1
|
Incontro S, Musella ML, Sammari M, Di Scala C, Fantini J, Debanne D. Lipids shape brain function through ion channel and receptor modulations: physiological mechanisms and clinical perspectives. Physiol Rev 2025; 105:137-207. [PMID: 38990068 DOI: 10.1152/physrev.00004.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/28/2024] [Accepted: 07/01/2024] [Indexed: 07/12/2024] Open
Abstract
Lipids represent the most abundant molecular type in the brain, with a fat content of ∼60% of the dry brain weight in humans. Despite this fact, little attention has been paid to circumscribe the dynamic role of lipids in brain function and disease. Membrane lipids such as cholesterol, phosphoinositide, sphingolipids, arachidonic acid, and endocannabinoids finely regulate both synaptic receptors and ion channels that ensure critical neural functions. After a brief introduction on brain lipids and their respective properties, we review here their role in regulating synaptic function and ion channel activity, action potential propagation, neuronal development, and functional plasticity and their contribution in the development of neurological and neuropsychiatric diseases. We also provide possible directions for future research on lipid function in brain plasticity and diseases.
Collapse
Affiliation(s)
| | | | - Malika Sammari
- UNIS, INSERM, Aix-Marseille Université, Marseille, France
| | | | | | | |
Collapse
|
2
|
Loiola RA, Nguyen C, Dib S, Saint-Pol J, Dehouck L, Sevin E, Naudot M, Landry C, Pahnke J, Pot C, Gosselet F. 25-Hydroxycholesterol attenuates tumor necrosis factor alpha-induced blood-brain barrier breakdown in vitro. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167479. [PMID: 39181516 DOI: 10.1016/j.bbadis.2024.167479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 08/05/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024]
Abstract
Intracellular cholesterol metabolism is regulated by the SREBP-2 and LXR signaling pathways. The effects of inflammation on these molecular mechanisms remain poorly studied, especially at the blood-brain barrier (BBB) level. Tumor necrosis factor α (TNFα) is a proinflammatory cytokine associated with BBB dysfunction. Therefore, the aim of our study was to investigate the effects of TNFα on BBB cholesterol metabolism, focusing on its underlying signaling pathways. Using a human in vitro BBB model composed of human brain-like endothelial cells (hBLECs) and brain pericytes (HBPs), we observed that TNFα increases BBB permeability by degrading the tight junction protein CLAUDIN-5 and activating stress signaling pathways in both cell types. TNFα also promotes cholesterol release and decreases cholesterol accumulation and APOE secretion. In hBLECs, the expression of SREBP-2 targets (LDLR and HMGCR) is increased, while ABCA1 expression is decreased. In HBPs, only LDLR and ABCA1 expression is increased. TNFα treatment also induces 25-hydroxycholesterol (25-HC) production, a cholesterol metabolite involved in the immune response and intracellular cholesterol metabolism. 25-HC pretreatment attenuates TNFα-induced BBB leakage and partially alleviates the effects of TNFα on ABCA1, LDLR, and HMGCR expression. Overall, our results suggest that TNFα favors cholesterol efflux via an LXR/ABCA1-independent mechanism at the BBB, while it activates the SREBP-2 pathway. Treatment with 25-HC partially reversed the effect of TNFα on the LXR/SREBP-2 pathways. Our study provides novel perspectives for better understanding cerebrovascular signaling events linked to BBB dysfunction and cholesterol metabolism in neuroinflammatory diseases.
Collapse
Affiliation(s)
- Rodrigo Azevedo Loiola
- University of Artois, UR2465, Blood-Brain Barrier (BBB) Laboratory, F-62300 Lens, France
| | - Cindy Nguyen
- University of Artois, UR2465, Blood-Brain Barrier (BBB) Laboratory, F-62300 Lens, France
| | - Shiraz Dib
- University of Artois, UR2465, Blood-Brain Barrier (BBB) Laboratory, F-62300 Lens, France
| | - Julien Saint-Pol
- University of Artois, UR2465, Blood-Brain Barrier (BBB) Laboratory, F-62300 Lens, France
| | - Lucie Dehouck
- University of Artois, UR2465, Blood-Brain Barrier (BBB) Laboratory, F-62300 Lens, France
| | - Emmanuel Sevin
- University of Artois, UR2465, Blood-Brain Barrier (BBB) Laboratory, F-62300 Lens, France
| | - Marie Naudot
- Plateforme d'Ingénierie Cellulaire & Analyses des Protéines ICAP, FR CNRS 3085 ICP, Université de Picardie Jules Verne, F-80039 Amiens, France
| | - Christophe Landry
- University of Artois, UR2465, Blood-Brain Barrier (BBB) Laboratory, F-62300 Lens, France
| | - Jens Pahnke
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO), Section of Neuropathology Research, Department of Pathology (PAT), Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, NO-0372 Oslo, Norway; Institute of Nutritional Medicine (INUM)/Lübeck Institute of Dermatology (LIED), University of Lübeck (UzL), University Medical Center Schleswig-Holstein (UKSH), Ratzeburger Allee 160, D-23538 Lübeck, Germany; Department of Pharmacology, Faculty of Medicine and Life Sciences, University of Latvia (LU), Jelgavas iela 3, LV-1004 Rīga, Latvia; School of Neurobiology, Biochemistry and Biophysics, The Georg S. Wise Faculty of Life Sciences, Tel Aviv University (TAU), Ramat Aviv, IL-6997801, Israel
| | - Caroline Pot
- Lausanne University Hospital (CHUV), University of Lausanne, Laboratories of Neuroimmunology, Service of Neurology and Neuroscience Research Center, Department of Clinical Neurosciences, CH-1011 Lausanne, Vaud, Switzerland
| | - Fabien Gosselet
- University of Artois, UR2465, Blood-Brain Barrier (BBB) Laboratory, F-62300 Lens, France.
| |
Collapse
|
3
|
Zhang B, Ban M, Chen X, Hu J, Cui L, Chen ZJ. Altered metabolic profiles in male offspring conceived from intracytoplasmic sperm injection. BMC Med 2024; 22:462. [PMID: 39402563 PMCID: PMC11476986 DOI: 10.1186/s12916-024-03654-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 09/25/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND While most research has focused on the association between intracytoplasmic sperm injection (ICSI) and neurodevelopmental disorders in children, relatively little attention has been given to its metabolic effects. Previous studies have reported that low serum lipid levels are associated with mental health problems. Our objective was to analyze the impact of ICSI on metabolic alterations compared to their in vitro fertilization (IVF) counterparts in male offspring, as well as its interaction with paternal overweight/obesity. METHODS We recruited families between January 2006 and December 2017 at the Center for Reproductive Medicine, Shandong University, China. Prospective data of offspring were obtained for body mass index (BMI), blood pressure, glucose, and lipid profile in their 0-11 years old. Linear mixed models were utilized to compute the mean difference and 95% confidence intervals (CI). RESULTS A total of 14,196 offspring visits were identified. In offspring aged 4-11 years, ICSI-conceived offspring exhibited significantly lower fasting glucose z-scores, total cholesterol z-scores, and low-density lipoprotein cholesterol (LDL-C) z-scores compared with their IVF counterparts (fasting glucose z-score: adjusted mean difference: - 0.13, 95% CI: - 0.23 to - 0.03; total cholesterol z-score: adjusted mean difference: - 0.13, 95% CI: - 0.23 to - 0.02; LDL-C z-score: adjusted mean difference: - 0.12, 95% CI: - 0.22 to - 0.01). Paternal overweight/obesity significantly influenced the relationship between ICSI and metabolic changes in offspring. In offspring born from fathers with overweight/obesity, ICSI-conceived offspring displayed significantly lower fasting glucose and total cholesterol z-scores than their IVF controls (fasting glucose z-score: adjusted mean difference: - 0.20, 95% CI: - 0.32 to - 0.08; total cholesterol z-score: adjusted mean difference: - 0.15, 95% CI: - 0.27 to - 0.02). In offspring born to fathers with normal weight, ICSI-conceived offspring showed significantly lower systolic blood pressure z-scores compared to those conceived via the IVF procedures (adjusted mean difference: - 0.21, 95% CI: - 0.37 to - 0.05). CONCLUSIONS The findings of this study suggested that ICSI was associated with altered glucose and lipid profiles compared to their IVF controls, characterized by lower fasting glucose z-scores, total cholesterol z-scores, and LDL-C z-scores. Encouraging fathers to reduce their body weight could potentially improve the metabolic health of their ICSI-conceived children.
Collapse
Affiliation(s)
- Bingqian Zhang
- Children and Reproductive Health, Institute of Women, Jinan, Shandong, 250012, China
- School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong, 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China
- ResearchUnit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan, Shandong, 250012, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250012, China
| | - Miaomiao Ban
- Children and Reproductive Health, Institute of Women, Jinan, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong, 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China
- ResearchUnit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan, Shandong, 250012, China
| | - Xiaojing Chen
- Children and Reproductive Health, Institute of Women, Jinan, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong, 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China
- ResearchUnit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan, Shandong, 250012, China
| | - Jingmei Hu
- Children and Reproductive Health, Institute of Women, Jinan, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong, 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China
- ResearchUnit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan, Shandong, 250012, China
| | - Linlin Cui
- The Second Hospital, Children and Reproductive Health, Institute of Women, Shandong University, Jinan, Shandong, 250012, China.
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, 250012, China.
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China.
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong, 250012, China.
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, 250012, China.
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, 250012, China.
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China.
- ResearchUnit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan, Shandong, 250012, China.
| | - Zi-Jiang Chen
- Children and Reproductive Health, Institute of Women, Jinan, Shandong, 250012, China
- The Second Hospital, Children and Reproductive Health, Institute of Women, Shandong University, Jinan, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong, 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China
- ResearchUnit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan, Shandong, 250012, China
- Department of Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200135, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, China
| |
Collapse
|
4
|
Kang S, Lee J, Ali DN, Choi S, Nesbitt J, Min PH, Trushina E, Choi DS. Low to moderate ethanol exposure reduces astrocyte-induced neuroinflammatory signaling and cognitive decline in presymptomatic APP/PS1 mice. Sci Rep 2024; 14:23989. [PMID: 39402264 PMCID: PMC11473946 DOI: 10.1038/s41598-024-75202-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 10/03/2024] [Indexed: 10/19/2024] Open
Abstract
Alcohol use disorder has been associated with the development of neurodegenerative diseases, including Alzheimer's disease (AD). However, recent studies demonstrate that moderate alcohol consumption may be protective against dementia and cognitive decline. We examined astrocyte function, low-density lipoprotein (LDL) receptor-related protein 1 (LRP1), and the NF-κB p65 and IKK-α/β signaling pathways in modulating neuroinflammation and amyloid beta (Aβ) deposition. We assessed apolipoprotein E (ApoE) in the brain of APP/PS1 mice using IHC and ELISA in response to low to moderate ethanol exposure (MEE). First, to confirm the intracerebral distribution of ApoE, we co-stained with GFAP, a marker for astrocytes that biosynthesize ApoE. We sought to investigate whether the ethanol-induced upregulation of LRP1 could potentially inhibit the activity of IL-1β and TNF-α induced IKK-α/β towards NF-κB p65, resulting in a reduction of pro-inflammatory cytokines. To evaluate the actual Aβ load in the brains of APP/PS1 mice, we performed with a specific antibody Aβ (Thioflavin S) on both air- and ethanol-exposed groups, subsequently analyzing Aβ levels. We also measured glucose uptake using 18F- fluorodeoxyglucose (FDG)-positron emission tomography (PET). Finally, we investigated whether MEE induced cognitive and memory changes using the Y maze, noble object recognition test, and Morris water maze. Our findings demonstrate that MEE reduced astrocytic glial fibrillary acidic protein (GFAP) and ApoE levels in the cortex and hippocampus in presymptomatic APP/PS1 mice. Interestingly, increased LRP1 protein expression was accompanied by dampening the IKK-α/β-NF-κB p65 pathway, resulting in decreased IL-1β and TNF-α levels in male mice. Notably, female mice show reduced levels of anti-inflammatory cytokines IL-4, and IL-10 without altering IL-1β and TNF-α concentrations. In both males and females, Aβ plaques, a hallmark of AD, were reduced in the cortex and hippocampus of APP/PS1 mice exposed to ethanol starting at pre-symptomatic stage. Consistently, MEE increased FDG-PET-based brain activities and normalized cognitive and memory deficits in the APP/PS1 mice. Our findings suggest that MEE may benefit AD pathology via modulating LRP1 expression, potentially reducing neuroinflammation and attenuating Aβ deposition. Our study implies that reduced astrocyte-derived ApoE and LDL cholesterol levels are critical for attenuating AD pathology.
Collapse
Affiliation(s)
- Shinwoo Kang
- Department of Molecular Pharmacology and Experimental Therapeutics, Clinic College of Medicine, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Pharmacology College of Medicine, Soonchunhyang University, 22 Soonchunhyango-ro, Ansan, Chungcheongnam-do, 31508, South Korea
| | - Jeyeon Lee
- Department of Radiology, Mayo Clinic College of Medicine and Science, 200 First Street SW, Rochester, MN, 55905, USA
| | - Dina N Ali
- Department of Molecular Pharmacology and Experimental Therapeutics, Clinic College of Medicine, 200 First Street SW, Rochester, MN, 55905, USA
| | - Sun Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Clinic College of Medicine, 200 First Street SW, Rochester, MN, 55905, USA
| | - Jarred Nesbitt
- Department of Neurology, Mayo Clinic College of Medicine and Science, 200 First Street SW, Rochester, MN, 55905, USA
| | - Paul H Min
- Department of Radiology, Mayo Clinic College of Medicine and Science, 200 First Street SW, Rochester, MN, 55905, USA
| | - Eugenia Trushina
- Department of Molecular Pharmacology and Experimental Therapeutics, Clinic College of Medicine, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Neurology, Mayo Clinic College of Medicine and Science, 200 First Street SW, Rochester, MN, 55905, USA
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Clinic College of Medicine, 200 First Street SW, Rochester, MN, 55905, USA.
- Department of Psychiatry and Psychology, Mayo Clinic College of Medicine and Science, 200 First Street SW, Rochester, MN, 55905, USA.
- Neuroscience Program, Mayo Clinic College of Medicine and Science, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
5
|
Parente M, Tonini C, Caputo S, Fiocchetti M, Pallottini V. Mechanisms of Sigma-2/TMEM97 Involvement in Cholesterol Metabolism. J Cell Biochem 2024; 125:e30645. [PMID: 39300897 DOI: 10.1002/jcb.30645] [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: 12/12/2023] [Revised: 08/21/2024] [Accepted: 08/25/2024] [Indexed: 09/22/2024]
Abstract
Extensive research has focused on cellular cholesterol and its regulation, primarily due to its crucial physiological roles and its association with numerous diseases resulting from dysregulated homeostasis. Consequently, investigating cholesterol metabolism and the network of regulating proteins remains an ongoing challenge for biomedical research seeking new molecular targets to manage aberrant cholesterol levels in pathologic conditions. There is evidence that Sigma-2/TMEM97 receptor regulates cholesterol metabolism. However, the mechanisms remain incompletely understood to date. Therefore, this study aimed to employ a pharmacological approach based on selective Sigma-2/TMEM97 agonists, rimcazole and siramesine, to uncover the contribution of this receptor to cholesterol homeostasis. Our results indicate that Sigma-2/TMEM97 activation modulates cholesterol uptake by altering key proteins involved in, leading to free cholesterol and neutral lipids accumulation. This sheds light on potential mechanisms implied, contributing a new piece to the intricate puzzle of cholesterol metabolism homeostasis.
