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
|
Xing C, Zhang X, Wang D, Chen H, Gao X, Sun C, Guo W, Roshan S, Li Y, Hang Z, Cai S, Lei T, Bi W, Hou L, Li L, Wu Y, Li L, Zeng Z, Du H. Neuroprotective effects of mesenchymal stromal cells in mouse models of Alzheimer's Disease: The Mediating role of gut microbes and their metabolites via the Microbiome-Gut-Brain axis. Brain Behav Immun 2024; 122:510-526. [PMID: 39191350 DOI: 10.1016/j.bbi.2024.08.047] [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: 12/17/2023] [Revised: 08/03/2024] [Accepted: 08/24/2024] [Indexed: 08/29/2024] Open
Abstract
The intricacy and multifaceted nature of Alzheimer's disease (AD) necessitate therapies that target multiple aspects of the disease. Mesenchymal stromal cells (MSCs) emerge as potential agents to mitigate AD symptoms; however, whether their therapeutic efficacy involves modulation of gut microbiota and the microbiome-gut-brain axis (MGBA) remains unexplored. In this study, we evaluated the effects of three distinct MSCs types-derived from the umbilical cord (UCMSC), dental pulp (SHED), and adipose tissue (ADSC)-in an APP/PS1 mouse model of AD. In comparison to saline control, MSCs administration resulted in a significant reduction of behavioral disturbances, amyloid plaques, and phosphorylated tau in the hippocampus and frontal cortex, accompanied by an increase in neuronal count and Nissl body density across AD-afflicted brain regions. Through 16S rRNA gene sequencing, we identified partial restoration of gut microbial balance in AD mice post-MSCs treatment, evidenced by the elevation of neuroprotective Akkermansia and reduction of the AD-associated Sphingomonas. To examine whether gut microbiota involved in MSCs efficacy in treating AD, SHED with better anti-inflammatory and gut microbiota recovery effects among three MSCs, and another AD model 5 × FAD mice with earlier and more pathological proteins in brain than APP/PS1, were selected for further studies. Antibiotic-mediated gut microbial inactivation attenuated MSCs efficacy in 5 × FAD mice, implicating the involvement of gut microbiota in the therapeutic mechanism. Functional analysis of altered gut microbiota and targeted bile acid metabolism profiling revealed a significant enhancement in bile acid variety following MSCs therapy. A chief bile acid constituent, taurocholic acid (TCA), was orally administered to AD mice and similarly abated AD symptoms. Nonetheless, the disruption of intestinal neuronal integrity with enterotoxin abrogated the ameliorative impact of both MSCs and TCA treatments. Collectively, our findings substantiate that MSCs confer therapeutic benefits in AD within a paradigm that primarily involves regulation of gut microbiota and their metabolites through the MGBA.
Collapse
Affiliation(s)
- Cencan Xing
- Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, No. 30 XueYuan Road, Haidian District, Beijing 100083, China
| | - Xiaoshuang Zhang
- Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, No. 30 XueYuan Road, Haidian District, Beijing 100083, China; Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
| | - Donghui Wang
- Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, No. 30 XueYuan Road, Haidian District, Beijing 100083, China
| | - Hongyu Chen
- Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, No. 30 XueYuan Road, Haidian District, Beijing 100083, China
| | - Xiaoyu Gao
- Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, No. 30 XueYuan Road, Haidian District, Beijing 100083, China
| | - Chunbin Sun
- Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, No. 30 XueYuan Road, Haidian District, Beijing 100083, China
| | - Wenhua Guo
- Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, No. 30 XueYuan Road, Haidian District, Beijing 100083, China; Reproductive Center, Peking University Third Hospital, Beijing, China
| | - Shah Roshan
- Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, No. 30 XueYuan Road, Haidian District, Beijing 100083, China
| | - Yingxian Li
- Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, No. 30 XueYuan Road, Haidian District, Beijing 100083, China
| | - Zhongci Hang
- Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, No. 30 XueYuan Road, Haidian District, Beijing 100083, China
| | - Shanglin Cai
- Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, No. 30 XueYuan Road, Haidian District, Beijing 100083, China
| | - Tong Lei
- Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, No. 30 XueYuan Road, Haidian District, Beijing 100083, China
| | - Wangyu Bi
- Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, No. 30 XueYuan Road, Haidian District, Beijing 100083, China
| | - Liangxuan Hou
- Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, No. 30 XueYuan Road, Haidian District, Beijing 100083, China
| | - Luping Li
- Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, No. 30 XueYuan Road, Haidian District, Beijing 100083, China
| | - Yawen Wu
- Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, No. 30 XueYuan Road, Haidian District, Beijing 100083, China
| | - Liang Li
- Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, No. 30 XueYuan Road, Haidian District, Beijing 100083, China
| | - Zehua Zeng
- Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, No. 30 XueYuan Road, Haidian District, Beijing 100083, China.
