; Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi", 20122 Milan, Italy.
| | - Ilaria Ottonelli
- Nanotech Lab, Te.Far.T.I. Center, Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Francesca Talpo
- Department of Biology and Biotechnologies, University of Pavia, 27100 Pavia, Italy
| | - Lucia Minoli
- Pathology Department, Evotec, 37135 Verona, Italy; Mouse & Animal Pathology Lab (MAPLab), Fondazione UniMi, 20139 Milan, Italy
| | - Andrea Cappelleri
- Dipartimento di Medicina Veterinaria e Scienze Animali, Università degli Studi di Milano, 26900 Lodi, Italy; Mouse & Animal Pathology Lab (MAPLab), Fondazione UniMi, 20139 Milan, Italy
| | - Mauro Bombaci
- Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi", 20122 Milan, Italy
| | - Claudio Caccia
- Unit of Medical Genetics and Neurogenetics. Fondazione IRCCS Istituto Neurologico Carlo Besta, 20131 Milan, Italy
| | - Caterina Canevari
- Department of Biology and Biotechnologies, University of Pavia, 27100 Pavia, Italy
| | - Arianna Trucco
- Department of Biology and Biotechnologies, University of Pavia, 27100 Pavia, Italy
| | - Valerio Leoni
- Laboratory of Clinical Chemistry, Hospital Pio XI of Desio, ASST-Brianza and Department of Medicine and Surgery, University of Milano Bicocca, 20900 Monza, Italy
| | - Alice Passoni
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
| | - Monica Favagrossa
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
| | - Maria Rosaria Nucera
- Department of Biosciences, University of Milan, 20133 Milan, Italy; Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi", 20122 Milan, Italy
| | - Laura Colombo
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
| | - Saverio Paltrinieri
- Dipartimento di Medicina Veterinaria e Scienze Animali, Università degli Studi di Milano, 26900 Lodi, Italy
| | - Renzo Bagnati
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
| | - Jason Thomas Duskey
- Nanotech Lab, Te.Far.T.I. Center, Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Riccardo Caraffi
- Nanotech Lab, Te.Far.T.I. Center, Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Maria Angela Vandelli
- Nanotech Lab, Te.Far.T.I. Center, Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Franco Taroni
- Unit of Medical Genetics and Neurogenetics. Fondazione IRCCS Istituto Neurologico Carlo Besta, 20131 Milan, Italy
| | - Mario Salmona
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
| | - Eugenio Scanziani
- Dipartimento di Medicina Veterinaria e Scienze Animali, Università degli Studi di Milano, 26900 Lodi, Italy; Mouse & Animal Pathology Lab (MAPLab), Fondazione UniMi, 20139 Milan, Italy
| | - Gerardo Biella
- Department of Biology and Biotechnologies, University of Pavia, 27100 Pavia, Italy
| | - Barbara Ruozi
- Nanotech Lab, Te.Far.T.I. Center, Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Giovanni Tosi
- Nanotech Lab, Te.Far.T.I. Center, Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Elena Cattaneo
- Department of Biosciences, University of Milan, 20133 Milan, Italy; Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi", 20122 Milan, Italy.
| |
Collapse
|
9
|
Ferlazzo GM, Gambetta AM, Amato S, Cannizzaro N, Angiolillo S, Arboit M, Diamante L, Carbognin E, Romani P, La Torre F, Galimberti E, Pflug F, Luoni M, Giannelli S, Pepe G, Capocci L, Di Pardo A, Vanzani P, Zennaro L, Broccoli V, Leeb M, Moro E, Maglione V, Martello G. Genome-wide screening in pluripotent cells identifies Mtf1 as a suppressor of mutant huntingtin toxicity. Nat Commun 2023; 14:3962. [PMID: 37407555 DOI: 10.1038/s41467-023-39552-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/19/2023] [Indexed: 07/07/2023] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disorder caused by CAG-repeat expansions in the huntingtin (HTT) gene. The resulting mutant HTT (mHTT) protein induces toxicity and cell death via multiple mechanisms and no effective therapy is available. Here, we employ a genome-wide screening in pluripotent mouse embryonic stem cells (ESCs) to identify suppressors of mHTT toxicity. Among the identified suppressors, linked to HD-associated processes, we focus on Metal response element binding transcription factor 1 (Mtf1). Forced expression of Mtf1 counteracts cell death and oxidative stress caused by mHTT in mouse ESCs and in human neuronal precursor cells. In zebrafish, Mtf1 reduces malformations and apoptosis induced by mHTT. In R6/2 mice, Mtf1 ablates motor defects and reduces mHTT aggregates and oxidative stress. Our screening strategy enables a quick in vitro identification of promising suppressor genes and their validation in vivo, and it can be applied to other monogenic diseases.