Collapse
Grants
- This work has been supported by Together Strong NPC Foundation and Niemann-Pick Selbsthilfegruppe e.V. toVP, moreover by Excellence Departments, MUR-Italy, Grant/Award Numbers: ARTICOLO 1, COMMI 314-337 LEGGE 232/2016, ARTICOLO 1, awarded to Dept. of Science, University Roma Tre, Rome, Italy for 2023-2027, and Rome Technopole.
Collapse
Affiliation(s)
| | | | - Sara Caputo
- Department of Science, University Roma Tre, Rome, Italy
| | - Marco Fiocchetti
- Department of Science, University Roma Tre, Rome, Italy
- Neuroendocrinology Metabolism and Neuropharmacology Unit, IRCSS Fondazione Santa Lucia, Rome, Italy
| | - Valentina Pallottini
- Department of Science, University Roma Tre, Rome, Italy
- Neuroendocrinology Metabolism and Neuropharmacology Unit, IRCSS Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
6
|
Chalitsios CV, Ley H, Gao J, Turner MR, Thompson AG. Apolipoproteins, lipids, lipid-lowering drugs and risk of amyotrophic lateral sclerosis and frontotemporal dementia: a meta-analysis and Mendelian randomisation study. J Neurol 2024; 271:6956-6969. [PMID: 39230722 PMCID: PMC11447100 DOI: 10.1007/s00415-024-12665-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/05/2024]
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) have clinical, pathological and genetic overlapping. Lipid pathways are implicated in ALS. This study examined the effect of blood lipid levels on ALS, FTD risk, and survival in ALS. METHODS A systematic review and meta-analysis of high and low-density lipoprotein cholesterol (HDL-c and LDL-c), total cholesterol, triglycerides, apolipoproteins B and A1 levels with ALS was performed. Two-sample Mendelian randomisation (MR) analysis sought the causal effects of these exposures on ALS, FTD, and survival in ALS. The effect of lipid-lowering drugs was also examined using genetic proxies for targets of lipid-lowering medications. RESULTS Three cohort studies met the inclusion criteria for meta-analysis. Meta-analysis indicated an association between higher LDL-c (HRper mmol/L = 1.07, 95%CI:1.02-1.12;I 2 =18%) and lower HDL-c (HRper mmol/L = 0.83, 95%CI:0.74-0.94;I 2 =0%) with an increased risk of ALS. MR suggested causal effects of higher LDL-c (ORIVW = 1.085, 95%:CI 1.008-1.168, pFDR = 0.0406), total cholesterol (ORIVW = 1.081, 95%:CI 1.013-1.154, pFDR = 0.0458) and apolipoprotein B (ORIVW = 1.104, 95%:CI 1.041-1.171, pFDR = 0.0061) increasing ALS risk, and higher apolipoprotein B level increasing FTD risk (ORIVW = 1.424, 95%CI 1.072-1.829, pFDR = 0.0382). Reducing LDL-c through APOB inhibition was associated with lower ALS (ORIVW = 0.84, 95%CI 0.759-0.929, pFDR = 0.00275) and FTD risk (ORIVW = 0.581, 95%CI 0.387-0.874, pFDR = 0.0362). CONCLUSION These data support the influence of LDL-c and total cholesterol on ALS risk and apolipoprotein B on the risk of ALS and FTD. Potential APOB inhibition might decrease the risk of sporadic ALS and FTD. Further work in monogenic forms of ALS and FTD is necessary to determine whether blood lipids influence penetrance and phenotype.
Collapse
Affiliation(s)
- Christos V Chalitsios
- Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Level 6, West Wing, Oxford, OX3 9DU, UK
| | - Harriet Ley
- Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Level 6, West Wing, Oxford, OX3 9DU, UK
| | - Jiali Gao
- Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Level 6, West Wing, Oxford, OX3 9DU, UK
| | - Martin R Turner
- Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Level 6, West Wing, Oxford, OX3 9DU, UK
| | - Alexander G Thompson
- Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Level 6, West Wing, Oxford, OX3 9DU, UK.
| |
Collapse
|
7
|
Chandramouli A, Kamat SS. A Facile LC-MS Method for Profiling Cholesterol and Cholesteryl Esters in Mammalian Cells and Tissues. Biochemistry 2024; 63:2300-2309. [PMID: 38986142 DOI: 10.1021/acs.biochem.4c00160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
Cholesterol is central to mammalian lipid metabolism and serves many critical functions in the regulation of diverse physiological processes. Dysregulation in cholesterol metabolism is causally linked to numerous human diseases, and therefore, in vivo, the concentrations and flux of cholesterol and cholesteryl esters (fatty acid esters of cholesterol) are tightly regulated. While mass spectrometry has been an analytical method of choice for detecting cholesterol and cholesteryl esters in biological samples, the hydrophobicity, chemically inert nature, and poor ionization of these neutral lipids have often proved a challenge in developing lipidomics compatible liquid chromatography-mass spectrometry (LC-MS) methods to study them. To overcome this problem, here, we report a reverse-phase LC-MS method that is compatible with existing high-throughput lipidomics strategies and capable of identifying and quantifying cholesterol and cholesteryl esters from mammalian cells and tissues. Using this sensitive yet robust LC-MS method, we profiled different mammalian cell lines and tissues and provide a comprehensive picture of cholesterol and cholesteryl esters content in them. Specifically, among cholesteryl esters, we find that mammalian cells and tissues largely possess monounsaturated and polyunsaturated variants. Taken together, our lipidomics compatible LC-MS method to study this lipid class opens new avenues in understanding systemic and tissue-level cholesterol metabolism under various physiological conditions.
Collapse
Affiliation(s)
- Aakash Chandramouli
- Department of Biology, Indian Institute of Science Education and Research (IISER) Pune, Dr. Homi Bhabha Road, Pashan, Pune, Maharashtra 411008, India
| | - Siddhesh S Kamat
- Department of Biology, Indian Institute of Science Education and Research (IISER) Pune, Dr. Homi Bhabha Road, Pashan, Pune, Maharashtra 411008, India
| |
Collapse
|
8
|
Sánchez-Ramírez E, Ung TPL, Stringari C, Aguilar-Arnal L. Emerging Functional Connections Between Metabolism and Epigenetic Remodeling in Neural Differentiation. Mol Neurobiol 2024; 61:6688-6707. [PMID: 38340204 PMCID: PMC11339152 DOI: 10.1007/s12035-024-04006-w] [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: 09/13/2023] [Accepted: 01/30/2024] [Indexed: 02/12/2024]
Abstract
Stem cells possess extraordinary capacities for self-renewal and differentiation, making them highly valuable in regenerative medicine. Among these, neural stem cells (NSCs) play a fundamental role in neural development and repair processes. NSC characteristics and fate are intricately regulated by the microenvironment and intracellular signaling. Interestingly, metabolism plays a pivotal role in orchestrating the epigenome dynamics during neural differentiation, facilitating the transition from undifferentiated NSC to specialized neuronal and glial cell types. This intricate interplay between metabolism and the epigenome is essential for precisely regulating gene expression patterns and ensuring proper neural development. This review highlights the mechanisms behind metabolic regulation of NSC fate and their connections with epigenetic regulation to shape transcriptional programs of stemness and neural differentiation. A comprehensive understanding of these molecular gears appears fundamental for translational applications in regenerative medicine and personalized therapies for neurological conditions.
Collapse
Affiliation(s)
- Edgar Sánchez-Ramírez
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Thi Phuong Lien Ung
- Laboratory for Optics and Biosciences, Ecole Polytechnique, CNRS, INSERM, Institut Polytechnique de Paris, Palaiseau, France
| | - Chiara Stringari
- Laboratory for Optics and Biosciences, Ecole Polytechnique, CNRS, INSERM, Institut Polytechnique de Paris, Palaiseau, France
| | - Lorena Aguilar-Arnal
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico.
| |
Collapse
|
9
|
Ma J, Lin Z, Zhang Y, Ding Y, Tang Q, Qian Y, Jin B, Luo RY, Liao WL, Thyparambil S, Han Z, Chou CJ, Schilling J, Li Q, Zhang M, Lin Y, Ma Y, Sylvester KG, Nagpal S, McElhinney DB, Ling XB, Chen B. Targeted multiplex validation of CSF proteomic biomarkers: implications for differentiation of PCNSL from tumor-free controls and other brain tumors. Front Immunol 2024; 15:1343109. [PMID: 39144147 PMCID: PMC11322575 DOI: 10.3389/fimmu.2024.1343109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 07/17/2024] [Indexed: 08/16/2024] Open
Abstract
Introduction Primary central nervous system lymphoma (PCNSL) is a rare type of non-Hodgkin's lymphoma that affects brain parenchyma, eyes, cerebrospinal fluid, and spinal cord. Diagnosing PCNSL can be challenging because imaging studies often show similar patterns as other brain tumors, and stereotactic brain lesion biopsy conformation is invasive and not always possible. This study aimed to validate a previous proteomic profiling (PMID: 32610669) of cerebrospinal fluid (CSF) and develop a CSF-based proteomic panel for accurate PCNSL diagnosis and differentiation. Methods CSF samples were collected from patients of 30 PCNSL, 30 other brain tumors, and 31 tumor-free/benign controls. Liquid chromatography tandem-mass spectrometry targeted proteomics analysis was used to establish CSF-based proteomic panels. Results Final proteomic panels were selected and optimized to diagnose PCNSL from tumor-free controls or other brain tumor lesions with an area under the curve (AUC) of 0.873 (95%CI: 0.723-0.948) and 0.937 (95%CI: 0.807- 0.985), respectively. Pathways analysis showed diagnosis panel features were significantly enriched in pathways related to extracellular matrices-receptor interaction, focal adhesion, and PI3K-Akt signaling, while prion disease, mineral absorption and HIF-1 signaling were significantly enriched with differentiation panel features. Discussion This study suggests an accurate clinical test panel for PCNSL diagnosis and differentiation with CSF-based proteomic signatures, which may help overcome the challenges of current diagnostic methods and improve patient outcomes.
Collapse
Affiliation(s)
- Jingjing Ma
- Department of Hematology, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhiguang Lin
- Department of Hematology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yaqi Zhang
- College of Automation, Guangdong Polytechnic Normal University, Guangzhou, China
| | - Yun Ding
- Research and Development, mProbe Inc.Palo Alto, CA, United States
| | - Qiming Tang
- Research and Development, mProbe Inc.Palo Alto, CA, United States
| | - Yufeng Qian
- Research and Development, mProbe Inc.Palo Alto, CA, United States
| | - Bo Jin
- Research and Development, mProbe Inc.Palo Alto, CA, United States
| | - Ruben Y. Luo
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States
| | - Wei-Li Liao
- Research and Development, mProbe Inc.Palo Alto, CA, United States
| | | | - Zhi Han
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, United States
| | - C. James Chou
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - James Schilling
- Research and Development, mProbe Inc.Palo Alto, CA, United States
| | - Qing Li
- Department of Hematology, Huashan Hospital, Fudan University, Shanghai, China
| | - Mengxue Zhang
- Department of Hematology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yunan Lin
- Department of Hematology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yan Ma
- Department of Hematology, Huashan Hospital, Fudan University, Shanghai, China
| | - Karl G. Sylvester
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Seema Nagpal
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, United States
| | - Doff B. McElhinney
- Departments of Cardiothoracic Surgery and Pediatrics (Cardiology), Stanford University School of Medicine, Stanford, CA, United States
| | - Xuefeng B. Ling
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Bobin Chen
- Department of Hematology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
10
|
Pan Y, Liang J, Zhang W, Gao D, Li C, Xie W, Zheng F. Association between Age at Diagnosis of Hyperlipidemia and Subsequent Risk of Dementia. J Am Med Dir Assoc 2024; 25:104960. [PMID: 38453136 DOI: 10.1016/j.jamda.2024.01.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/30/2024] [Accepted: 01/30/2024] [Indexed: 03/09/2024]
Abstract
OBJECTIVES The relationship between age at diagnosis of hyperlipidemia and dementia remains unclear. We examined whether younger age at diagnosis of hyperlipidemia is associated with higher risk of subsequent dementia. DESIGN A longitudinal population-based study with a median follow-up of 12.8 years. SETTING AND PARTICIPANTS We analyzed data on a sample of 489,642 participants in the United Kingdom. METHODS This study was based on the UK Biobank. Information on hyperlipidemia and dementia diagnoses was collected at baseline (2006-2010) and follow-up [median = 12.8 years, interquartile range (IQR): 12.1-13.6 years]. Propensity score matching method and Cox proportional hazards models were used to assess the association between age at diagnosis of hyperlipidemia and dementia. RESULTS Among 489,642 participants (mean age: 56.9 ± 8.1 years; female: 54.7%), 114,112 (23.3%) were diagnosed with hyperlipidemia. Younger age at diagnosis of hyperlipidemia (per 10-year decrease) was significantly associated with higher risks of all-cause dementia [hazard ratio (HR), 1.12; 95% CI, 1.07-1.18; P < .001], Alzheimer's disease (AD) (HR, 1.22; 95% CI, 1.14-1.31; P < .001), and vascular dementia (VD) (HR, 1.16; 95% CI, 1.05-1.27; P < .001). After propensity score matching, patients with hyperlipidemia diagnosed before 50 years had the highest HR for all-cause dementia (HR, 1.46; 95% CI, 1.15-1.86; P = .002), followed by patients diagnosed between 50 and 69 years (HR, 1.21; 95% CI, 1.12-1.31; P < .001) and then patients diagnosed aged 70 years and older (HR, 0.94; 95% CI, 0.84-1.06; P = .302). Similar results were observed for AD and VD. CONCLUSIONS AND IMPLICATIONS A dose-response relationship between age at hyperlipidemia diagnosis and risk of dementia was found in the longitudinal cohort study, with younger age at diagnosis of hyperlipidemia being associated with higher subsequent risk.