| | - Hongwu Du
- Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, No. 30 XueYuan Road, Haidian District, Beijing 100083, China.
| |
Collapse
|
3
|
Zhang MY, Yin C, Ding L, Cheng L, Lv Q, Wang P, Zhang SB, You QY. Mechanism of Panax notoginseng saponins in improving cognitive impairment induced by chronic sleep deprivation based on the integrative analysis of serum metabolomics and network pharmacology. JOURNAL OF ETHNOPHARMACOLOGY 2024; 337:118988. [PMID: 39447711 DOI: 10.1016/j.jep.2024.118988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 10/26/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Panax notoginseng saponin (PNS) has a variety of biological activities, such as improvement of myocardial ischemia, improvement of learning and memory, hypolipidemia, and immunomodulation. However, its protective mechanism on the central nervous system (CNS) is not clear. AIM OF THE STUDY The present study initially evaluated the possible mechanism of PNS to improve cognitive dysfunction due to chronic sleep deprivation (CSD). MATERIALS AND METHODS In the present study, we used a modified multi-platform aquatic environment sleep deprivation method to induce a cognitively impaired rat model, and explored the mechanism of action of PNS by integrating serum metabolomics and network pharmacology, which was further verified by molecular docking and experiments. RESULTS The results showed that PNS significantly shortened the escape latency, increased the target quadrant time and the number of traversing platforms, and attenuated the inflammatory damage in the hippocampal Cornu Ammonis 1 (CA1) region in CSD rats. The non-targeted metabolomics results indicated that 35 biomarkers significantly altered following PNS therapy intervention, with metabolic pathways enriched for the effects of One carbon pool by folate, Riboflavin metabolism, Glycerophospholipid metabolism, Sphingolipid metabolism, Glycerolipid metabolism, Arachidonic acid metabolism, and Tryptophan metabolism. In addition, network pharmacology identified 234 potential targets for PNS intervention in CSD with cognitive impairment. Metabolite-response-enzyme-gene network was constructed by MetaScape and matched with the network pharmacology results to identify a total of five shared targets (LPL, GPAM, HSD11B1, HSD11B2, and SULT2A1) and two metabolic pathways (Sphingolipid metabolism and Steroid hormone biosynthesis). The results of molecular docking revealed that the five active ingredients had good binding ability with the five core targets. qPCR analysis confirmed the ability of PNS to modulate the above five targets. CONCLUSIONS The combination of metabolomics and network analysis provides a scientific basis for promoting the clinical application of PNS in cognitive impairment.
Collapse
Affiliation(s)
- Mei-Ya Zhang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China; Hubei Shizhen Laboratory, Hubei University of Chinese Medicine, Wuhan, 430065, China; Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Chao Yin
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China; Hubei Shizhen Laboratory, Hubei University of Chinese Medicine, Wuhan, 430065, China; Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Li Ding
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China; Hubei Shizhen Laboratory, Hubei University of Chinese Medicine, Wuhan, 430065, China; Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Li Cheng
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Qing Lv
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Ping Wang
- Hubei Shizhen Laboratory, Hubei University of Chinese Medicine, Wuhan, 430065, China; Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Shun-Bo Zhang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China; Hubei Shizhen Laboratory, Hubei University of Chinese Medicine, Wuhan, 430065, China; Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Qiu-Yun You
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China; Hubei Shizhen Laboratory, Hubei University of Chinese Medicine, Wuhan, 430065, China; Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| |
Collapse
|
4
|
Jeong C, Jung JH, Oh DJ, Han K, Kim MK. Association between fluctuations in blood cholesterol levels and the risk of suicide death in the general population. J Psychosom Res 2024; 187:111956. [PMID: 39490294 DOI: 10.1016/j.jpsychores.2024.111956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 10/09/2024] [Accepted: 10/12/2024] [Indexed: 11/05/2024]
Abstract