Collapse
Affiliation(s)
- Giorgia Maria Ferlazzo
- Department of Molecular Medicine, Medical School, University of Padua, 35131, Padua, Italy
- Aptuit (Verona) S.r.l., an Evotec Company, Campus Levi-Montalcini, 37135, Verona, Italy
| | - Anna Maria Gambetta
- Department of Molecular Medicine, Medical School, University of Padua, 35131, Padua, Italy
- Department of Biology, University of Padova, Via U. Bassi 58B, 35131, Padua, Italy
| | - Sonia Amato
- Department of Biology, University of Padova, Via U. Bassi 58B, 35131, Padua, Italy
- Department of Neuroscience, University of Padova, Via Belzoni, 160, 35131, Padua, Italy
| | - Noemi Cannizzaro
- Department of Molecular Medicine, Medical School, University of Padua, 35131, Padua, Italy
| | - Silvia Angiolillo
- Department of Molecular Medicine, Medical School, University of Padua, 35131, Padua, Italy
| | - Mattia Arboit
- Department of Molecular Medicine, Medical School, University of Padua, 35131, Padua, Italy
| | - Linda Diamante
- Department of Biology, University of Padova, Via U. Bassi 58B, 35131, Padua, Italy
| | - Elena Carbognin
- Department of Biology, University of Padova, Via U. Bassi 58B, 35131, Padua, Italy
| | - Patrizia Romani
- Department of Molecular Medicine, Medical School, University of Padua, 35131, Padua, Italy
| | - Federico La Torre
- Department of Biology, University of Padova, Via U. Bassi 58B, 35131, Padua, Italy
| | - Elena Galimberti
- Max Perutz Laboratories Vienna, University of Vienna, Vienna Biocenter, Dr Bohr Gasse 9, 1030, Vienna, Austria
| | - Florian Pflug
- Max Perutz Laboratories Vienna, University of Vienna, Vienna Biocenter, Dr Bohr Gasse 9, 1030, Vienna, Austria
| | - Mirko Luoni
- Division of Neuroscience, San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Serena Giannelli
- Division of Neuroscience, San Raffaele Scientific Institute, 20132, Milan, Italy
| | | | | | | | - Paola Vanzani
- Department of Molecular Medicine, Medical School, University of Padua, 35131, Padua, Italy
| | - Lucio Zennaro
- Department of Molecular Medicine, Medical School, University of Padua, 35131, Padua, Italy
| | - Vania Broccoli
- Division of Neuroscience, San Raffaele Scientific Institute, 20132, Milan, Italy
- CNR Institute of Neuroscience, 20854, Vedrano al Lambro, Italy
| | - Martin Leeb
- Max Perutz Laboratories Vienna, University of Vienna, Vienna Biocenter, Dr Bohr Gasse 9, 1030, Vienna, Austria
| | - Enrico Moro
- Department of Molecular Medicine, Medical School, University of Padua, 35131, Padua, Italy
| | | | - Graziano Martello
- Department of Biology, University of Padova, Via U. Bassi 58B, 35131, Padua, Italy.