Collapse
Affiliation(s)
- Yang Pan
- School of Nursing, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie Liang
- School of Nursing, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenya Zhang
- School of Nursing, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Darui Gao
- Peking University Clinical Research Institute, Peking University First Hospital, Beijing, China
| | - Chenglong Li
- Peking University Clinical Research Institute, Peking University First Hospital, Beijing, China
| | - Wuxiang Xie
- Peking University Clinical Research Institute, Peking University First Hospital, Beijing, China; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China.
| | - Fanfan Zheng
- School of Nursing, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
11
|
Phua YL, D'Annibale OM, Karunanidhi A, Mohsen AW, Kirmse B, Dobrowolski SF, Vockley J. A multiomics approach reveals evidence for phenylbutyrate as a potential treatment for combined D,L-2- hydroxyglutaric aciduria. Mol Genet Metab 2024; 142:108495. [PMID: 38772223 DOI: 10.1016/j.ymgme.2024.108495] [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: 02/01/2024] [Revised: 03/30/2024] [Accepted: 05/13/2024] [Indexed: 05/23/2024]
Abstract
PURPOSE To identify therapies for combined D, L-2-hydroxyglutaric aciduria (C-2HGA), a rare genetic disorder caused by recessive variants in the SLC25A1 gene. METHODS Patients C-2HGA were identified and diagnosed by whole exome sequencing and biochemical genetic testing. Patient derived fibroblasts were then treated with phenylbutyrate and the functional effects assessed by metabolomics and RNA-sequencing. RESULTS In this study, we demonstrated that C-2HGA patient derived fibroblasts exhibited impaired cellular bioenergetics. Moreover, Fibroblasts form one patient exhibited worsened cellular bioenergetics when supplemented with citrate. We hypothesized that treating patient cells with phenylbutyrate (PB), an FDA approved pharmaceutical drug that conjugates glutamine for renal excretion, would reduce mitochondrial 2-ketoglutarate, thereby leading to improved cellular bioenergetics. Metabolomic and RNA-seq analyses of PB-treated fibroblasts demonstrated a significant decrease in intracellular 2-ketoglutarate, 2-hydroxyglutarate, and in levels of mRNA coding for citrate synthase and isocitrate dehydrogenase. Consistent with the known action of PB, an increased level of phenylacetylglutamine in patient cells was consistent with the drug acting as 2-ketoglutarate sink. CONCLUSION Our pre-clinical studies suggest that citrate supplementation has the possibility exacerbating energy metabolism in this condition. However, improvement in cellular bioenergetics suggests phenylbutyrate might have interventional utility for this rare disease.
Collapse
MESH Headings
- Humans
- Phenylbutyrates/pharmacology
- Phenylbutyrates/therapeutic use
- Fibroblasts/metabolism
- Fibroblasts/drug effects
- Glutarates/metabolism
- Ketoglutaric Acids/metabolism
- Energy Metabolism/drug effects
- Energy Metabolism/genetics
- Mitochondria/drug effects
- Mitochondria/metabolism
- Mitochondria/genetics
- Metabolomics
- Exome Sequencing
- Citrate (si)-Synthase/metabolism
- Citrate (si)-Synthase/genetics
- Brain Diseases, Metabolic, Inborn/drug therapy
- Brain Diseases, Metabolic, Inborn/genetics
- Brain Diseases, Metabolic, Inborn/metabolism
- Isocitrate Dehydrogenase/genetics
- Isocitrate Dehydrogenase/metabolism
- Brain Diseases, Metabolic/drug therapy
- Brain Diseases, Metabolic/genetics
- Brain Diseases, Metabolic/metabolism
- Brain Diseases, Metabolic/pathology
- Multiomics
- Mitochondrial Proteins
- Organic Anion Transporters
Collapse
Affiliation(s)
- Yu Leng Phua
- Department of Pediatrics, Division of Genetic and Genomic Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA; Department of Pathology, Clinical Biochemical Genetics Laboratory, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Olivia M D'Annibale
- Department of Pediatrics, Division of Genetic and Genomic Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA; Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| | - Anuradha Karunanidhi
- Department of Pediatrics, Division of Genetic and Genomic Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Al-Walid Mohsen
- Department of Pediatrics, Division of Genetic and Genomic Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA; Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| | - Brian Kirmse
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS, USA
| | - Steven F Dobrowolski
- Department of Pathology, Clinical Biochemical Genetics Laboratory, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Jerry Vockley
- Department of Pediatrics, Division of Genetic and Genomic Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA; Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA.
| |
Collapse
|
12
|
Żulińska S, Strosznajder AK, Strosznajder JB. Current View on PPAR-α and Its Relation to Neurosteroids in Alzheimer's Disease and Other Neuropsychiatric Disorders: Promising Targets in a Therapeutic Strategy. Int J Mol Sci 2024; 25:7106. [PMID: 39000217 PMCID: PMC11241121 DOI: 10.3390/ijms25137106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/19/2024] [Accepted: 06/22/2024] [Indexed: 07/16/2024] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) may play an important role in the pathomechanism/pathogenesis of Alzheimer's disease (AD) and several other neurological/neuropsychiatric disorders. AD leads to progressive alterations in the redox state, ion homeostasis, lipids, and protein metabolism. Significant alterations in molecular processes and the functioning of several signaling pathways result in the degeneration and death of synapses and neuronal cells, leading to the most severe dementia. Peroxisome proliferator-activated receptor alpha (PPAR-α) is among the processes affected by AD; it regulates the transcription of genes related to the metabolism of cholesterol, fatty acids, other lipids and neurotransmission, mitochondria biogenesis, and function. PPAR-α is involved in the cholesterol transport to mitochondria, the substrate for neurosteroid biosynthesis. PPAR-α-coding enzymes, such as sulfotransferases, which are responsible for neurosteroid sulfation. The relation between PPAR-α and cholesterol/neurosteroids may have a significant impact on the course and progression of neurodegeneration/neuroprotection processes. Unfortunately, despite many years of intensive studies, the pathogenesis of AD is unknown and therapy for AD and other neurodegenerative diseases is symptomatic, presenting a significant goal and challenge today. This review presents recent achievements in therapeutic approaches for AD, which are targeting PPAR-α and its relation to cholesterol and neurosteroids in AD and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Sylwia Żulińska
- Department of Cellular Signaling, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego St., 02-106 Warsaw, Poland;
| | - Anna K. Strosznajder
- Department of Psychiatry, Medical University of Warsaw, Nowowiejska St. 27, 00-665 Warsaw, Poland;
| | - Joanna B. Strosznajder
- Department of Cellular Signaling, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego St., 02-106 Warsaw, Poland;
| |
Collapse
|
13
|
Rahman MM, Islam A, Mamun MA, Afroz MS, Nabi MM, Sakamoto T, Sato T, Kahyo T, Takahashi Y, Okino A, Setou M. Low-Temperature Plasma Pretreatment Enhanced Cholesterol Detection in Brain by Desorption Electrospray Ionization-Mass Spectrometry Imaging. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2024; 35:1227-1236. [PMID: 38778699 DOI: 10.1021/jasms.4c00045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Cholesterol is a primary lipid molecule in the brain that contains one-fourth of the total body cholesterol. Abnormal cholesterol homeostasis is associated with neurodegenerative disorders. Mass spectrometry imaging (MSI) technique is a powerful tool for studying lipidomics and metabolomics. Among the MSI techniques, desorption electrospray ionization-MSI (DESI-MSI) has been used advantageously to study brain lipidomics due to its soft and ambient ionization nature. However, brain cholesterol is poorly ionized. To this end, we have developed a new method for detecting brain cholesterol by DESI-MSI using low-temperature plasma (LTP) pretreatment as an ionization enhancement. In this method, the brain sections were treated with LTP for 1 and 2 min prior to DESI-MSI analyses. Interestingly, the MS signal intensity of cholesterol (at m/z 369.35 [M + H - H2O]+) was more than 2-fold higher in the 1 min LTP-treated brain section compared to the untreated section. In addition, we detected cholesterol, more specifically excluding isomers by targeted-DESI-MSI in multiple reaction monitoring (MRM) mode and similar results were observed: the signal intensity of each cholesterol transition (m/z 369.4 → 95.1, 109.1, 135.1, 147.1, and 161.1) was increased by more than 2-fold due to 1 min LTP treatment. Cholesterol showed characteristic distributions in the fiber tract region, including the corpus callosum and anterior commissure, anterior part of the brain where LTP markedly (p < 0.001) enhanced the cholesterol intensity. In addition, the distributions of some unknown analytes were exclusively detected in the LTP-treated section. Our study revealed LTP pretreatment as a potential strategy to ionize molecules that show poor ionization efficiency in the MSI technique.
Collapse
Affiliation(s)
- Md Muedur Rahman
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka 431-3192, Japan
- Preppers Co., Ltd., Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Ariful Islam
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka 431-3192, Japan
- Preppers Co., Ltd., Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka 431-3192, Japan
- Department of Biochemistry and Microbiology, School of Health and Life Sciences, North South University, Bashundhara, Dhaka 1229, Bangladesh
| | - Md Al Mamun
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka 431-3192, Japan
- Preppers Co., Ltd., Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Mst Sayela Afroz
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Md Mahamodun Nabi
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Takumi Sakamoto
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka 431-3192, Japan
- Preppers Co., Ltd., Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Tomohito Sato
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Tomoaki Kahyo
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka 431-3192, Japan
- Quantum Imaging Laboratory, International Mass Imaging and Spatial Omics Center, Institute of Photonics Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Yutaka Takahashi
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka 431-3192, Japan
- Preppers Co., Ltd., Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Akitoshi Okino
- Laboratory for Future Interdisciplinary Research of Science and Technology, Institute of Innovative Research, Tokyo Institute of Technology, J2-32, 4259 Nagatsuta, Midori-ku, Yokohama 226-8502, Japan
| | - Mitsutoshi Setou
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka 431-3192, Japan
- International Mass Imaging and Spatial Omics Center, Institute of Photonics Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu, Shizuoka 431-3192, Japan
| |
Collapse
|
14
|
Grenon MB, Papavergi MT, Bathini P, Sadowski M, Lemere CA. Temporal Characterization of the Amyloidogenic APPswe/PS1dE9;hAPOE4 Mouse Model of Alzheimer's Disease. Int J Mol Sci 2024; 25:5754. [PMID: 38891941 PMCID: PMC11172317 DOI: 10.3390/ijms25115754] [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: 05/04/2024] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Alzheimer's disease (AD) is a devastating disorder with a global prevalence estimated at 55 million people. In clinical studies administering certain anti-beta-amyloid (Aβ) antibodies, amyloid-related imaging abnormalities (ARIAs) have emerged as major adverse events. The frequency of these events is higher among apolipoprotein ε4 allele carriers (APOE4) compared to non-carriers. To reflect patients most at risk for vascular complications of anti-Aβ immunotherapy, we selected an APPswe/PS1dE9 transgenic mouse model bearing the human APOE4 gene (APPPS1:E4) and compared it with the same APP/PS1 mouse model bearing the human APOE3 gene (APOE ε3 allele; APPPS1:E3). Using histological and biochemical analyses, we characterized mice at three ages: 8, 12, and 16 months. Female and male mice were assayed for general cerebral fibrillar and pyroglutamate (pGlu-3) Aβ deposition, cerebral amyloid angiopathy (CAA), microhemorrhages, apoE and cholesterol composition, astrocytes, microglia, inflammation, lysosomal dysfunction, and neuritic dystrophy. Amyloidosis, lipid deposition, and astrogliosis increased with age in APPPS1:E4 mice, while inflammation did not reveal significant changes with age. In general, APOE4 carriers showed elevated Aβ, apoE, reactive astrocytes, pro-inflammatory cytokines, microglial response, and neuritic dystrophy compared to APOE3 carriers at different ages. These results highlight the potential of the APPPS1:E4 mouse model as a valuable tool in investigating the vascular side effects associated with anti-amyloid immunotherapy.
Collapse
Affiliation(s)
- Martine B. Grenon
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (M.B.G.); (M.-T.P.); (P.B.)
- Section Neuropsychology & Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Maria-Tzousi Papavergi
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (M.B.G.); (M.-T.P.); (P.B.)
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Praveen Bathini
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (M.B.G.); (M.-T.P.); (P.B.)
| | - Martin Sadowski
- Departments of Neurology, Psychiatry, and Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA;
| | - Cynthia A. Lemere
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (M.B.G.); (M.-T.P.); (P.B.)
| |
Collapse
|
15
|
Casas M, Dickson EJ. Unraveling the Connection: Cholesterol, Calcium Signaling, and Neurodegeneration. Neurosci Insights 2024; 19:26331055241252772. [PMID: 38737295 PMCID: PMC11088808 DOI: 10.1177/26331055241252772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 04/19/2024] [Indexed: 05/14/2024] Open
Abstract
Cholesterol and calcium play crucial roles as integral structural components and functional signaling entities within the central nervous system. Disruption in cholesterol homeostasis has been linked to Alzheimer's, Parkinson's, and Huntington's Disease while alterations in calcium signaling is hypothesized to be a key substrate for neurodegeneration across many disorders. Despite the importance of regulated cholesterol and calcium homeostasis for brain health there has been an absence of research investigating the interdependence of these signaling molecules and how they can tune each other's abundance at membranes to influence membrane identity. Here, we discuss the role of cholesterol in shaping calcium dynamics in a neurodegenerative disorder that arises due to mutations in the lysosomal cholesterol transporter, Niemann Pick Type C1 (NPC1). We discuss the molecular mechanisms through which altered lysosomal cholesterol transport influences calcium signaling pathways through remodeling of ion channel distribution at organelle-organelle membrane contacts leading to neurodegeneration. This scientific inquiry not only sheds light on NPC disease but also holds implications for comprehending other cholesterol-associated neurodegenerative disorders.
Collapse
Affiliation(s)
- Maria Casas
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA, USA
| | - Eamonn J Dickson
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA, USA
| |
Collapse
|
16
|
Alashmali S. Nutritional roles and therapeutic potentials of dietary sphingomyelin in brain diseases. J Clin Biochem Nutr 2024; 74:185-191. [PMID: 38799143 PMCID: PMC11111474 DOI: 10.3164/jcbn.23-97] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 12/12/2023] [Indexed: 05/29/2024] Open
Abstract
Sphingolipids have recently gained interest as potential players in variety of diseases due to their import roles in human body particularly, the brain. As sphingomyelin is the most common type of sphingolipids, deficits in its distribution to brain cells may contribute to neurological anomalies. However, data is limited regarding the impact of different levels of dietary sphingomyelin intake on neural function especially if this approach can boost cognition and prevent neurological disorders. This review evaluates the effect of dietary sphingomyelin and its metabolites (ceramide and sphingosine-1-phosphate) in animal models and in humans, with a primary focus on its impact on brain health. Additionally, it proposes multiple neuroenhancing effects of sphingomyelin-rich diet. This presents an opportunity to stimulate further research that aims to determine the therapeutic value of dietary sphingomyelin in preventing, improving or slowing the progression of central nervous system disorders.