OBJECTIVE Suicide is a pressing global public health issue. While recent studies have explored the association between serum cholesterol levels and suicide risk, the association between cholesterol variability and suicide risk is not well characterized. METHODS This was a nationwide population-based cohort study using data from the Korean National Health Insurance Service database. A total of 1,983,701 patients with at least three measurements of TC between 2004 and 2009 were included. Participants were followed until death by suicide or the study's end in December 2021. Participants were categorized based on baseline TC or TC variability. TC variability was assessed using coefficient of variation (CV), variability independent of the mean (VIM), and average successive variability (ASV). RESULTS Over a median follow-up of 11 years, 5883 (0.3 %) patients died by suicide. Compared to subjects with TC <200 mg/dL, those with TC ≥240 mg/dL had a lower risk of suicide death (hazard ratio [HR]: 0.85, 95 % confidence interval [95 % CI]: 0.78-0.93). In the unadjusted model, the HR for suicide death in the highest quartile (Q4) compared to the lowest quartile (Q1) of TC variability was 1.36 (95 % CI: 1.26-1.46). After adjusting for potential confounders, high variability in TC levels was associated with a higher risk of suicide death (HR 1.27, 95 % CI: 1.18-1.37). When analyzed based on the combination of TC baseline and variability, the highest quartile of TC variability showed a higher rate of suicide death compared to the lowest quartile, regardless of baseline TC level. CONCLUSION High TC variability and low TC levels were associated with an increased risk of suicide.
Collapse
Affiliation(s)
- Chaiho Jeong
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jin-Hyung Jung
- Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Dae Jong Oh
- Workplace Mental Health Institute, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Kyungdo Han
- Department of Medical Statistics, Soongsil University of Korea, Seoul, Republic of Korea.
| | - Mee Kyoung Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
5
|
Meng H, Elliott A, Mansfield J, Bailey M, Frogley M, Cinque G, Moger J, Stone N, Tamagnini F, Palombo F. Identification of tauopathy-associated lipid signatures in Alzheimer's disease mouse brain using label-free chemical imaging. Commun Biol 2024; 7:1341. [PMID: 39420210 PMCID: PMC11487145 DOI: 10.1038/s42003-024-07034-3] [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: 05/23/2024] [Accepted: 10/08/2024] [Indexed: 10/19/2024] Open
Abstract
There is cumulative evidence that lipid metabolism plays a key role in the pathogenesis of various neurodegenerative disorders including Alzheimer's disease (AD). Visualising lipid content in a non-destructive label-free manner can aid in elucidating the AD phenotypes towards a better understanding of the disease. In this study, we combined multiple optical molecular-specific methods, Fourier transform infrared (FTIR) spectroscopic imaging, synchrotron radiation-infrared (SR-IR) microscopy, Raman and stimulated Raman scattering (SRS) microscopy, and optical-photothermal infrared (O-PTIR) microscopy with multivariate data analysis, to investigate the biochemistry of brain hippocampus in situ using a mouse model of tauopathy (rTg4510). We observed a significant difference in the morphology and lipid content between transgenic (TG) and wild type (WT) samples. Immunohistochemical staining revealed some degree of microglia co-localisation with elevated lipids in the brain. These results provide new evidence of tauopathy-related dysfunction in a preclinical study at a subcellular level.
Collapse
Affiliation(s)
- Hao Meng
- Department of Physics and Astronomy, University of Exeter, Exeter, EX4 4QL, UK
| | - Alicia Elliott
- Department of Physics and Astronomy, University of Exeter, Exeter, EX4 4QL, UK
| | - Jessica Mansfield
- Department of Physics and Astronomy, University of Exeter, Exeter, EX4 4QL, UK
| | - Michelle Bailey
- Department of Physics and Astronomy, University of Exeter, Exeter, EX4 4QL, UK
| | - Mark Frogley
- Diamond Light Source, MIRIAM beamline B22, Harwell Science & Innovation Campus, Didcot, OX11 0DE, UK
| | - Gianfelice Cinque
- Diamond Light Source, MIRIAM beamline B22, Harwell Science & Innovation Campus, Didcot, OX11 0DE, UK
| | - Julian Moger
- Department of Physics and Astronomy, University of Exeter, Exeter, EX4 4QL, UK
| | - Nick Stone
- Department of Physics and Astronomy, University of Exeter, Exeter, EX4 4QL, UK
| | - Francesco Tamagnini
- School of Pharmacy, University of Reading, Reading, RG6 6UB, UK
- Centro Studi Biomedici, Università degli Studi della Repubblica di San Marino, Salita alla Rocca, 44 - 47890, San Marino Città, Republic of San Marino
| | - Francesca Palombo
- Department of Physics and Astronomy, University of Exeter, Exeter, EX4 4QL, UK.