| |
Collapse
|
10
|
Tshilenge KT, Aguirre CG, Bons J, Gerencser AA, Basisty N, Song S, Rose J, Lopez-Ramirez A, Naphade S, Loureiro A, Battistoni E, Milani M, Wehrfritz C, Holtz A, Hetz C, Mooney SD, Schilling B, Ellerby LM. Proteomic Analysis of Huntington's Disease Medium Spiny Neurons Identifies Alterations in Lipid Droplets. Mol Cell Proteomics 2023; 22:100534. [PMID: 36958627 PMCID: PMC10165459 DOI: 10.1016/j.mcpro.2023.100534] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 03/15/2023] [Accepted: 03/19/2023] [Indexed: 03/25/2023] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disease caused by a CAG repeat expansion in the Huntingtin (HTT) gene. The resulting polyglutamine (polyQ) tract alters the function of the HTT protein. Although HTT is expressed in different tissues, the medium spiny projection neurons (MSNs) in the striatum are particularly vulnerable in HD. Thus, we sought to define the proteome of human HD patient-derived MSNs. We differentiated HD72 induced pluripotent stem cells and isogenic controls into MSNs and carried out quantitative proteomic analysis. Using data-dependent acquisitions with FAIMS for label-free quantification on the Orbitrap Lumos mass spectrometer, we identified 6,323 proteins with at least two unique peptides. Of these, 901 proteins were altered significantly more in the HD72-MSNs than in isogenic controls. Functional enrichment analysis of upregulated proteins demonstrated extracellular matrix and DNA signaling (DNA replication pathway, double-strand break repair, G1/S transition) with the highest significance. Conversely, processes associated with the downregulated proteins included neurogenesis-axogenesis, the brain-derived neurotrophic factor-signaling pathway, Ephrin-A: EphA pathway, regulation of synaptic plasticity, triglyceride homeostasis cholesterol, plasmid lipoprotein particle immune response, interferon-γ signaling, immune system major histocompatibility complex, lipid metabolism and cellular response to stimulus. Moreover, proteins involved in the formation and maintenance of axons, dendrites, and synapses (e.g., Septin protein members) were dysregulated in HD72-MSNs. Importantly, lipid metabolism pathways were altered, and using quantitative image, we found analysis that lipid droplets accumulated in the HD72-MSN, suggesting a deficit in the turnover of lipids possibly through lipophagy. Our proteomics analysis of HD72-MSNs identified relevant pathways that are altered in MSNs and confirm current and new therapeutic targets for HD.
Collapse
Affiliation(s)
| | - Carlos Galicia Aguirre
- The Buck Institute for Research on Aging, Novato, California, 94945, USA; University of Southern California, Leonard Davis School of Gerontology, 3715 McClintock Ave, Los Angeles, CA 90893, USA
| | - Joanna Bons
- The Buck Institute for Research on Aging, Novato, California, 94945, USA
| | - Akos A Gerencser
- The Buck Institute for Research on Aging, Novato, California, 94945, USA
| | - Nathan Basisty
- The Buck Institute for Research on Aging, Novato, California, 94945, USA; Translational Gerontology Branch, National Institute on Aging (NIA), NIH, Baltimore, Maryland, 21244, USA
| | - Sicheng Song
- Department of Biomedical Informatics and Medical Education, School of Medicine, University of Washington, Seattle, WA, 98109, USA
| | - Jacob Rose
- The Buck Institute for Research on Aging, Novato, California, 94945, USA
| | | | - Swati Naphade
- The Buck Institute for Research on Aging, Novato, California, 94945, USA
| | - Ashley Loureiro
- The Buck Institute for Research on Aging, Novato, California, 94945, USA
| | - Elena Battistoni
- The Buck Institute for Research on Aging, Novato, California, 94945, USA
| | - Mateus Milani
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile; Center for Geroscience, Brain Health and Metabolism (GERO), Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile
| | - Cameron Wehrfritz
- The Buck Institute for Research on Aging, Novato, California, 94945, USA
| | - Anja Holtz
- The Buck Institute for Research on Aging, Novato, California, 94945, USA
| | - Claudio Hetz
- The Buck Institute for Research on Aging, Novato, California, 94945, USA; Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile; Center for Geroscience, Brain Health and Metabolism (GERO), Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile
| | - Sean D Mooney
- Department of Biomedical Informatics and Medical Education, School of Medicine, University of Washington, Seattle, WA, 98109, USA
| | - Birgit Schilling
- The Buck Institute for Research on Aging, Novato, California, 94945, USA; University of Southern California, Leonard Davis School of Gerontology, 3715 McClintock Ave, Los Angeles, CA 90893, USA.