Collapse
Affiliation(s)
- Shoug Alashmali
- Department of Clinical Nutrition, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
17
|
Brase L, Yu Y, McDade E, Harari O, Benitez BA. Comparative gene regulatory networks modulating APOE expression in microglia and astrocytes. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.19.24306098. [PMID: 38699303 PMCID: PMC11065001 DOI: 10.1101/2024.04.19.24306098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
Background Single-cell technologies have unveiled various transcriptional states in different brain cell types. Transcription factors (TFs) regulate the expression of related gene sets, thereby controlling these diverse expression states. Apolipoprotein E (APOE), a pivotal risk-modifying gene in Alzheimer's disease (AD), is expressed in specific glial transcriptional states associated with AD. However, it is still unknown whether the upstream regulatory programs that modulate its expression are shared across brain cell types or specific to microglia and astrocytes. Methods We used pySCENIC to construct state-specific gene regulatory networks (GRNs) for resting and activated cell states within microglia and astrocytes based on single-nucleus RNA sequencing data from AD patients' cortices from the Knight ADRC-DIAN cohort. We then identified replicating TF using data from the ROSMAP cohort. We identified sets of genes co-regulated with APOE by clustering the GRN target genes and identifying genes differentially expressed after the virtual knockout of TFs regulating APOE. We performed enrichment analyses on these gene sets and evaluated their overlap with genes found in AD GWAS loci. Results We identified an average of 96 replicating regulators for each microglial and astrocyte cell state. Our analysis identified the CEBP, JUN, FOS, and FOXO TF families as key regulators of microglial APOE expression. The steroid/thyroid hormone receptor families, including the THR TF family, consistently regulated APOE across astrocyte states, while CEBP and JUN TF families were also involved in resting astrocytes. AD GWAS-associated genes (PGRN, FCGR3A, CTSH, ABCA1, MARCKS, CTSB, SQSTM1, TSC22D4, FCER1G, and HLA genes) are co-regulated with APOE. We also uncovered that APOE-regulating TFs were linked to circadian rhythm (BHLHE40, DBP, XBP1, CREM, SREBF1, FOXO3, and NR2F1). Conclusions Our findings reveal a novel perspective on the transcriptional regulation of APOE in the human brain. We found a comprehensive and cell-type-specific regulatory landscape for APOE, revealing distinct and shared regulatory mechanisms across microglia and astrocytes, underscoring the complexity of APOE regulation. APOE-co-regulated genes might also affect AD risk. Furthermore, our study uncovers a potential link between circadian rhythm disruption and APOE regulation, shedding new light on the pathogenesis of AD.
Collapse
Affiliation(s)
- Logan Brase
- Department of Psychiatry, Washington University, Saint Louis, St. Louis, Missouri, United States of America
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University, St. Louis, Missouri, United States of America
| | - Yanbo Yu
- Department of Psychiatry, Washington University, Saint Louis, St. Louis, Missouri, United States of America
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University, St. Louis, Missouri, United States of America
| | - Eric McDade
- Department of Neurology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | | | - Oscar Harari
- Department of Psychiatry, Washington University, Saint Louis, St. Louis, Missouri, United States of America
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University, St. Louis, Missouri, United States of America
- Department of Neurology, Division of Neurogenetics, The Ohio State University, Columbus, OH, United States of America
| | - Bruno A. Benitez
- Department of Neurology and Neuroscience, Harvard Medical School and Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| |
Collapse
|
18
|
Malara M, Prestel M, Tahirovic S. Endo-lysosomal dysfunction and neuronal-glial crosstalk in Niemann-Pick type C disease. Philos Trans R Soc Lond B Biol Sci 2024; 379:20220388. [PMID: 38368932 PMCID: PMC10874755 DOI: 10.1098/rstb.2022.0388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 11/27/2023] [Indexed: 02/20/2024] Open
Abstract
Niemann-Pick type C (NPC) disease is a rare progressive lysosomal lipid storage disorder that manifests with a heterogeneous spectrum of clinical syndromes, including visceral, neurological and psychiatric symptoms. This monogenetic autosomal recessive disease is largely caused by mutations in the NPC1 gene, which controls intracellular lipid homeostasis. Vesicle-mediated endo-lysosomal lipid trafficking and non-vesicular lipid exchange via inter-organelle membrane contact sites are both regulated by the NPC1 protein. Loss of NPC1 function therefore triggers intracellular accumulation of diverse lipid species, including cholesterol, glycosphingolipids, sphingomyelin and sphingosine. The NPC1-mediated dysfunction of lipid transport has severe consequences for all brain cells, leading to neurodegeneration. Besides the cell-autonomous contribution of neuronal NPC1, aberrant NPC1 signalling in other brain cells is critical for the pathology. We discuss here the importance of endo-lysosomal dysfunction and a tight crosstalk between neurons, oligodendrocytes, astrocytes and microglia in NPC pathology. We strongly believe that a cell-specific rescue may not be sufficient to counteract the severity of the NPC pathology, but targeting common mechanisms, such as endo-lysosomal and lipid trafficking dysfunction, may ameliorate NPC pathology. This article is part of a discussion meeting issue 'Understanding the endo-lysosomal network in neurodegeneration'.
Collapse
Affiliation(s)
- Mariagiovanna Malara
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
| | - Matthias Prestel
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
| | - Sabina Tahirovic
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
| |
Collapse
|
19
|
O’Donnell C, Demler TL, Trigoboff E, Lee C. The Impact of High-density Lipoprotein Cholesterol (HDL-C) Levels and Risk of Movement Disorders in Patients Taking Antipsychotics. INNOVATIONS IN CLINICAL NEUROSCIENCE 2024; 21:27-30. [PMID: 38938537 PMCID: PMC11208005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
Abstract
Introduction Well-known adverse events of antipsychotics are movement disorders, or extrapyramidal symptoms, such as drug-induced parkinsonism and tardive dyskinesia. Objective With new evidence suggesting a link between low high-density lipoprotein cholesterol (HDL-C) and risk of Parkinson's disease, this study sought to investigate if that link also translated to patients taking antipsychotics with low HDL-C and an increased risk for developing a movement disorder. Design Adult patients (n=89) at an inpatient state psychiatric facility taking at least one antipsychotic with at least one HDL-C level were assessed for signs of a movement disorder through their history and physical, progress notes, and Abnormal Involuntary Movement Scale (AIMS) score. Results There was no statistical significance when comparing a patient's movement disorder, AIMS scores, and HDL-C levels to suggest that the HDL-C level influenced a patient's movement disorder. Conclusion This study did not show a correlation between HDL-C levels and a patient's risk of developing a movement disorder while taking an antipsychotic.
Collapse
Affiliation(s)
- Carolyn O’Donnell
- Dr. O’Donnell is with Edward Hines Jr. Veterans Administration Hospital in Hines, Illinois, and was with the New York State Office of Mental Health at Buffalo Psychiatric Center in Buffalo, New York, at the time of the study
| | - Tammie Lee Demler
- Dr. Demler is with Department of Pharmacy Practice, State University of New York at Buffalo, School of Pharmacy and Pharmaceutical Sciences in Buffalo, New York; Department of Pharmacy, New York State Office of Mental Health at Buffalo Psychiatric Center in Buffalo, New York; and Department of Psychiatry, State University of New York at Buffalo, Jacobs School of Medicine and Biomedical Sciences in Buffalo, New York
| | - Eileen Trigoboff
- Dr. Trigoboff is with SCH Pharmacy, SCH Medicine Department of Psychiatry at The State University of New York at Buffalo in Buffalo, New York
| | - Claudia Lee
- Dr. Lee is with Buffalo Psychiatric Center in Buffalo, New York
| |
Collapse
|
20
|
Szepanowski LP, Wruck W, Kapr J, Rossi A, Fritsche E, Krutmann J, Adjaye J. Cockayne Syndrome Patient iPSC-Derived Brain Organoids and Neurospheres Show Early Transcriptional Dysregulation of Biological Processes Associated with Brain Development and Metabolism. Cells 2024; 13:591. [PMID: 38607030 PMCID: PMC11011893 DOI: 10.3390/cells13070591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/24/2024] [Accepted: 03/26/2024] [Indexed: 04/13/2024] Open
Abstract
Cockayne syndrome (CS) is a rare hereditary autosomal recessive disorder primarily caused by mutations in Cockayne syndrome protein A (CSA) or B (CSB). While many of the functions of CSB have been at least partially elucidated, little is known about the actual developmental dysregulation in this devasting disorder. Of particular interest is the regulation of cerebral development as the most debilitating symptoms are of neurological nature. We generated neurospheres and cerebral organoids utilizing Cockayne syndrome B protein (CSB)-deficient induced pluripotent stem cells derived from two patients with distinct severity levels of CS and healthy controls. The transcriptome of both developmental timepoints was explored using RNA-Seq and bioinformatic analysis to identify dysregulated biological processes common to both patients with CS in comparison to the control. CSB-deficient neurospheres displayed upregulation of the VEGFA-VEGFR2 signalling pathway, vesicle-mediated transport and head development. CSB-deficient cerebral organoids exhibited downregulation of brain development, neuron projection development and synaptic signalling. We further identified the upregulation of steroid biosynthesis as common to both timepoints, in particular the upregulation of the cholesterol biosynthesis branch. Our results provide insights into the neurodevelopmental dysregulation in patients with CS and strengthen the theory that CS is not only a neurodegenerative but also a neurodevelopmental disorder.
Collapse
Affiliation(s)
- Leon-Phillip Szepanowski
- Institute for Stem Cell Research and Regenerative Medicine, University Hospital Düsseldorf, Moorenstrasse 5, D-40225 Duesseldorf, Germany; (L.-P.S.)
- IUF—Leibniz Research Institute for Environmental Medicine, Auf’m Hennekamp 50, D-40225 Duesseldorf, Germany
| | - Wasco Wruck
- Institute for Stem Cell Research and Regenerative Medicine, University Hospital Düsseldorf, Moorenstrasse 5, D-40225 Duesseldorf, Germany; (L.-P.S.)
| | - Julia Kapr
- IUF—Leibniz Research Institute for Environmental Medicine, Auf’m Hennekamp 50, D-40225 Duesseldorf, Germany
| | - Andrea Rossi
- IUF—Leibniz Research Institute for Environmental Medicine, Auf’m Hennekamp 50, D-40225 Duesseldorf, Germany
| | - Ellen Fritsche
- IUF—Leibniz Research Institute for Environmental Medicine, Auf’m Hennekamp 50, D-40225 Duesseldorf, Germany
| | - Jean Krutmann
- IUF—Leibniz Research Institute for Environmental Medicine, Auf’m Hennekamp 50, D-40225 Duesseldorf, Germany
| | - James Adjaye
- Institute for Stem Cell Research and Regenerative Medicine, University Hospital Düsseldorf, Moorenstrasse 5, D-40225 Duesseldorf, Germany; (L.-P.S.)
- Zayed Centre for Research into Rare Diseases in Children (ZCR), University College London (UCL)—EGA Institute for Women’s Health, 20 Guilford Street, London WC1N 1DZ, UK
| |
Collapse
|
21
|
Peeples ES, Mirnics K, Korade Z. Chemical Inhibition of Sterol Biosynthesis. Biomolecules 2024; 14:410. [PMID: 38672427 PMCID: PMC11048061 DOI: 10.3390/biom14040410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 03/22/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
Cholesterol is an essential molecule of life, and its synthesis can be inhibited by both genetic and nongenetic mechanisms. Hundreds of chemicals that we are exposed to in our daily lives can alter sterol biosynthesis. These also encompass various classes of FDA-approved medications, including (but not limited to) commonly used antipsychotic, antidepressant, antifungal, and cardiovascular medications. These medications can interfere with various enzymes of the post-lanosterol biosynthetic pathway, giving rise to complex biochemical changes throughout the body. The consequences of these short- and long-term homeostatic disruptions are mostly unknown. We performed a comprehensive review of the literature and built a catalogue of chemical agents capable of inhibiting post-lanosterol biosynthesis. This process identified significant gaps in existing knowledge, which fall into two main areas: mechanisms by which sterol biosynthesis is altered and consequences that arise from the inhibitions of the different steps in the sterol biosynthesis pathway. The outcome of our review also reinforced that sterol inhibition is an often-overlooked mechanism that can result in adverse consequences and that there is a need to develop new safety guidelines for the use of (novel and already approved) medications with sterol biosynthesis inhibiting side effects, especially during pregnancy.
Collapse
Affiliation(s)
- Eric S. Peeples
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE 68198, USA;
- Child Health Research Institute, Omaha, NE 68198, USA;
- Division of Neonatology, Children’s Nebraska, Omaha, NE 68114, USA
| | - Karoly Mirnics
- Child Health Research Institute, Omaha, NE 68198, USA;
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Pharmacology & Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Zeljka Korade
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE 68198, USA;
- Child Health Research Institute, Omaha, NE 68198, USA;
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
22
|
Petrov AM. Oxysterols in Central and Peripheral Synaptic Communication. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1440:91-123. [PMID: 38036877 DOI: 10.1007/978-3-031-43883-7_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Cholesterol is a key molecule for synaptic transmission, and both central and peripheral synapses are cholesterol rich. During intense neuronal activity, a substantial portion of synaptic cholesterol can be oxidized by either enzymatic or non-enzymatic pathways to form oxysterols, which in turn modulate the activities of neurotransmitter receptors (e.g., NMDA and adrenergic receptors), signaling molecules (nitric oxide synthases, protein kinase C, liver X receptors), and synaptic vesicle cycling involved in neurotransmitters release. 24-Hydroxycholesterol, produced by neurons in the brain, could directly affect neighboring synapses and change neurotransmission. 27-Hydroxycholesterol, which can cross the blood-brain barrier, can alter both synaptogenesis and synaptic plasticity. Increased generation of 25-hydroxycholesterol by activated microglia and macrophages could link inflammatory processes to learning and neuronal regulation. Amyloids and oxidative stress can lead to an increase in the levels of ring-oxidized sterols and some of these oxysterols (4-cholesten-3-one, 5α-cholestan-3-one, 7β-hydroxycholesterol, 7-ketocholesterol) have a high potency to disturb or modulate neurotransmission at both the presynaptic and postsynaptic levels. Overall, oxysterols could be used as "molecular prototypes" for therapeutic approaches. Analogs of 24-hydroxycholesterol (SGE-301, SGE-550, SAGE718) can be used for correction of NMDA receptor hypofunction-related states, whereas inhibitors of cholesterol 24-hydroxylase, cholestane-3β,5α,6β-triol, and cholest-4-en-3-one oxime (olesoxime) can be utilized as potential anti-epileptic drugs and (or) protectors from excitotoxicity.
Collapse
Affiliation(s)
- Alexey M Petrov
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", Kazan, RT, Russia.
- Kazan State Medial University, Kazan, RT, Russia.
- Kazan Federal University, Kazan, RT, Russia.
| |
Collapse
|
23
|
Vallée M. Advances in steroid research from the pioneering neurosteroid concept to metabolomics: New insights into pregnenolone function. Front Neuroendocrinol 2024; 72:101113. [PMID: 37993022 DOI: 10.1016/j.yfrne.2023.101113] [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: 09/22/2023] [Revised: 11/13/2023] [Accepted: 11/19/2023] [Indexed: 11/24/2023]
Abstract
Advances in neuroendocrinology have led to major discoveries since the 19th century, identifying adaptive loops for maintaining homeostasis. One of the most remarkable discoveries was the concept of neurosteroids, according to which the brain is not only a target but also a source of steroid production. The identification of new membrane steroid targets now underpins the neuromodulatory effects of neurosteroids such as pregnenolone, which is involved in functions mediated by the GPCR CB1 receptor. Structural analysis of steroids is a key feature of their interactions with the phospholipid membrane, receptors and resulting activity. Therefore, mass spectrometry-based methods have been developed to elucidate the metabolic pathways of steroids, the ultimate approach being metabolomics, which allows the identification of a large number of metabolites in a single sample. This approach should enable us to make progress in understanding the role of neurosteroids in the functioning of physiological and pathological processes.