| |
Collapse
|
6
|
Chauhan P, Wadhwa K, Singh G, Gupta S, Iqbal D, Abomughaid MM, Almutary AG, Mishra PC, Nelson VK, Jha NK. Exploring complexities of Alzheimer's disease: New insights into molecular and cellular mechanisms of neurodegeneration and targeted therapeutic interventions. Ageing Res Rev 2024:102548. [PMID: 39419399 DOI: 10.1016/j.arr.2024.102548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/19/2024]
Abstract
Alzheimer's disease (AD), the common form of dementia globally, is a complex condition including neurodegeneration; shares incompletely known pathogenesis. Signal transduction and biological activities, including cell metabolism, growth, and death are regulated by different signaling pathways including AKT/MAPK, Wnt, Leptin, mTOR, ubiquitin, Sirt1, and insulin. Absolute evidence linking specific molecular pathways with the genesis and/or progression of AD is still lacking. Changes in gut microbiota and blood-brain barrier also cause amyloid β aggregation in AD. The current review reports significant characteristics of various signaling pathways, their relationship with each other, and how they interact in disease genesis and/or progression. Nevertheless, due to the enormous complexity of the brain and numerous chemical linkages between these pathways, the use of signaling pathways as possible targets for drug development against AD is minimal. Currently, there is no permanent cure for AD, and there is no way to stop brain cell loss. This review also aimed to draw attention to the role of a novel group of signaling pathways, which can be collectively dubbed "anti-AD pathways", in multi-target therapy for AD, where cellular metabolic functions are severely impaired. Thus, different hypotheses have been formulated and elaborated to explain the genesis of AD, which can be further explored for drug development too.
Collapse
Affiliation(s)
- Payal Chauhan
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Karan Wadhwa
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Govind Singh
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, 124001, India.
| | - Saurabh Gupta
- Deparment of Biotechnology, GLA University, Mathura, India
| | - Danish Iqbal
- Department of Health Information Management, College of Applied Medical Sciences, Buraydah Private Colleges, Buraydah 51418, Saudi Arabia
| | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha, 61922, Saudi Arabia
| | - Abdulmajeed G Almutary
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi P.O. Box 59911, United Arab Emirates
| | - Prabhu Chandra Mishra
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, India
| | - Vinod Kumar Nelson
- Raghavendra Institute of Pharmaceutical Education and Research, Anantapur, India
| | - Niraj Kumar Jha
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Centre of Research Impact and Outreach, Chitkara University Institute of Engineering and Technology, Chitkara University, Punjab, India; School of Bioengineering & Biosciences, Lovely Professional University, Phagwara 144411, India.
| |
Collapse
|
7
|
Chase BA, Frigerio R, Yucus CJ, Patel S, Maraganore D, Sanders AR, Duan J, Markopoulou K. Lipid Trajectories Improve Risk Models for Alzheimer's Disease and Mild Cognitive Impairment. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.27.24314494. [PMID: 39399044 PMCID: PMC11469357 DOI: 10.1101/2024.09.27.24314494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
To assess the relationship between lipids and cognitive dysfunction, we retrospectively analyzed blood-lipid levels in clinically well-characterized individuals with stable mild cognitive impairment (MCI) or Alzheimer's disease (AD) over the decade prior to first cognitive symptoms. In this case/control cohort study, AD and MCI cases were diagnosed using DSM-IV criteria; MCI cases had not progressed to dementia for ≥5 years; and controls were propensity matched to cases at age of symptom onset (MCI: 116 cases, 435 controls; AD: 215 cases, 483 controls). Participants were grouped based on longitudinal trajectories and quintile of variability independent of the mean (VIM) for total cholesterol, HDL-C, LDL-C, non-HDL-C and ln(triglycerides). Models for the risk of cognitive dysfunction evaluated trajectory and VIM groups, APOE genotype, polygenic risk scores (PRS) for AD and lipid levels, age, comorbidities, and longitudinal correlates of blood-lipid concentrations. Lower HDL-C trajectories (OR = 3.8, 95% CI = 1.3-11.3) and the lowest VIM quintile of non-HDL-C (OR = 2.2, 95% CI = 1.3-3.0) were associated with higher MCI risk. Lower HDL-C trajectories (OR = 3.0, 95% CI = 1.6-5.7) and the lowest VIM quintile of total cholesterol (OR = 2.4, 95% CI = 1.5-3.9) were associated with higher AD risk. The inclusion of lipid-trajectory and VIM groups improved risk-model predictive performance independent of APOE genotype or PRS for AD and lipid levels. These results provide an important real-world perspective on the influence of lipid metabolism and blood-lipid levels on the development of stable MCI and AD.