| | - Lisa M Ellerby
- The Buck Institute for Research on Aging, Novato, California, 94945, USA; University of Southern California, Leonard Davis School of Gerontology, 3715 McClintock Ave, Los Angeles, CA 90893, USA.
| |
Collapse
|
11
|
Lor YCM, Tsou MT, Tsai LW, Tsai SY. The factors associated with cognitive function among community-dwelling older adults in Taiwan. BMC Geriatr 2023; 23:116. [PMID: 36864383 PMCID: PMC9983251 DOI: 10.1186/s12877-023-03806-4] [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: 08/25/2022] [Accepted: 02/07/2023] [Indexed: 03/04/2023] Open
Abstract
BACKGROUND This research aimed to investigate the associations of anthropometric measurements, physiological parameters, chronic disease comorbidities, and social and lifestyle factors with cognitive function amongst community-dwelling older adults in Taiwan. METHODS This was an observational, cross-sectional study involving 4,578 participants at least 65 years old, recruited between January 2008 and December 2018 from the Annual Geriatric Health Examinations Program. Cognitive function was assessed using the short portable mental state questionnaire (SPMSQ). Multivariable logistic regression was done to analyze the factors associated with cognitive impairment. RESULTS Among the 4,578 participants, 103 people (2.3%) with cognitive impairment were identified. Associated factors were age (odds ratio (OR) = 1.16, 95% confidence interval (CI) = 1.13,1.20), male gender (OR = 0.39, 95% CI = 0.21,0.72), diabetes mellitus (DM) (OR = 1.70, 95% CI = 1.03, 2.82), hyperlipidemia (OR = 0.47, 95% CI = 0.25, 0.89), exercise (OR = 0.44, 95% CI = 0.34, 0.56), albumin (OR = 0.37, 95% CI = 0.15, 0.88), and high-density lipoprotein (HDL) (OR = 0.98, 95% CI = 0.97, 1.00). Whereas waistline, alcohol intake in recent six months, and hemoglobin was not significantly associated with cognitive impairment (all p > 0.05). CONCLUSIONS Our findings suggested that people with older age and a history of DM had a higher risk of cognitive impairment. Male gender, a history of hyperlipidemia, exercise, a high albumin level, and a high HDL level seemed to be associated with a lower risk of cognitive impairment amongst older adults.
Collapse
Affiliation(s)
- You-Chen Mary Lor
- Department of Family Medicine, Hsinchu MacKay Memorial Hospital, No. 690, Section 2, Guangfu Road, East District, Hsinchu, 300, Taiwan
| | - Meng-Ting Tsou
- Department of Family Medicine, MacKay Memorial Hospital, Taipei, Taiwan.,Department of Nursing and Management, MacKay Junior College of Medicine, New Taipei City, Taiwan
| | - Li-Wei Tsai
- Department of Surgical Oncology, National Taiwan University Cancer Center, Taipei, Taiwan.,Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Szu-Ying Tsai
- Department of Family Medicine, Hsinchu MacKay Memorial Hospital, No. 690, Section 2, Guangfu Road, East District, Hsinchu, 300, Taiwan.