Collapse
Affiliation(s)
- Monique Vallée
- University Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000 Bordeaux, France.
| |
Collapse
|
24
|
Pappolla MA, Refolo L, Sambamurti K, Zambon D, Duff K. Hypercholesterolemia and Alzheimer's Disease: Unraveling the Connection and Assessing the Efficacy of Lipid-Lowering Therapies. J Alzheimers Dis 2024; 101:S371-S393. [PMID: 39422957 DOI: 10.3233/jad-240388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
This article examines the relationship between cholesterol levels and Alzheimer's disease (AD), beginning with the early observation that individuals who died from heart attacks often had brain amyloid deposition. Subsequent animal model research proved that high cholesterol could hasten amyloid accumulation. In contrast, cholesterol-lowering treatments appeared to counteract this effect. Human autopsy studies reinforced the cholesterol-AD connection, revealing that higher cholesterol levels during midlife significantly correlated with higher brain amyloid pathology. This effect was especially pronounced in individuals aged 40 to 55. Epidemiological data supported animal research and human tissue observations and suggested that managing cholesterol levels in midlife could reduce the risk of developing AD. We analyze the main observational studies and clinical trials on the efficacy of statins. While observational data often suggest a potential protective effect against AD, clinical trials have not consistently shown benefit. The failure of these trials to demonstrate a clear advantage is partially attributed to multiple factors, including the timing of statin therapy, the type of statin and the appropriate selection of patients for treatment. Many studies failed to target individuals who might benefit most from early intervention, such as high-risk patients like APOE4 carriers. The review addresses how cholesterol is implicated in AD through various biological pathways, the potential preventive role of cholesterol management as suggested by observational studies, and the difficulties encountered in clinical trials, particularly related to statin use. The paper highlights the need to explore alternate therapeutic targets and mechanisms that escape statin intervention.
Collapse
Affiliation(s)
- Miguel A Pappolla
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Lorenzo Refolo
- Translational Research Branch, Division of Neuroscience, Bethesda, MD, USA
| | - Kumar Sambamurti
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC, USA
| | - Daniel Zambon
- Universitat Internacional de Catalunya, Barcelona, Spain
| | - Karen Duff
- Karen Duff, UK Dementia Research Institute at University College London, London, UK
| |
Collapse
|
25
|
Biojone C, Cannarozzo C, Seiffert N, Diniz CRAF, Brunello CA, Castrén E, Casarotto P. Mutation in the TRKB Cholesterol Recognition Site that blocks Antidepressant Binding does not Influence the Basal or BDNF-Stimulated Activation of TRKB. Cell Mol Neurobiol 2023; 44:4. [PMID: 38104054 PMCID: PMC10725331 DOI: 10.1007/s10571-023-01438-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 11/04/2023] [Indexed: 12/19/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) acting upon its receptor Neurotrophic tyrosine kinase receptor 2 (NTRK2, TRKB) plays a central role in the development and maintenance of synaptic function and activity- or drug-induced plasticity. TRKB possesses an inverted cholesterol recognition and alignment consensus sequence (CARC), suggesting this receptor can act as a cholesterol sensor. We have recently shown that antidepressant drugs directly bind to the CARC domain of TRKB dimers, and that this binding as well as biochemical and behavioral responses to antidepressants are lost with a mutation in the TRKB CARC motif (Tyr433Phe). However, it is not clear if this mutation can also compromise the receptor function and lead to behavioral alterations. Here, we observed that Tyr433Phe mutation does not alter BDNF binding to TRKB, or BDNF-induced dimerization of TRKB. In this line, primary cultures from embryos of heterozygous Tyr433Phe mutant mice (hTRKB.Tyr433Phe) are responsive to BDNF-induced activation of TRKB, and samples from adult mice do not show any difference on TRKB activation compared to wild-type littermates (TRKB.wt). The behavioral phenotype of hTRKB.Tyr433Phe mice is indistinguishable from the wild-type mice in cued fear conditioning, contextual discrimination task, or the elevated plus maze, whereas mice heterozygous to BDNF null allele show a phenotype in context discrimination task. Taken together, our results indicate that Tyr433Phe mutation in the TRKB CARC motif does not show signs of loss-of-function of BDNF responses, while antidepressant binding to TRKB and responses to antidepressants are lost in Tyr433Phe mutants, making them an interesting mouse model for antidepressant research.
Collapse
Affiliation(s)
- Caroline Biojone
- Neuroscience Center - HiLife, University of Helsinki, P. O. Box 63, 00014, Helsinki, Finland
- Department of Biomedicine, Faculty of Health, Aarhus University, Aarhus, Denmark
- Translational Neuropsychiatry Unit (TNU), Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Cecilia Cannarozzo
- Neuroscience Center - HiLife, University of Helsinki, P. O. Box 63, 00014, Helsinki, Finland
| | - Nina Seiffert
- Neuroscience Center - HiLife, University of Helsinki, P. O. Box 63, 00014, Helsinki, Finland
| | - Cassiano R A F Diniz
- Neuroscience Center - HiLife, University of Helsinki, P. O. Box 63, 00014, Helsinki, Finland
- Center for Neuroscience, University of California, Davis, CA, USA
| | - Cecilia A Brunello
- Neuroscience Center - HiLife, University of Helsinki, P. O. Box 63, 00014, Helsinki, Finland
| | - Eero Castrén
- Neuroscience Center - HiLife, University of Helsinki, P. O. Box 63, 00014, Helsinki, Finland
| | - Plinio Casarotto
- Neuroscience Center - HiLife, University of Helsinki, P. O. Box 63, 00014, Helsinki, Finland.
| |
Collapse
|
26
|
Kang S, Lee J, Choi S, Nesbitt J, Min PH, Trushina E, Choi DS. Moderate ethanol exposure reduces astrocyte-induced neuroinflammatorysignaling and cognitive decline in presymptomatic APP/PS1 mice. RESEARCH SQUARE 2023:rs.3.rs-3627637. [PMID: 38077051 PMCID: PMC10705690 DOI: 10.21203/rs.3.rs-3627637/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Background Alcohol use disorder (AUD) has been associated with the development of neurodegenerative diseases, including Alzheimer's disease (AD). However, recent studies demonstrate that moderate alcohol consumption may be protective against dementia and cognitive decline. Methods We examined astrocyte function, low-density lipoprotein (LDL) receptor-related protein 1 (LRP1), and the NF-κB p65 and IKK-α/β signaling pathways in modulating neuroinflammation and amyloid beta (Aβ) deposition. We assessed apolipoprotein E (ApoE) in the mouse brain using IHC and ELISA in response to moderate ethanol exposure (MEE). First, to confirm the intracerebral distribution of ApoE, we co-stained with GFAP, a marker for astrocytes that biosynthesize ApoE. We sought to investigate whether the ethanol-induced upregulation of LRP1 could potentially inhibit the activity of IL-1β and TNF-α induced IKK-α/β towards NF-κB p65, resulting in a reduction of pro-inflammatory cytokines. To evaluate the actual Aβ load in the brains of APP/PS1 mice, we performed with a specific antibody Aβ (Thioflavin S) on both air- and ethanol-exposed groups, subsequently analyzing Aβ levels. We also measured glucose uptake activity using 18F-FDG in APP/PS1 mice. Finally, we investigated whether MEE induced cognitive and memory changes using the Y maze, noble objective recognition (NOR) test, and Morris water maze (MWM). Results Our findings demonstrate that MEE reduced astrocytic glial fibrillary acidic protein (GFAP) and ApoE levels in the cortex and hippocampus in presymptomatic APP/PS1 mice. Interestingly, increased LRP1 protein expression is accompanied by dampening the IKK-α/β-NF-κB p65 pathway, resulting in decreased IL-1β and TNF-α levels in male mice. Notably, female mice show reduced anti-inflammatory cytokines, IL-4, and IL-10 levels without altering IL-1β and TNF-α concentrations. In both males and females, Aβ plaques, a hallmark of AD, were reduced in the cortex and hippocampus of ethanol-exposed presymptomatic APP/PS1 mice. Consistently, MEE increased fluorodeoxyglucose (FDG)-positron emission tomography (PET)-based brain activities and normalized cognitive and memory deficits in the APP/PS1 mice. Conclusions Our findings suggest that MEE may benefit AD pathology via modulating LRP1 expression, potentially reducing neuroinflammation and attenuating Aβ deposition. Our study implies that reduced astrocyte derived ApoE and LDL cholesterol levels are critical for attenuating AD pathology.
Collapse
Affiliation(s)
| | - Jeyeon Lee
- Mayo Clinic College of Medicine, and Science
| | - Sun Choi
- Mayo Clinic College of Medicine, and Science
| | | | - Paul H Min
- Mayo Clinic College of Medicine, and Science
| | | | | |
Collapse
|
27
|
Hu ZL, Yuan YQ, Tong Z, Liao MQ, Yuan SL, Jian Y, Yang JL, Liu WF. Reexamining the Causes and Effects of Cholesterol Deposition in the Brains of Patients with Alzheimer's Disease. Mol Neurobiol 2023; 60:6852-6868. [PMID: 37507575 DOI: 10.1007/s12035-023-03529-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 07/20/2023] [Indexed: 07/30/2023]
Abstract
Alzheimer's disease (AD) is a degenerative disease of the central nervous system. Numerous studies have shown that imbalances in cholesterol homeostasis in the brains of AD patients precede the onset of clinical symptoms. In addition, cholesterol deposition has been observed in the brains of AD patients even though peripheral cholesterol does not enter the brain through the blood‒brain barrier (BBB). Studies have demonstrated that cholesterol metabolism in the brain is associated with many pathological conditions, such as amyloid beta (Aβ) production, Tau protein phosphorylation, oxidative stress, and inflammation. In 2022, some scholars put forward a new hypothesis of AD: the disease involves lipid invasion and its exacerbation of the abnormal metabolism of cholesterol in the brain. In this review, by discussing the latest research progress, the causes and effects of cholesterol retention in the brains of AD patients are analyzed and discussed. Additionally, the possible mechanism through which AD may be improved by targeting cholesterol is described. Finally, we propose that improving the impairments in cholesterol removal observed in the brains of AD patients, instead of further reducing the already impaired cholesterol synthesis in the brain, may be the key to preventing cholesterol deposition and improving the corresponding pathological symptoms.
Collapse
Affiliation(s)
- Ze-Lin Hu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Yang-Qi Yuan
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Zhen Tong
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Mei-Qing Liao
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Shun-Ling Yuan
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Ye Jian
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Jia-Lun Yang
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Wen-Feng Liu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China.
- Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, Hunan Normal University, Changsha, 410081, China.
| |
Collapse
|
28
|
Constantinescu CC, Brown T, Wang S, Yin W, Barret O, Jennings D, Tauscher J. Clinical Characterization of [ 18F]T-008, a Cholesterol 24-Hydroxylase PET Ligand: Dosimetry, Kinetic Modeling, Variability, and Soticlestat Occupancy. J Nucl Med 2023; 64:1972-1979. [PMID: 37770111 PMCID: PMC10690114 DOI: 10.2967/jnumed.123.265912] [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: 04/20/2023] [Revised: 08/18/2023] [Indexed: 10/03/2023] Open
Abstract
This series of studies characterized [18F]T-008, a PET radiotracer for imaging cholesterol 24-hydroxylase (CH24H), in healthy volunteers (ClinicalTrials.gov identifier NCT02497235). Assessments included radiation dosimetry, kinetic modeling, test-retest variability (TRT) evaluation, and a dose occupancy evaluation using soticlestat, a selective CH24H inhibitor. Soticlestat is currently in phase 3 development for the treatment of seizures in Dravet syndrome and Lennox-Gastaut syndrome. Methods: In the dosimetry study, 5 participants (3 men) underwent serial whole-body scans to estimate organ-absorbed doses and effective doses of [18F]T-008 using OLINDA/EXM 1.1. For the kinetic modeling and TRT study, 6 participants (all men) underwent two 210-min dynamic [18F]T-008 PET scans with arterial blood sampling. The regional total volume of distribution was estimated using a 1-tissue-compartment model, a 2-tissue-compartment model, and Logan graphic analysis. In the dose occupancy study, 11 participants (all men) underwent 120-min scans at baseline and 2 time points (peak and trough) after receiving single oral doses of soticlestat (50-600 mg). The relationship between effect-site soticlestat concentration and brain occupancy was evaluated with a specially developed pharmacokinetic model and a saturable maximal occupancy model. Results: The estimated mean whole-body effective dose was 0.0292 mSv/MBq (SD, 0.00147 mSv/MBq). [18F]T-008 entered the brain rapidly, with a distribution consistent with known CH24H distribution densities. The 2-tissue-compartment model and Logan graphic analysis best described the tracer kinetics. The mean TRT for estimating total volume of distribution was 7%-15%. Single doses of soticlestat in the range 50-600 mg resulted in occupancies of 64%-96% at 2 h and 11%-79% at 24 h. The estimated half-maximal effect-site concentration of soticlestat was 5.52 ng/mL. Conclusion: [18F]T-008 is a suitable PET radiotracer for quantitatively analyzing CH24H in the human brain. Using [18F]T-008 and PET, we demonstrated that soticlestat was brain-penetrant and established target engagement by displacing [18F]T-008 in a dose-dependent manner in the brain.
Collapse
Affiliation(s)
| | - Terry Brown
- Takeda Pharmaceutical Co. Ltd., Cambridge, Massachusetts
| | - Shining Wang
- Takeda Pharmaceutical Co. Ltd., Cambridge, Massachusetts
| | - Wei Yin
- Takeda Pharmaceutical Co. Ltd., Cambridge, Massachusetts
| | | | | | | |
Collapse
|
29
|
Sviridov D, Bukrinsky M. Neuro-HIV-New insights into pathogenesis and emerging therapeutic targets. FASEB J 2023; 37:e23301. [PMID: 37942865 PMCID: PMC11032165 DOI: 10.1096/fj.202301239rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 10/22/2023] [Accepted: 10/25/2023] [Indexed: 11/10/2023]
Abstract
HIV-associated neurocognitive disorders (HAND) is a term describing a complex set of cognitive impairments accompanying HIV infection. Successful antiretroviral therapy (ART) reduces the most severe forms of HAND, but milder forms affect over 50% of people living with HIV (PLWH). Pathogenesis of HAND in the ART era remains unknown. A variety of pathogenic factors, such as persistent HIV replication in the brain reservoir, HIV proteins released from infected brain cells, HIV-induced neuroinflammation, and some components of ART, have been implicated in driving HAND pathogenesis in ART-treated individuals. Here, we propose another factor-impairment of cholesterol homeostasis and lipid rafts by HIV-1 protein Nef-as a possible contributor to HAND pathogenesis. These effects of Nef on cholesterol may also underlie the effects of other pathogenic factors that constitute the multifactorial nature of HAND pathogenesis. The proposed Nef- and cholesterol-focused mechanism may provide a long-sought unified explanation of HAND pathogenesis that takes into account all contributing factors. Evidence for the impairment by Nef of cellular cholesterol balance, potential effects of this impairment on brain cells, and opportunities to therapeutically target this element of HAND pathogenesis are discussed.