Collapse
Affiliation(s)
- Bruce A. Chase
- Information Technology, Endeavor Health, Skokie, IL USA
- Pritzker School of Medicine, Chicago, USA
| | - Roberta Frigerio
- Pritzker School of Medicine, Chicago, USA
- Research Institute, Endeavor Health, Evanston, IL USA
| | - Chad J. Yucus
- Department of Neurology, Endeavor Health, Evanston, IL USA
| | - Smita Patel
- Department of Neurology, Endeavor Health, Evanston, IL USA
| | - Demetrius Maraganore
- Department of Neurology, Tulane University School of Medicine, New Orleans, LA USA
| | - Alan R. Sanders
- Center for Psychiatric Genetics, Endeavor Health Research Institute, Evanston, IL USA
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL USA
| | - Jubao Duan
- Center for Psychiatric Genetics, Endeavor Health Research Institute, Evanston, IL USA
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL USA
| | - Katerina Markopoulou
- Department of Neurology, Endeavor Health, Evanston, IL USA
- Department of Neurology, Pritzker School of Medicine, University of Chicago, Chicago, IL USA
| |
Collapse
|
8
|
Ury-Thiery V, Fichou Y, Alves I, Molinari M, Lecomte S, Feuillie C. Interaction of full-length Tau with negatively charged lipid membranes leads to polymorphic aggregates. NANOSCALE 2024; 16:17141-17153. [PMID: 39189914 DOI: 10.1039/d4nr01343c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
The Tau protein is implicated in various diseases collectively known as tauopathies, including Alzheimer's disease and frontotemporal dementia. The precise mechanism underlying Tau pathogenicity remains elusive. Recently, the role of lipids has garnered interest due to their implications in Tau aggregation, secretion, uptake, and pathogenic dysregulation. Previous investigations have highlighted critical aspects: (i) Tau's tendency to aggregate into fibers when interacting with negatively charged lipids, (ii) its ability to form structured species upon contact with anionic membranes, and (iii) the potential disruption of the membrane upon Tau binding. In this study, we examine the disease-associated P301L mutation of the 2N4R isoform of Tau and its effects on membranes composed on phosphatidylserine (PS) lipids. Aggregation studies and liposome leakage assays demonstrate Tau's ability to bind to anionic lipid vesicles, leading to membrane disruption. Attenuated total reflection Fourier-transform infrared spectroscopy (ATR-FTIR) reveals the accumulation of Tau on the membrane surface without protein insertion, structuration, or lipid removal. Plasmon waveguide resonance (PWR) demonstrates a strong binding of Tau on PS bilayers with an apparent Kd in the micromolar range, indicating the deposition of a thick protein layer. Atomic force microscopy (AFM) real-time imaging allows the observation of partial lipid solubilization and the deposition of polymorphic aggregates in the form of thick patches and fibrillary structures resembling amyloid fibers, which could grow from a combination of extracted anionic phospholipids from the membrane and Tau protein. This study deepens our understanding of full-length Tau's multifaceted interactions with lipids, shedding light on potential mechanisms leading to the formation of pathogenic Tau assemblies.
Collapse
Affiliation(s)
- Vicky Ury-Thiery
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, F-33600 Pessac, France.
| | - Yann Fichou
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, F-33600 Pessac, France.
| | - Isabel Alves
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, F-33600 Pessac, France.
| | - Michael Molinari
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, F-33600 Pessac, France.
| | - Sophie Lecomte
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, F-33600 Pessac, France.
| | - Cécile Feuillie
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, F-33600 Pessac, France.