| |
Collapse
|
12
|
Improvement of synaptic plasticity by nanoparticles and the related mechanisms: Applications and prospects. J Control Release 2022; 347:143-163. [PMID: 35513209 DOI: 10.1016/j.jconrel.2022.04.049] [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: 01/26/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 11/20/2022]
Abstract
Synaptic plasticity is an important basis of learning and memory and participates in brain network remodelling after different types of brain injury (such as that caused by neurodegenerative diseases, cerebral ischaemic injury, posttraumatic stress disorder (PTSD), and psychiatric disorders). Therefore, improving synaptic plasticity is particularly important for the treatment of nervous system-related diseases. With the rapid development of nanotechnology, increasing evidence has shown that nanoparticles (NPs) can cross the blood-brain barrier (BBB) in different ways, directly or indirectly act on nerve cells, regulate synaptic plasticity, and ultimately improve nerve function. Therefore, to better elucidate the effect of NPs on synaptic plasticity, we review evidence showing that NPs can improve synaptic plasticity by regulating different influencing factors, such as neurotransmitters, receptors, presynaptic membrane proteins and postsynaptic membrane proteins, and further discuss the possible mechanism by which NPs improve synaptic plasticity. We conclude that NPs can improve synaptic plasticity and restore the function of damaged nerves by inhibiting neuroinflammation and oxidative stress, inducing autophagy, and regulating ion channels on the cell membrane. By reviewing the mechanism by which NPs regulate synaptic plasticity and the applications of NPs for the treatment of neurological diseases, we also propose directions for future research in this field and provide an important reference for follow-up research.
Collapse
|
13
|
Kacher R, Mounier C, Caboche J, Betuing S. Altered Cholesterol Homeostasis in Huntington’s Disease. Front Aging Neurosci 2022; 14:797220. [PMID: 35517051 PMCID: PMC9063567 DOI: 10.3389/fnagi.2022.797220] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 03/18/2022] [Indexed: 12/25/2022] Open
Abstract
Huntington’s disease (HD) is an autosomal dominant genetic disorder caused by an expansion of the CAG repeat in the first exon of Huntingtin’s gene. The associated neurodegeneration mainly affects the striatum and the cortex at early stages and progressively spreads to other brain structures. Targeting HD at its earlier stages is under intense investigation. Numerous drugs were tested, with a rate of success of only 3.5% approved molecules used as symptomatic treatment. The restoration of cholesterol metabolism, which is central to the brain homeostasis and strongly altered in HD, could be an interesting disease-modifying strategy. Cholesterol is an essential membrane component in the central nervous system (CNS); alterations of its homeostasis have deleterious consequences on neuronal functions. The levels of several sterols, upstream of cholesterol, are markedly decreased within the striatum of HD mouse model. Transcription of cholesterol biosynthetic genes is reduced in HD cell and mouse models as well as post-mortem striatal and cortical tissues from HD patients. Since the dynamic of brain cholesterol metabolism is complex, it is essential to establish the best method to target it in HD. Cholesterol, which does not cross the blood-brain-barrier, is locally synthesized and renewed within the brain. All cell types in the CNS synthesize cholesterol during development but as they progress through adulthood, neurons down-regulate their cholesterol synthesis and turn to astrocytes for their full supply. Cellular levels of cholesterol reflect the dynamic balance between synthesis, uptake and export, all integrated into the context of the cross talk between neurons and glial cells. In this review, we describe the latest advances regarding the role of cholesterol deregulation in neuronal functions and how this could be a determinant factor in neuronal degeneration and HD progression. The pathways and major mechanisms by which cholesterol and sterols are regulated in the CNS will be described. From this overview, we discuss the main clinical strategies for manipulating cholesterol metabolism in the CNS, and how to reinstate a proper balance in HD.