Collapse
Affiliation(s)
- Dmitri Sviridov
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Michael Bukrinsky
- The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| |
Collapse
|
30
|
Shaheen A, Richter Gorey CL, Sghaier A, Dason JS. Cholesterol is required for activity-dependent synaptic growth. J Cell Sci 2023; 136:jcs261563. [PMID: 37902091 DOI: 10.1242/jcs.261563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 10/24/2023] [Indexed: 10/31/2023] Open
Abstract
Changes in cholesterol content of neuronal membranes occur during development and brain aging. Little is known about whether synaptic activity regulates cholesterol levels in neuronal membranes and whether these changes affect neuronal development and function. We generated transgenic flies that express the cholesterol-binding D4H domain of perfringolysin O toxin and found increased levels of cholesterol in presynaptic terminals of Drosophila larval neuromuscular junctions following increased synaptic activity. Reduced cholesterol impaired synaptic growth and largely prevented activity-dependent synaptic growth. Presynaptic knockdown of adenylyl cyclase phenocopied the impaired synaptic growth caused by reducing cholesterol. Furthermore, the effects of knocking down adenylyl cyclase and reducing cholesterol were not additive, suggesting that they function in the same pathway. Increasing cAMP levels using a dunce mutant with reduced phosphodiesterase activity failed to rescue this impaired synaptic growth, suggesting that cholesterol functions downstream of cAMP. We used a protein kinase A (PKA) sensor to show that reducing cholesterol levels reduced presynaptic PKA activity. Collectively, our results demonstrate that enhanced synaptic activity increased cholesterol levels in presynaptic terminals and that these changes likely activate the cAMP-PKA pathway during activity-dependent growth.
Collapse
Affiliation(s)
- Amber Shaheen
- Department of Biomedical Sciences, University of Windsor, Windsor, Ontario, N9B 3P4, Canada
| | - Claire L Richter Gorey
- Department of Biomedical Sciences, University of Windsor, Windsor, Ontario, N9B 3P4, Canada
| | - Adam Sghaier
- Department of Biomedical Sciences, University of Windsor, Windsor, Ontario, N9B 3P4, Canada
| | - Jeffrey S Dason
- Department of Biomedical Sciences, University of Windsor, Windsor, Ontario, N9B 3P4, Canada
| |
Collapse
|
31
|
Islam M, Behura SK. Role of caveolin-1 in metabolic programming of fetal brain. iScience 2023; 26:107710. [PMID: 37720105 PMCID: PMC10500482 DOI: 10.1016/j.isci.2023.107710] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/10/2023] [Accepted: 08/23/2023] [Indexed: 09/19/2023] Open
Abstract
Mice lacking caveolin-1 (Cav1), a key protein of plasma membrane, exhibit brain aging at an early adult stage. Here, integrative analyses of metabolomics, transcriptomics, epigenetics, and single-cell data were performed to test the hypothesis that metabolic deregulation of fetal brain due to the ablation of Cav1 is linked to brain aging in these mice. The results of this study show that lack of Cav1 caused deregulation in the lipid and amino acid metabolism in the fetal brain, and genes associated with these deregulated metabolites were significantly altered in the brain upon aging. Moreover, ablation of Cav1 deregulated several metabolic genes in specific cell types of the fetal brain and impacted DNA methylation of those genes in coordination with mouse epigenetic clock. The findings of this study suggest that the aging program of brain is confounded by metabolic abnormalities in the fetal stage due to the absence of Cav1.
Collapse
Affiliation(s)
- Maliha Islam
- Division of Animal Sciences, 920 East Campus Drive, University of Missouri, Columbia, MO 65211, USA
| | - Susanta K. Behura
- Division of Animal Sciences, 920 East Campus Drive, University of Missouri, Columbia, MO 65211, USA
- MU Institute for Data Science and Informatics, University of Missouri, Columbia, MO, USA
- Interdisciplinary Reproduction and Health Group, University of Missouri, Columbia, MO, USA
- Interdisciplinary Neuroscience Program, University of Missouri, Columbia, MO, USA
| |
Collapse
|
32
|
Parente M, Tonini C, Segatto M, Pallottini V. Regulation of cholesterol metabolism: New players for an old physiological process. J Cell Biochem 2023; 124:1449-1465. [PMID: 37796135 DOI: 10.1002/jcb.30477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/30/2023] [Accepted: 09/12/2023] [Indexed: 10/06/2023]
Abstract
Identified more than two centuries ago, cholesterol plays a pivotal role in human physiology. Since cholesterol metabolism is a physiologically significant process, it is not surprising that its alterations are associated with several pathologies. The discovery of new molecular targets or compounds able to modulate this sophisticated metabolism has been capturing the attention of research groups worldwide since many years. Endogenous and exogenous compounds are known to regulate cellular cholesterol synthesis and uptake, or reduce cholesterol absorption at the intestinal level, thereby regulating cholesterol homeostasis. However, there is a great need of new modulators and diverse new pathways have been uncovered. Here, after illustrating cholesterol metabolism and its well-known regulators, some new players of this important physiological process are also described.
Collapse
Affiliation(s)
| | | | - Marco Segatto
- Department of Bioscience and Territory, University of Molise, Pesche, Italy
| | - Valentina Pallottini
- Department of Science, University Roma Tre, Rome, Italy
- Neuroendocrinology Metabolism and Neuropharmacology Unit, IRCSS Fondazione Santa Lucia, Via del Fosso Fiorano, Rome, Italy
| |
Collapse
|
33
|
Matza Porges S, Mor-Shaked H, Shaag A, Porat S, Daum H. Prenatal diagnosis of lanosterol synthase deficiency: Fetal ultrasound findings as a window on family genetics. Eur J Med Genet 2023; 66:104825. [PMID: 37659595 DOI: 10.1016/j.ejmg.2023.104825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/18/2023] [Accepted: 08/29/2023] [Indexed: 09/04/2023]
Abstract
Cholesterol is essential in the brain from the earliest stages of embryonic development. Disruption of cholesterol synthesis pathways that leads to cholesterol deficiency underlies a few syndromes, including desmosterolosis and Smith-Lemli-Opitz syndrome. In both syndromes, brain anomalies can occur. The LSS gene encodes lanosterol synthase (LSS), an important enzyme in the cholesterol biosynthesis pathway. Biallelic pathogenic variants in this gene cause alopecia-intellectual disability type 4 syndrome (APMR4, MIM 618840), a rare autosomal recessive disorder. Here, we describe two new LSS variants (c.1016C > T; p. Ser339Leu and c.1522G > C; p. Gly508Arg) found in a compound heterozygous fetus diagnosed prenatally with brain abnormalities by ultrasound scanning. Two of his siblings from the same parents also harbored these variants. Both siblings had alopecia, mild intellectual disability, autism spectrum disorder, and cataracts. To the best of our knowledge, this case represents the first prenatal diagnosis of APMR4 first suspected by ultrasound. In addition, the phenotypic features of the siblings are extensive compared with those described in previous reports and include abnormal corpus callosum, cataracts, alopecia, and developmental delay.
Collapse
Affiliation(s)
- Sigal Matza Porges
- Department of Genetics, Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel; Department of Biotechnology, Hadassah Academic College, Jerusalem, Israel.
| | - Hagar Mor-Shaked
- Department of Genetics, Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel; Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Avraham Shaag
- Department of Genetics, Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Shay Porat
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel; Department of Obstetrics and Gynecology, Hadassah Medical Organization, Jerusalem, Israel
| | - Hagit Daum
- Department of Genetics, Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel; Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
34
|
Chen Y, Yang K, Huang Y, Wang X, Zhao Y, Ping P, Guan S, Fu S. Associations between lipid profiles and late-life cognitive impairment among oldest-old and centenarian adults. MedComm (Beijing) 2023; 4:e362. [PMID: 37692108 PMCID: PMC10484073 DOI: 10.1002/mco2.362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 07/23/2023] [Accepted: 08/06/2023] [Indexed: 09/12/2023] Open
Abstract
Dyslipidemia and cognitive impairment are common among old adults and the occurrence of them rises exponentially with increasing age. Evidences of the relationships between serum lipids and cognitive impairment are inconsistent or equivocal among older adults. This study aimed to investigate the associations between lipid profiles and late-life cognitive impairment among oldest-old and centenarian adults. In this cross-sectional study, serum lipids were biochemically measured among 606 oldest-old adults and 653 centenarians, and cognitive function was evaluated using mini-mental state examination (MMSE). Multivariate linear and logistic regression analyses were performed to explore the associations between serum lipids and cognitive impairment. Results showed participants with cognitive impairment had lower total cholesterol (TC) levels compared with those without cognitive impairment (p < 0.05). TC levels were positively associated with MMSE (p < 0.05). Furthermore, a negative association was observed between TC levels and cognitive impairment (p for trend = 0.002). This negative association remained statistically significant after adjusting for confounders (p for trend = 0.028). These results suggested that older adults with higher TC levels were likely to have better cognitive function. Taking immoderate cholesterol-lowering drugs among older adults is questionable and requires investigation, and cognitive performance of old adults with lower TC levels deserves more attention.
Collapse
Affiliation(s)
- Yujian Chen
- Central LaboratoryHainan Hospital of Chinese People's Liberation Army General HospitalSanyaChina
| | - Kaidi Yang
- Oncology DepartmentHainan Hospital of Chinese People's Liberation Army General HospitalSanyaChina
| | - Ya Huang
- Blood Transfusion DepartmentHainan Hospital of Chinese People's Liberation Army General HospitalSanyaChina
| | - Xuejiao Wang
- Pediatric DepartmentHainan Hospital of Chinese People's Liberation Army General HospitalSanyaChina
| | - Yali Zhao
- Central LaboratoryHainan Hospital of Chinese People's Liberation Army General HospitalSanyaChina
| | - Ping Ping
- General Station for Drug and Instrument Supervision and ControlJoint Logistic Support Force of Chinese People's Liberation ArmyBeijingChina
| | - Shasha Guan
- Oncology DepartmentHainan Hospital of Chinese People's Liberation Army General HospitalSanyaChina
| | - Shihui Fu
- Department of CardiologyHainan Hospital of Chinese People's Liberation Army General HospitalSanyaChina
- Department of Geriatric CardiologyChinese People's Liberation Army General HospitalBeijingChina
| |
Collapse
|
35
|
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: 6.0] [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
|
36
|
Benachenhou S, Laroui A, Dionne O, Rojas D, Toupin A, Çaku A. Cholesterol alterations in fragile X syndrome, autism spectrum disorders and other neurodevelopmental disorders. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 173:115-139. [PMID: 37993175 DOI: 10.1016/bs.irn.2023.08.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Abstract
Neurodevelopmental disorders (NDDs) are a group of etiologically diverse diseases primarily associated with abnormal brain development, impaired cognition, and various behavioral problems. The majority of NDDs present a wide range of clinical phenotypes while sharing distinct cellular and biochemical alterations. Low plasma cholesterol levels have been reported in a subset of NNDs including, autism spectrum disorder (ASD) and fragile X syndrome (FXS). The present review focuses on cholesterol metabolism and discusses the current evidence of lipid disruption in ASD, FXS, and other genetically related NDDs. The characterization of these common deficits might provide valuable insights into their underlying physiopathology and help identify potential therapeutic targets.
Collapse
Affiliation(s)
- Sérine Benachenhou
- Biochemistry and Functional Genomic Department, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Asma Laroui
- Biochemistry and Functional Genomic Department, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Olivier Dionne
- Biochemistry and Functional Genomic Department, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Daniela Rojas
- Biochemistry and Functional Genomic Department, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Amanda Toupin
- Biochemistry and Functional Genomic Department, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Artuela Çaku
- Biochemistry and Functional Genomic Department, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada.
| |
Collapse
|
37
|
Liu B, Meng Q, Gao X, Sun H, Xu Z, Wang Y, Zhou H. Lipid and glucose metabolism in senescence. Front Nutr 2023; 10:1157352. [PMID: 37680899 PMCID: PMC10481967 DOI: 10.3389/fnut.2023.1157352] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 08/09/2023] [Indexed: 09/09/2023] Open
Abstract
Senescence is an inevitable biological process. Disturbances in glucose and lipid metabolism are essential features of cellular senescence. Given the important roles of these types of metabolism, we review the evidence for how key metabolic enzymes influence senescence and how senescence-related secretory phenotypes, autophagy, apoptosis, insulin signaling pathways, and environmental factors modulate glucose and lipid homeostasis. We also discuss the metabolic alterations in abnormal senescence diseases and anti-cancer therapies that target senescence through metabolic interventions. Our work offers insights for developing pharmacological strategies to combat senescence and cancer.
Collapse
Affiliation(s)
- Bin Liu
- Department of Urology II, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Qingfei Meng
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin, China
| | - Xin Gao
- Department of Urology II, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Huihui Sun
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin, China
| | - Zhixiang Xu
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin, China
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin, China
| | - Honglan Zhou
- Department of Urology II, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
38
|
Sodero AO, Castagna VC, Elorza SD, Gonzalez-Rodulfo SM, Paulazo MA, Ballestero JA, Martin MG, Gomez-Casati ME. Phytosterols reverse antiretroviral-induced hearing loss, with potential implications for cochlear aging. PLoS Biol 2023; 21:e3002257. [PMID: 37619212 PMCID: PMC10449472 DOI: 10.1371/journal.pbio.3002257] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 07/18/2023] [Indexed: 08/26/2023] Open
Abstract
Cholesterol contributes to neuronal membrane integrity, supports membrane protein clustering and function, and facilitates proper signal transduction. Extensive evidence has shown that cholesterol imbalances in the central nervous system occur in aging and in the development of neurodegenerative diseases. In this work, we characterize cholesterol homeostasis in the inner ear of young and aged mice as a new unexplored possibility for the prevention and treatment of hearing loss. Our results show that cholesterol levels in the inner ear are reduced during aging, an effect that is associated with an increased expression of the cholesterol 24-hydroxylase (CYP46A1), the main enzyme responsible for cholesterol turnover in the brain. In addition, we show that pharmacological activation of CYP46A1 with the antiretroviral drug efavirenz reduces the cholesterol content in outer hair cells (OHCs), leading to a decrease in prestin immunolabeling and resulting in an increase in the distortion product otoacoustic emissions (DPOAEs) thresholds. Moreover, dietary supplementation with phytosterols, plant sterols with structure and function similar to cholesterol, was able to rescue the effect of efavirenz administration on the auditory function. Altogether, our findings point towards the importance of cholesterol homeostasis in the inner ear as an innovative therapeutic strategy in preventing and/or delaying hearing loss.