| |
Collapse
|
9
|
Wang W, Xia H. The role of lipid species in Alzheimer's disease onset: A comprehensive Mendelian randomization analysis. Brain Res 2024; 1846:149238. [PMID: 39278307 DOI: 10.1016/j.brainres.2024.149238] [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: 05/05/2024] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 09/18/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) remains a significant global health challenge, with its etiology intricately linked to a variety of genetic and environmental factors. Among these, lipid metabolism has been hypothesized to play a crucial role, though the causal pathways remain inadequately elucidated. This study aims to employ Mendelian Randomization (MR) to unravel the potential causal relationships between a comprehensive array of lipid species and the risk of developing AD. METHODS Utilizing a two-sample MR framework, we analyzed data from genome-wide association studies (GWAS) encompassing 487,511 individuals of European descent. A total of 179 lipid species across 13 lipid categories were investigated for their causal association with AD. Genetic variants serving as instrumental variables (IVs) were carefully selected based on stringent criteria to ensure validity. The statistical analyses, including inverse variance weighting (IVW), weighted median-based estimation, and sensitivity analyses, were conducted using the R software environment. RESULTS Our findings reveal a significant causal relationship between ten specific lipid species and the risk of AD. Notably, certain lipids such as Sterol ester (27:1/15:0) and Phosphatidylcholine (16:0_22:4) exhibited a protective effect against AD, as evidenced by their inverse correlation with the disease's risk. Additionally, a reciprocal analysis suggested a negative causal impact of AD on the levels of certain Triacylglycerol species. The integrity of our results was reinforced by sensitivity analyses, including the MR Egger intercept test, indicating the absence of horizontal pleiotropy and confirming the reliability of our findings. CONCLUSIONS This study substantiates the causal link between specific lipid species and Alzheimer's disease, highlighting the complex interplay between lipid metabolism and AD pathogenesis. The identified lipid biomarkers offer new insights into the disease's etiology and potential therapeutic targets. Furthermore, our rigorous methodological approach demonstrates the utility of MR in disentangling the causal relationships in complex diseases.
Collapse
Affiliation(s)
- Wen Wang
- Department of Gerontology, Tongde Hospital of Zhejiang Province, Psychiatric and Geriatric Critical Medicine Department, Tongde Hospital of Zhejiang Province, China
| | - HongLian Xia
- Department of Gerontology, Tongde Hospital of Zhejiang Province, No. 234, Gucui Road, Hangzhou, China.
| |
Collapse
|
10
|
Cao Y, Zhao LW, Chen ZX, Li SH. New insights in lipid metabolism: potential therapeutic targets for the treatment of Alzheimer's disease. Front Neurosci 2024; 18:1430465. [PMID: 39323915 PMCID: PMC11422391 DOI: 10.3389/fnins.2024.1430465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 08/14/2024] [Indexed: 09/27/2024] Open
Abstract
Alzheimer's disease (AD) is increasingly recognized as being intertwined with the dysregulation of lipid metabolism. Lipids are a significant class of nutrients vital to all organisms, playing crucial roles in cellular structure, energy storage, and signaling. Alterations in the levels of various lipids in AD brains and dysregulation of lipid pathways and transportation have been implicated in AD pathogenesis. Clinically, evidence for a high-fat diet firmly links disrupted lipid metabolism to the pathogenesis and progression of AD, although contradictory findings warrant further exploration. In view of the significance of various lipids in brain physiology, the discovery of complex and diverse mechanisms that connect lipid metabolism with AD-related pathophysiology will bring new hope for patients with AD, underscoring the importance of lipid metabolism in AD pathophysiology, and promising targets for therapeutic intervention. Specifically, cholesterol, sphingolipids, and fatty acids have been shown to influence amyloid-beta (Aβ) accumulation and tau hyperphosphorylation, which are hallmarks of AD pathology. Recent studies have highlighted the potential therapeutic targets within lipid metabolism, such as enhancing apolipoprotein E lipidation, activating liver X receptors and retinoid X receptors, and modulating peroxisome proliferator-activated receptors. Ongoing clinical trials are investigating the efficacy of these strategies, including the use of ketogenic diets, statin therapy, and novel compounds like NE3107. The implications of these findings suggest that targeting lipid metabolism could offer new avenues for the treatment and management of AD. By concentrating on alterations in lipid metabolism within the central nervous system and their contribution to AD development, this review aims to shed light on novel research directions and treatment approaches for combating AD, offering hope for the development of more effective management strategies.