Collapse
Affiliation(s)
- Radhia Kacher
- Institut du Cerveau - Paris Brain Institute (ICM), AP-HP, INSERM, CNRS, University Hospital Pitié-Salpêtrière, Sorbonne Université, Paris, France
- INSERM, U1216, Grenoble Institut Neurosciences, Université Grenoble Alpes, Grenoble, France
| | - Coline Mounier
- Neuroscience Paris Seine, Institut de Biologie Paris-Seine, Faculté des Sciences et Ingénierie, Sorbonne Université, Paris, France
- Centre National de la Recherche Scientifique, UMR 8246, Paris, France
- U1130, Institut National de la Santé et de la Recherche Médicale, Paris, France
| | - Jocelyne Caboche
- Neuroscience Paris Seine, Institut de Biologie Paris-Seine, Faculté des Sciences et Ingénierie, Sorbonne Université, Paris, France
- Centre National de la Recherche Scientifique, UMR 8246, Paris, France
- U1130, Institut National de la Santé et de la Recherche Médicale, Paris, France
| | - Sandrine Betuing
- Neuroscience Paris Seine, Institut de Biologie Paris-Seine, Faculté des Sciences et Ingénierie, Sorbonne Université, Paris, France
- Centre National de la Recherche Scientifique, UMR 8246, Paris, France
- U1130, Institut National de la Santé et de la Recherche Médicale, Paris, France
- *Correspondence: Sandrine Betuing,
| |
Collapse
|
14
|
Liu H, Zou L, Zhou R, Zhang M, Gu S, Zheng J, Hukportie DN, Wu K, Huang Z, Yuan Z, Wu X. Long-Term Increase in Cholesterol Is Associated With Better Cognitive Function: Evidence From a Longitudinal Study. Front Aging Neurosci 2021; 13:691423. [PMID: 34220488 PMCID: PMC8248815 DOI: 10.3389/fnagi.2021.691423] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 05/18/2021] [Indexed: 12/16/2022] Open
Abstract
Background: Higher visit-to-visit cholesterol has been associated with cognitive decline. However, the association between long-term increase or decrease in cholesterol and cognitive decline remains unclear. Methods: A total of 4,915 participants aged ≥45 years with normal cognition in baseline were included. The participants were divided into four groups, namely low-low, low-high, high-low, and high-high, according to the diagnostic thresholds of total cholesterol (TC), non-high-density lipoprotein cholesterol (NHDL-C), low-density lipoprotein cholesterol, and high-density lipoprotein cholesterol (HDL-C) after 4 years of follow-up. Cognitive function was assessed by episodic memory and mental intactness. Binary logistic regression was used to analyse the association of cholesterol variation with cognitive decline. Results: Among the participants, 979 (19.9%) experienced global cognitive decline. The odds ratio (OR) of global cognitive and memory function decline were remarkably lower in participants in the low-high NHDL-C group than those in the low-low group [OR and 95% confidence interval (CI): 0.50 [0.26-0.95] for global cognitive decline, 0.45 [0.25-0.82] for memory function decline]. The lower OR was also significant in females (OR [95% CI]: 0.38 [0.17-0.87] for global cognitive decline; 0.44 [0.19-0.97] for memory function decline) and participants without cardiovascular disease (OR [95% CI]: 0.31 [0.11-0.87] for global cognitive decline; 0.34 [0.14-0.83] for memory function decline). The increases in other cholesterol were also negatively associated with the risk of cognitive decline although not significantly. Conclusions: A longitudinal increase in NHDL-C may be protective for cognition in females or individuals without cardiovascular disease.