Collapse
Affiliation(s)
- Alejandro O. Sodero
- Instituto de Investigaciones Biomédicas, Pontificia Universidad Católica Argentina, Consejo Nacional de Investigaciones Científicas y Técnicas (BIOMED, UCA-CONICET), Buenos Aires, Argentina
| | - Valeria C. Castagna
- Instituto de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Dr. Héctor N. Torres, Consejo Nacional de Investigaciones Científicas y Técnicas (INGEBI-CONICET), Buenos Aires, Argentina
| | - Setiembre D. Elorza
- Laboratorio de Neurobiología, Instituto de Investigaciones Médicas Mercedes y Martín Ferreyra, Consejo Nacional de Investigaciones Científicas y Técnicas (INIMEC-CONICET-UNC), Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Sara M. Gonzalez-Rodulfo
- Instituto de Investigaciones Biomédicas, Pontificia Universidad Católica Argentina, Consejo Nacional de Investigaciones Científicas y Técnicas (BIOMED, UCA-CONICET), Buenos Aires, Argentina
| | - María A. Paulazo
- Instituto de Investigaciones Biomédicas, Pontificia Universidad Católica Argentina, Consejo Nacional de Investigaciones Científicas y Técnicas (BIOMED, UCA-CONICET), Buenos Aires, Argentina
| | - Jimena A. Ballestero
- Instituto de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Mauricio G. Martin
- Laboratorio de Neurobiología, Instituto de Investigaciones Médicas Mercedes y Martín Ferreyra, Consejo Nacional de Investigaciones Científicas y Técnicas (INIMEC-CONICET-UNC), Universidad Nacional de Córdoba, Córdoba, Argentina
| | - María Eugenia Gomez-Casati
- Instituto de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
39
|
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
|
40
|
You M, Sun L, Li C, Zhu S. ATGL-mediated lipophagy balances cholesterol-induced inflammation in pathogen infected Apostichopus japonicus coelomocytes. FISH & SHELLFISH IMMUNOLOGY 2023; 139:108863. [PMID: 37277050 DOI: 10.1016/j.fsi.2023.108863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 05/18/2023] [Accepted: 05/29/2023] [Indexed: 06/07/2023]
Abstract
Cholesterol metabolism can be dynamically altered in response to pathogen infection that ensure proper macrophage inflammatory function in mammals. However, it is unclear whether the dynamic between cholesterol accumulation and breakdown could induce or suppress inflammation in aquatic animal. Here, we aimed to investigate the cholesterol metabolic response to LPS stimulation in coelomocytes of Apostichopus japonicus, and to elucidate the mechanism of lipophagy in regulating cholesterol-related inflammation. LPS stimulation significantly increased intracellular cholesterol levels at early time point (12 h), and the increase in cholesterol levels is associated with AjIL-17 upregulation. Excessive cholesterol in coelomocytes of A. japonicus was rapidly converted to cholesteryl esters (CEs) and stored in lipid droplets (LDs) after 12 h of LPS stimulation and prolonged for 18 h. Then, increased colocalization of LDs with lysosomes was observed at late time point of LPS treatment (24 h), accompanied by elevated expression of AjLC3 and decreased expression of Ajp62. At the same time, the expression of AjABCA1 rapidly increased, suggesting lipophagy induction. Moreover, we demonstrated that AjATGL is required for induction of lipophagy. Inducing lipophagy by AjATGL overexpression attenuated cholesterol-induced AjIL-17 expression. Overall, our study provides evidence that cholesterol metabolic response occurs upon LPS stimulation, which is actively involved in regulating the inflammatory response of coelomocytes. AjATGL-mediated lipophagy is responsible for cholesterol hydrolysis, thereby balancing cholesterol-induced inflammation in the coelomocytes of A. japonicus.
Collapse
Affiliation(s)
- Meixiang You
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China
| | - Lianlian Sun
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China
| | - Chenghua Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, PR China.
| | - Si Zhu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China.
| |
Collapse
|
41
|
Zhang WB, Huang Y, Guo XR, Zhang MQ, Yuan XS, Zu HB. DHCR24 reverses Alzheimer's disease-related pathology and cognitive impairment via increasing hippocampal cholesterol levels in 5xFAD mice. Acta Neuropathol Commun 2023; 11:102. [PMID: 37344916 DOI: 10.1186/s40478-023-01593-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 05/30/2023] [Indexed: 06/23/2023] Open
Abstract
Accumulating evidences reveal that cellular cholesterol deficiency could trigger the onset of Alzheimer's disease (AD). As a key regulator, 24-dehydrocholesterol reductase (DHCR24) controls cellular cholesterol homeostasis, which was found to be downregulated in AD vulnerable regions and involved in AD-related pathological activities. However, DHCR24 as a potential therapeutic target for AD remains to be identified. In present study, we demonstrated the role of DHCR24 in AD by employing delivery of adeno-associated virus carrying DHCR24 gene into the hippocampus of 5xFAD mice. Here, we found that 5xFAD mice had lower levels of cholesterol and DHCR24 expression, and the cholesterol loss was alleviated by DHCR24 overexpression. Surprisingly, the cognitive impairment of 5xFAD mice was significantly reversed after DHCR24-based gene therapy. Moreover, we revealed that DHCR24 knock-in successfully prevented or reversed AD-related pathology in 5xFAD mice, including amyloid-β deposition, synaptic injuries, autophagy, reactive astrocytosis, microglial phagocytosis and apoptosis. In conclusion, our results firstly demonstrated that the potential value of DHCR24-mediated regulation of cellular cholesterol level as a promising treatment for AD.
Collapse
Affiliation(s)
- Wen-Bin Zhang
- Department of Neurology, Jinshan Hospital Affiliated to Fudan University, No.1508 Long-Hang Road, Jinshan District, Shanghai, 201508, China
| | - Yue Huang
- Department of Neurology, Jinshan Hospital Affiliated to Fudan University, No.1508 Long-Hang Road, Jinshan District, Shanghai, 201508, China
| | - Xiao-Rou Guo
- Department of Neurology, Jinshan Hospital Affiliated to Fudan University, No.1508 Long-Hang Road, Jinshan District, Shanghai, 201508, China
| | - Meng-Qi Zhang
- Department of Neurology, Jinshan Hospital Affiliated to Fudan University, No.1508 Long-Hang Road, Jinshan District, Shanghai, 201508, China
| | - Xiang-Shan Yuan
- Department of Neurology, Jinshan Hospital Affiliated to Fudan University, No.1508 Long-Hang Road, Jinshan District, Shanghai, 201508, China.
- Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
- State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
| | - Heng-Bing Zu
- Department of Neurology, Jinshan Hospital Affiliated to Fudan University, No.1508 Long-Hang Road, Jinshan District, Shanghai, 201508, China.
| |
Collapse
|
42
|
Kim M, Bezprozvanny I. Potential direct role of synuclein in dopamine transport and its implications for Parkinson's disease pathogenesis. Biochem Biophys Res Commun 2023; 671:18-25. [PMID: 37290280 DOI: 10.1016/j.bbrc.2023.05.110] [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: 05/23/2023] [Accepted: 05/25/2023] [Indexed: 06/10/2023]
Abstract
Parkinson Disease (PD) is a progressive neurodegenerative disorder that is caused by dysfunction and death of dopaminergic neurons. Mutations in the gene encoding α-synuclein (ASYN) have been linked with familial PD (FPD). Despite important role of ASYN in PD pathology, its normal biological function has not been clarified, although direct action of ASYN in synaptic transmission and dopamine (DA+) release have been proposed. In the present report we propose a novel hypothesis that ASYN functions as DA+/H+ exchanger that can facilitate transport of dopamine across synaptic vesicle (SV) membrane by taking advantage of proton gradient between SV lumen and cytoplasm. According to this hypothesis, normal physiological role of ASYN consists of fine-tuning levels of dopamine in the SVs based on cytosolic concentration of dopamine and intraluminal pH. This hypothesis is based on similarity in domain structure of ASYN and pHILP, a designed peptide developed to mediate loading of lipid nanoparticles with the cargo molecules. We reason that carboxy-terminal acidic loop D2b domain in both ASYN and pHILP binds cargo molecules. By mimicking DA+ association with E/D residues in D2b domain of ASYN using Tyrosine replacement approach (TR) we have been able to estimate that ASYN is able to transfer 8-12 molecules of dopamine across SV membrane on each DA+/H+ exchange cycle. Our results suggest that familial PD mutations (A30P, E46K, H50Q, G51D, A53T and A53E) will interfere with different steps of the exchange cycle, resulting in partial loss of dopamine transport function phenotype. We also predict that similar impairment in ASYN DA+/H+ exchange function also occurs as a result on neuronal aging due to changes in SV lipid composition and size and also dissipation of pH gradient across SV membrane. Proposed novel functional role of ASYN provides novel insights into its biological role and its role in PD pathogenesis.
Collapse
Affiliation(s)
- Meewhi Kim
- Dept of Physiology, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
| | - Ilya Bezprozvanny
- Dept of Physiology, UT Southwestern Medical Center, Dallas, TX, 75390, USA; Laboratory of Molecular Neurodegeneration, St Petersburg State Polytechnical Universty, St Petersburg, 195251, Russian Federation.
| |
Collapse
|
43
|
Fan SM, Chiu PY, Liu CH, Liao YC, Chang HT. Predictive value of hypercholesterolemia, vegetarian diet, and hypertension for incident dementia among elderly Taiwanese individuals with low educational levels. Ther Adv Chronic Dis 2023; 14:20406223231171549. [PMID: 37255548 PMCID: PMC10226334 DOI: 10.1177/20406223231171549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 04/06/2023] [Indexed: 06/01/2023] Open
Abstract
Objective Early management of modifiable dementia-related factors is seen as a novel approach to preventing dementia onset; however, these efforts are often hindered by the complexity of interactions among these factors. In addition, different modifiable dementia-related factors may contribute to different etiologies of dementia. Design The current study investigated the effects of common modifiable dementia-related factors on prediction of incident dementia, dementia of the Alzheimer's type (DAT), and vascular dementia (VaD). Methods Vascular- and lifestyle-related factors were used as predictors of incident dementia, DAT, and VaD among 1,285 elderly individuals without obvious signs of dementia or mild cognitive impairment. Cox proportional hazard models were used to evaluate the risks associated with each modifiable factor. Results After controlling for factors other than stroke-related factors, hypercholesterolemia was correlated with a relatively low risk of all-cause incident dementia and DAT, whereas a vegetarian diet was correlated with an elevated risk of all-cause incident dementia and VaD. Hypertension was correlated with incident VaD. After controlling for stroke-related factors, a vegetarian diet was correlated with an elevated risk of all-cause dementia. A history of myocardiac infarction and the use of anti-platelet medication were, respectively, associated with a reduced risk of DAT and elevated risk of VaD. The use of anti-hypertensives was associated with a reduced risk of all-cause dementia, whereas the use of anti-lipid agents was associated with slow progression DAT (i.e. exceeding the average conversion time). Hypercholesterolemia was associated with an elevated risk for slow progression DAT. Conclusion These findings could perhaps be used as clinical markers in predicting and preventing incident dementia, DAT, and VaD.
Collapse
Affiliation(s)
| | | | - Chung-Hsiang Liu
- Department of Neurology, China Medical
University Hospital, College of Medicine, China Medical University,
Taichung
| | - Yu-Chi Liao
- Department of Psychology, College of Medical
and Health Sciences, Asia University, Taichung
| | | |
Collapse
|
44
|
Foest D, Knodel A, Ahrends R, Coman C, Franzke J, Brandt S. Flexible Microtube Plasma for the Consecutive-Ionization of Cholesterol in Nano-Electrospray Mass Spectrometry. Anal Chem 2023. [PMID: 37220280 DOI: 10.1021/acs.analchem.2c04052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Electrospray ionization mass spectrometry (ESI-MS) is an established method for the identification of biomarkers. By nano-ESI (nESI), the polar molecular fraction of complex biological samples can be successfully ionized. In contrast, the less-polar free cholesterol, which serves as an important biomarker for several human diseases, is barely accessible by nESI. Although, complex scan functions of modern high-resolution MS devices are able to increase the signal-to-noise ratio, they are limited by the ionization efficiency of the nESI. One possible method to increase the ionization efficiency is the derivatization with acetyl chloride, however interferences with cholesteryl esters must be considered, so chromatographic separation or complex scan functions may be required. A novel approach to increase the yield of cholesterol ions of the nESI could be the application of a second consecutive-ionization process. This publication presents the flexible microtube plasma (FμTP) as a consecutive-ionization source, which allows the determination of cholesterol in nESI-MS analysis. Focusing on the analytical performance, the nESI-FμTP approach increases the cholesterol signal yield in a complex liver extract by a factor of 49. The repeatability and long-term stability could be successfully evaluated. A linear dynamic range of 1.7 orders of magnitude, a minimum detectability of 5.46 mg/L, and a high accuracy (deviation, -8.1%) demonstrates the nESI-FμTP-MS as an excellent approach for a derivatization-free determination of cholesterol.
Collapse
Affiliation(s)
- Daniel Foest
- Miniaturisation, Leibniz-Institut für Analytische Wissenschaften─ISAS─e.V., Bunsen-Kirchhoff-Str. 11, 44139 Dortmund, Germany
| | - Alexander Knodel
- Miniaturisation, Leibniz-Institut für Analytische Wissenschaften─ISAS─e.V., Bunsen-Kirchhoff-Str. 11, 44139 Dortmund, Germany
| | - Robert Ahrends
- Department of Analytical Chemistry, University of Vienna, Währingstr. 38, A-1090 Vienna, Austria
| | - Cristina Coman
- Department of Analytical Chemistry, University of Vienna, Währingstr. 38, A-1090 Vienna, Austria
| | - Joachim Franzke
- Miniaturisation, Leibniz-Institut für Analytische Wissenschaften─ISAS─e.V., Bunsen-Kirchhoff-Str. 11, 44139 Dortmund, Germany
| | - Sebastian Brandt
- Miniaturisation, Leibniz-Institut für Analytische Wissenschaften─ISAS─e.V., Bunsen-Kirchhoff-Str. 11, 44139 Dortmund, Germany
| |
Collapse
|
45
|
Pakira V, Agarwal R, Chatterjee S, Mukherjee A, Chakraborty S. Lipidest: a lipid profile screening test under extreme point of care settings using a portable spinning disc and an office scanner. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2023; 15:2427-2440. [PMID: 37191178 DOI: 10.1039/d3ay00412k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
The demand for lipid profile (the cholesterol and triglyceride elements in the blood) testing outside resourced diagnostic centers is continuously increasing for personalized and community-based healthcare to ensure timely disease screening and management; however, it is inevitably challenged by several bottlenecks in the existing point of care technologies. These deficits include delicate sample pre-processing steps and device complexity, which give rise to unfavourable cost propositions to safeguard against compromised test accuracy. To circumvent these bottlenecks, herein, we introduce a new diagnostic technology, 'Lipidest', that integrates a portable spinning disc, a spin box, and an office scanner to reliably quantify the complete lipid panel from finger-prick blood. Our design facilitates the direct miniature adaptation of the established gold standard procedures as against any indirect sensing technologies that are otherwise common in point-of-care applications introduced commercially. The test procedure harmoniously connects all the elements of sample-to-answer integration in a single device, traversing the entire pipeline of the physical separation of plasma from the cellular components of the whole blood, the automated mixing with the test reagents on the same platform in situ, and office-scanner-adapted quantitative colorimetric analytics that eliminate any undesirable artefacts on account of variabilities in the background illumination and camera specifications. The exclusive value of eliminating sample preparation steps, including the rotationally actuated segregation of the specific blood constituents without any cross-interference between them, their automated homogeneous mixing with the respective test reagents, and the simultaneous, yet independent, quantitative readout without specialized instrumentation, render the test user-friendly and deployable in resource-constrained settings with a reasonably wide detection window. The extreme simplicity and modular nature of the device further make it amenable to mass manufacturing without incurring unfavourable costs. Extensive validation with laboratory-benchmark gold standards provide acceptable accuracy and indicates the value of the first-of-its-kind ultra-low-cost extreme-point-of-care test with a scientific foundation akin to highly accurate laboratory-centric technologies for cardiovascular health monitoring and beyond.