Collapse
Affiliation(s)
- Huamin Liu
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Lianwu Zou
- Department of Psychiatry, Baiyun Psychiatric Rehabilitation Hospital, Guangzhou, China
| | - Rui Zhou
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Minyi Zhang
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Shanyuan Gu
- Inpatient Department, Baiyun Psychiatric Rehabilitation Hospital, Guangzhou, China
| | - Jiazhen Zheng
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Daniel Nyarko Hukportie
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Keyi Wu
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Zhiwei Huang
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Zelin Yuan
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xianbo Wu
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| |
Collapse
|
15
|
Birolini G, Verlengia G, Talpo F, Maniezzi C, Zentilin L, Giacca M, Conforti P, Cordiglieri C, Caccia C, Leoni V, Taroni F, Biella G, Simonato M, Cattaneo E, Valenza M. SREBP2 gene therapy targeting striatal astrocytes ameliorates Huntington's disease phenotypes. Brain 2021; 144:3175-3190. [PMID: 33974044 DOI: 10.1093/brain/awab186] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 03/18/2021] [Accepted: 04/23/2021] [Indexed: 11/14/2022] Open
Abstract
Brain cholesterol is produced mainly by astrocytes and is important for neuronal function. Its biosynthesis is severely reduced in mouse models of Huntington's disease. One possible mechanism is a diminished nuclear translocation of the transcription factor sterol regulatory element binding protein 2 (SREBP2) and, consequently, reduced activation of SREBP-controlled genes in the cholesterol biosynthesis pathway. Here we evaluated the efficacy of a gene therapy based on the unilateral intra-striatal injection of a recombinant adeno-associated virus 2/5 (AAV2/5) targeting astrocytes specifically and carrying the transcriptionally active N-terminal fragment of human SREBP2. Robust hSREBP2 expression in striatal glial cells in R6/2 Huntington's disease mice activated the transcription of cholesterol biosynthesis pathway genes, restored synaptic transmission, reversed Drd2 transcript levels decline, cleared mutant Huntingtin aggregates and attenuated behavioral deficits. We conclude that glial SREBP2 participates in Huntington's disease brain pathogenesis in vivo and that AAV-based delivery of SREBP2 to astrocytes counteracts key features of the disease.
Collapse
Affiliation(s)
- Giulia Birolini
- Department of Biosciences, University of Milan, 20133, Milan, Italy.,Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi″, 20122, Milan, Italy
| | - Gianluca Verlengia
- Division of Neuroscience, IRCCS San Raffaele Hospital, 20132, Milan, Italy.,Department of BioMedical Sciences, Section of Pharmacology, University of Ferrara, 44121, Ferrara, Italy
| | - Francesca Talpo
- Department of Biology and Biotechnologies, University of Pavia, 27100, Pavia, Italy
| | - Claudia Maniezzi
- Department of Biology and Biotechnologies, University of Pavia, 27100, Pavia, Italy
| | - Lorena Zentilin
- International Centre for Genetic Engineering and Biotechnology, ICGEB, 34149, Trieste, Italy
| | - Mauro Giacca
- International Centre for Genetic Engineering and Biotechnology, ICGEB, 34149, Trieste, Italy.,School of Cardiovascular Medicine and Sciences, King's College London, SE5 9NU, UK
| | - Paola Conforti
- Department of Biosciences, University of Milan, 20133, Milan, Italy.,Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi″, 20122, Milan, Italy
| | - Chiara Cordiglieri
- Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi″, 20122, Milan, Italy
| | - Claudio Caccia
- Unit of Medical Genetics and Neurogenetics. Fondazione I.R.C.C.S. Istituto Neurologico Carlo Besta, 20131 Milan, Italy
| | - Valerio Leoni
- School of Medicine and Surgery, University of Milano-Bicocca, 20900, Monza, Italy.,Laboratory of Clinical Pathology, Hospital of Desio, ASST Monza, 20900, Monza, Italy
| | - Franco Taroni
- Unit of Medical Genetics and Neurogenetics. Fondazione I.R.C.C.S. Istituto Neurologico Carlo Besta, 20131 Milan, Italy
| | - Gerardo Biella
- Department of Biology and Biotechnologies, University of Pavia, 27100, Pavia, Italy
| | - Michele Simonato
- Division of Neuroscience, IRCCS San Raffaele Hospital, 20132, Milan, Italy.,Department of BioMedical Sciences, Section of Pharmacology, University of Ferrara, 44121, Ferrara, Italy
| | - Elena Cattaneo
- Department of Biosciences, University of Milan, 20133, Milan, Italy.,Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi″, 20122, Milan, Italy
| | - Marta Valenza
- Department of Biosciences, University of Milan, 20133, Milan, Italy.,Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi″, 20122, Milan, Italy
| |
Collapse
|