Collapse
Affiliation(s)
- Victor Pakira
- Advanced Technology Development Centre, Indian Institute of Technology Kharagpur, Kharagpur, 721302, India
| | - Rahul Agarwal
- Department of Mechanical Engineering, Indian Institute of Technology Kharagpur, Kharagpur, 721302, India.
| | - Subhamoy Chatterjee
- Department of Electronics and Electrical Communication Engineering, Indian Institute of Technology Kharagpur, Kharagpur, 721302, India
| | - Arghya Mukherjee
- Department of Mechanical Engineering, Indian Institute of Technology Kharagpur, Kharagpur, 721302, India.
| | - Suman Chakraborty
- Department of Mechanical Engineering, Indian Institute of Technology Kharagpur, Kharagpur, 721302, India.
| |
Collapse
|
46
|
Goicoechea L, Conde de la Rosa L, Torres S, García-Ruiz C, Fernández-Checa JC. Mitochondrial cholesterol: Metabolism and impact on redox biology and disease. Redox Biol 2023; 61:102643. [PMID: 36857930 PMCID: PMC9989693 DOI: 10.1016/j.redox.2023.102643] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/10/2023] [Accepted: 02/22/2023] [Indexed: 02/26/2023] Open
Abstract
Cholesterol is a crucial component of membrane bilayers by regulating their structural and functional properties. Cholesterol traffics to different cellular compartments including mitochondria, whose cholesterol content is low compared to other cell membranes. Despite the limited availability of cholesterol in the inner mitochondrial membrane (IMM), the metabolism of cholesterol in the IMM plays important physiological roles, acting as the precursor for the synthesis of steroid hormones and neurosteroids in steroidogenic tissues and specific neurons, respectively, or the synthesis of bile acids through an alternative pathway in the liver. Accumulation of cholesterol in mitochondria above physiological levels has a negative impact on mitochondrial function through several mechanisms, including the limitation of crucial antioxidant defenses, such as the glutathione redox cycle, increased generation of reactive oxygen species and consequent oxidative modification of cardiolipin, and defective assembly of respiratory supercomplexes. These adverse consequences of increased mitochondrial cholesterol trafficking trigger the onset of oxidative stress and cell death, and, ultimately, contribute to the development of diverse diseases, including metabolic liver diseases (i.e. fatty liver disease and liver cancer), as well as lysosomal disorders (i.e. Niemann-Pick type C disease) and neurodegenerative diseases (i.e. Alzheimer's disease). In this review, we summarize the metabolism and regulation of mitochondrial cholesterol and its potential impact on liver and neurodegenerative diseases.
Collapse
Affiliation(s)
- Leire Goicoechea
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain; Liver Unit, Hospital Clinic i Provincial de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red (CIBEREHD), Barcelona, Spain
| | - Laura Conde de la Rosa
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain; Liver Unit, Hospital Clinic i Provincial de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red (CIBEREHD), Barcelona, Spain
| | - Sandra Torres
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain; Liver Unit, Hospital Clinic i Provincial de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red (CIBEREHD), Barcelona, Spain
| | - Carmen García-Ruiz
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain; Liver Unit, Hospital Clinic i Provincial de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red (CIBEREHD), Barcelona, Spain; Research Center for ALPD, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
| | - José C Fernández-Checa
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain; Liver Unit, Hospital Clinic i Provincial de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red (CIBEREHD), Barcelona, Spain; Research Center for ALPD, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
| |
Collapse
|
47
|
Frutos MFD, Pardo-Marqués V, Torrecilla-Parra M, Rada P, Pérez-García A, Martín-Martín Y, de la Peña G, Gómez A, Toledano-Zaragoza A, Gómez-Coronado D, Casarejos MJ, Solís JM, Rotllán N, Pastor Ó, Ledesma MD, Valverde ÁM, Busto R, Ramírez CM. "MiR-7 controls cholesterol biosynthesis through posttranscriptional regulation of DHCR24 expression". BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2023; 1866:194938. [PMID: 37086967 DOI: 10.1016/j.bbagrm.2023.194938] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 04/13/2023] [Accepted: 04/14/2023] [Indexed: 04/24/2023]
Abstract
Dysregulation of cholesterol homeostasis is associated with several pathologies including cardiovascular diseases and neurological disorders such as Alzheimer's disease (AD). MicroRNAs (miRNAs) have emerged as key post-transcriptional regulators of cholesterol metabolism. We previously established the role of miR-7 in regulating insulin resistance and amyloidosis, which represents a common pathological feature between type 2 diabetes and AD. We show here an additional metabolic function of miR-7 in cholesterol biosynthesis. We found that miR-7 blocks the last steps of the cholesterol biosynthetic pathway in vitro by targeting relevant genes including DHCR24 and SC5D posttranscriptionally. Intracranial infusion of miR-7 on an adeno-associated viral vector reduced the expression of DHCR24 in the brain of wild-type mice, supporting in vivo miR-7 targeting. We also found that cholesterol regulates endogenous levels of miR-7 in vitro, correlating with transcriptional regulation through SREBP2 binding to its promoter region. In parallel to SREBP2 inhibition, the levels of miR-7 and hnRNPK (the host gene of miR-7) were concomitantly reduced in brain in a mouse model of Niemann Pick type C1 disease and in murine fatty liver, which are both characterized by intracellular cholesterol accumulation. Taken together, the results establish a novel regulatory feedback loop by which miR-7 modulates cholesterol homeostasis at the posttranscriptional level, an effect that could be exploited for therapeutic interventions against prevalent human diseases.
Collapse
Affiliation(s)
| | | | | | - Patricia Rada
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERdem), Instituto de Salud Carlos III, Madrid, Spain
| | - Ana Pérez-García
- IMDEA Research Institute of Food & Health Sciences, Madrid, Spain
| | | | - Gema de la Peña
- Department of Biochemistry-Research, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| | - Ana Gómez
- Department of Neurobiology-Research, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| | - Ana Toledano-Zaragoza
- Department of Molecular Neuropathology, Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Madrid, Spain
| | - Diego Gómez-Coronado
- Department of Biochemistry-Research, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| | - María José Casarejos
- Department of Neurobiology-Research, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| | - José M Solís
- Department of Neurobiology-Research, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| | - Noemí Rotllán
- Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Óscar Pastor
- Department of Clinical Biochemistry, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| | - María Dolores Ledesma
- Department of Molecular Neuropathology, Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Madrid, Spain
| | - Ángela M Valverde
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERdem), Instituto de Salud Carlos III, Madrid, Spain
| | - Rebeca Busto
- Department of Biochemistry-Research, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain.
| | | |
Collapse
|
48
|
Estrada-Cruz NA, Manuel-Apolinar L, Segura-Uribe JJ, Almanza-Pérez JC, Fortis-Barrera Á, Orozco-Suárez S, Bautista-Poblet G, Coyoy-Salgado A, Guerra-Araiza C. Short-term administration of tibolone reduces inflammation and oxidative stress in the hippocampus of ovariectomized rats fed high-fat and high-fructose. Nutr Neurosci 2023; 26:275-289. [PMID: 35282801 DOI: 10.1080/1028415x.2022.2046964] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Inflammation and oxidative stress are critical events involved in neurodegeneration. In animal models, it has been shown that chronic consumption of a hypercaloric diet, which leads to inflammatory processes, affects the hippocampus, a brain region fundamental for learning and memory processes. In addition, advanced age and menopause are risk factors for neurodegeneration. Hormone replacement therapy (HRT) ameliorates menopause symptoms. Tibolone (TB), a synthetic hormone, exerts estrogenic, progestogenic and androgenic effects on different tissues. We aimed to determine the effect of short-term TB administration on oxidative stress and inflammation markers in the hippocampus of ovariectomized rats fed a high-fat-and-fructose diet (HFFD). Adult female rats were ovariectomized (OVX) and fed standard diet or HFFD-consisting of 10% lard supplemented chow and 20% high-fructose syrup in the drinking water-and administered vehicle or TB (1 mg/kg for seven days). Finally, we administered hormone receptor antagonists (MPP, RU486 or FLU) to each of the OVX + HFFD + TB groups. Bodyweight, triglycerides and cholesterol, oxidative stress and inflammation markers, and the activity and expression of antioxidant enzymes were quantified in the hippocampus of each experimental group. We observed that short-term TB administration significantly reduced body weight, AGEs, MDA levels, increased SOD and GPx activity, improved GSH/GSSG ratio, and reduced IL-6 and TNF-α. Our findings suggest that short-term administration of TB decreases oxidative stress and reduces inflammation caused by HFFD and early estrogenic decline. These effects occurred via estrogen receptor alpha.
Collapse
Affiliation(s)
- Norma A Estrada-Cruz
- Unidad de Investigación Médica en Farmacología, Centro Médico Nacional (CMN) Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Leticia Manuel-Apolinar
- Unidad de Investigación Médica en Enfermedades Endocrinas, CMN Siglo XXI, IMSS, Mexico City, Mexico
| | - Julia J Segura-Uribe
- Subdirección de Gestión de la Investigación, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| | - Julio C Almanza-Pérez
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, UAM-I, Mexico City, Mexico
| | - Ángeles Fortis-Barrera
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, UAM-I, Mexico City, Mexico
| | - Sandra Orozco-Suárez
- Unidad de Investigación Médica en Enfermedades Neurológicas, CMN Siglo XXI, IMSS, Mexico City, Mexico
| | - Guadalupe Bautista-Poblet
- Unidad de Investigación Médica en Farmacología, Centro Médico Nacional (CMN) Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Angélica Coyoy-Salgado
- Cátedras CONACyT-Unidad de Investigación Médica en Enfermedades Neurológicas, IMSS, Mexico City, Mexico
| | - Christian Guerra-Araiza
- Unidad de Investigación Médica en Farmacología, Centro Médico Nacional (CMN) Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| |
Collapse
|
49
|
Dib S, Loiola RA, Sevin E, Saint-Pol J, Shimizu F, Kanda T, Pahnke J, Gosselet F. TNFα Activates the Liver X Receptor Signaling Pathway and Promotes Cholesterol Efflux from Human Brain Pericytes Independently of ABCA1. Int J Mol Sci 2023; 24:ijms24065992. [PMID: 36983062 PMCID: PMC10056409 DOI: 10.3390/ijms24065992] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/13/2023] [Accepted: 03/18/2023] [Indexed: 03/30/2023] Open
Abstract
Neuroinflammation and brain lipid imbalances are observed in Alzheimer's disease (AD). Tumor necrosis factor-α (TNFα) and the liver X receptor (LXR) signaling pathways are involved in both processes. However, limited information is currently available regarding their relationships in human brain pericytes (HBP) of the neurovascular unit. In cultivated HBP, TNFα activates the LXR pathway and increases the expression of one of its target genes, the transporter ATP-binding cassette family A member 1 (ABCA1), while ABCG1 is not expressed. Apolipoprotein E (APOE) synthesis and release are diminished. The cholesterol efflux is promoted, but is not inhibited, when ABCA1 or LXR are blocked. Moreover, as for TNFα, direct LXR activation by the agonist (T0901317) increases ABCA1 expression and the associated cholesterol efflux. However, this process is abolished when LXR/ABCA1 are both inhibited. Neither the other ABC transporters nor the SR-BI are involved in this TNFα-mediated lipid efflux regulation. We also report that inflammation increases ABCB1 expression and function. In conclusion, our data suggest that inflammation increases HBP protection against xenobiotics and triggers an LXR/ABCA1 independent cholesterol release. Understanding the molecular mechanisms regulating this efflux at the level of the neurovascular unit remains fundamental to the characterization of links between neuroinflammation, cholesterol and HBP function in neurodegenerative disorders.
Collapse
Affiliation(s)
- Shiraz Dib
- Blood-Brain Barrier Laboratory (LBHE), UR 2465, University of Artois, F-62300 Lens, France
| | - Rodrigo Azevedo Loiola
- Blood-Brain Barrier Laboratory (LBHE), UR 2465, University of Artois, F-62300 Lens, France
| | - Emmanuel Sevin
- Blood-Brain Barrier Laboratory (LBHE), UR 2465, University of Artois, F-62300 Lens, France
| | - Julien Saint-Pol
- Blood-Brain Barrier Laboratory (LBHE), UR 2465, University of Artois, F-62300 Lens, France
| | - Fumitaka Shimizu
- Department of Neurology and Clinical Neuroscience, Graduate School of Medicine, Yamaguchi University, Ube 755-8505, Japan
| | - Takashi Kanda
- Department of Neurology and Clinical Neuroscience, Graduate School of Medicine, Yamaguchi University, Ube 755-8505, Japan
| | - Jens Pahnke
- Department of Pathology, Section of Neuropathology, Translational Neurodegeneration Research and Neuropathology Lab, University of Oslo, Oslo University Hospital, Sognsvannsveien 20, 0372 Oslo, Norway
- Pahnke Lab (Drug Development and Chemical Biology), Lübeck Institute of Experimental Dermatology (LIED), University of Lübeck, University Medical Center Schleswig-Holstein, Ratzeburger Allee 160, 23538 Lübeck, Germany
- Department of Pharmacology, Faculty of Medicine, University of Latvia, Jelgavas iela 3, 1004 Riga, Latvia
- Department of Neurobiology, The Georg S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Fabien Gosselet
- Blood-Brain Barrier Laboratory (LBHE), UR 2465, University of Artois, F-62300 Lens, France
| |
Collapse
|
50
|
Puglisi A, Bognanni N, Vecchio G, Bayir E, van Oostrum P, Shepherd D, Platt F, Reimhult E. Grafting of Cyclodextrin to Theranostic Nanoparticles Improves Blood-Brain Barrier Model Crossing. Biomolecules 2023; 13:573. [PMID: 36979508 PMCID: PMC10046162 DOI: 10.3390/biom13030573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/19/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
Core-shell superparamagnetic iron oxide nanoparticles hold great promise as a theranostic platform in biological systems. Herein, we report the biological effect of multifunctional cyclodextrin-appended SPIONs (CySPION) in mutant Npc1-deficient CHO cells compared to their wild type counterparts. CySPIONs show negligible cytotoxicity while they are strongly endocytosed and localized in the lysosomal compartment. Through their bespoke pH-sensitive chemistry, these nanoparticles release appended monomeric cyclodextrins to mobilize over-accumulated cholesterol and eject it outside the cells. CySPIONs show a high rate of transport across blood-brain barrier models, indicating their promise as a therapeutic approach for cholesterol-impaired diseases affecting the brain.
Collapse
Affiliation(s)
- Antonino Puglisi
- Department of Bionanosciences, Institute of Biologically Inspired Materials, University of Natural Resources and Life Sciences (BOKU), 1190 Vienna, Austria
| | - Noemi Bognanni
- Department of Bionanosciences, Institute of Biologically Inspired Materials, University of Natural Resources and Life Sciences (BOKU), 1190 Vienna, Austria
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, 95125 Catania, Italy
| | - Graziella Vecchio
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, 95125 Catania, Italy
| | - Ece Bayir
- Central Research Testing and Analysis Laboratory Research and Application Center, Ege University Bornova, Izmir 35100, Turkey
| | - Peter van Oostrum
- Department of Bionanosciences, Institute of Biologically Inspired Materials, University of Natural Resources and Life Sciences (BOKU), 1190 Vienna, Austria
| | - Dawn Shepherd
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Frances Platt
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Erik Reimhult
- Department of Bionanosciences, Institute of Biologically Inspired Materials, University of Natural Resources and Life Sciences (BOKU), 1190 Vienna, Austria
| |
Collapse
|