Collapse
Affiliation(s)
- Yuan Cao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, Zhengzhou, China
- Clinical Systems Biology Laboratories, Translation Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Lin-Wei Zhao
- Department of Cardiology, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou University Central China Fuwai Hospital, Zhengzhou, China
| | - Zi-Xin Chen
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, Zhengzhou, China
- Clinical Systems Biology Laboratories, Translation Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Shao-Hua Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, Zhengzhou, China
- Clinical Systems Biology Laboratories, Translation Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
11
|
Barros LF, Schirmeier S, Weber B. The Astrocyte: Metabolic Hub of the Brain. Cold Spring Harb Perspect Biol 2024; 16:a041355. [PMID: 38438188 PMCID: PMC11368191 DOI: 10.1101/cshperspect.a041355] [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: 03/06/2024]
Abstract
Astrocytic metabolism has taken center stage. Interposed between the neuron and the vasculature, astrocytes exert control over the fluxes of energy and building blocks required for neuronal activity and plasticity. They are also key to local detoxification and waste recycling. Whereas neurons are metabolically rigid, astrocytes can switch between different metabolic profiles according to local demand and the nutritional state of the organism. Their metabolic state even seems to be instructive for peripheral nutrient mobilization and has been implicated in information processing and behavior. Here, we summarize recent progress in our understanding of astrocytic metabolism and its effects on metabolic homeostasis and cognition.
Collapse
Affiliation(s)
- L Felipe Barros
- Centro de Estudios Científicos, Valdivia 5110465, Chile
- Universidad San Sebastián, Facultad de Medicina y Ciencia, Valdivia 5110693, Chile
| | - Stefanie Schirmeier
- Technische Universität Dresden, Department of Biology, 01217 Dresden, Germany
| | - Bruno Weber
- University of Zurich, Institute of Pharmacology and Toxicology, 8057 Zurich, Switzerland
| |
Collapse
|
12
|
Wu M, Cheng Y, Zhang R, Han W, Jiang H, Bi C, Zhang Z, Ye M, Lin X, Liu Z. Molecular mechanism and therapeutic strategy of bile acids in Alzheimer's disease from the emerging perspective of the microbiota-gut-brain axis. Biomed Pharmacother 2024; 178:117228. [PMID: 39088965 DOI: 10.1016/j.biopha.2024.117228] [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: 04/21/2024] [Revised: 07/19/2024] [Accepted: 07/28/2024] [Indexed: 08/03/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by the accumulation of amyloid-β outside neurons and Tau protein inside neurons. Various pathological mechanisms are implicated in AD, including brain insulin resistance, neuroinflammation, and endocrinal dysregulation of adrenal corticosteroids. These factors collectively contribute to neuronal damage and destruction. Recently, bile acids (BAs), which are metabolites of cholesterol, have shown neuroprotective potential against AD by targeting the above pathological changes. BAs can enter the systematic circulation and cross the blood-brain barrier, subsequently exerting neuroprotective effects by targeting several endogenous receptors. Additionally, BAs interact with the microbiota-gut-brain (MGB) axis to improve immune and neuroendocrine function during AD episodes. Gut microbes impact BA signaling in the brain through their involvement in BA biotransformation. In this review, we summarize the role and molecular mechanisms of BAs in AD while considering the MGB axis and propose novel strategies for preventing the onset and progression of AD.
Collapse
Affiliation(s)
- Menglu Wu
- Clinical Laboratory, Shaoxing Seventh People's Hospital (Affiliated Mental Health Center, Medical College of Shaoxing University), Shaoxing, Zhejiang, China; Department of Behavioral Neurosciences, Science Research Center of Medical School, Shaoxing University, Shaoxing, Zhejiang, China
| | - Yongyi Cheng
- Department of Behavioral Neurosciences, Science Research Center of Medical School, Shaoxing University, Shaoxing, Zhejiang, China
| | - Ruolin Zhang
- Department of Behavioral Neurosciences, Science Research Center of Medical School, Shaoxing University, Shaoxing, Zhejiang, China
| | - Wenwen Han
- Department of Behavioral Neurosciences, Science Research Center of Medical School, Shaoxing University, Shaoxing, Zhejiang, China
| | - Hanqi Jiang
- Department of Behavioral Neurosciences, Science Research Center of Medical School, Shaoxing University, Shaoxing, Zhejiang, China
| | - Chenchen Bi
- Department of Behavioral Neurosciences, Science Research Center of Medical School, Shaoxing University, Shaoxing, Zhejiang, China
| | - Ziyi Zhang
- Department of Behavioral Neurosciences, Science Research Center of Medical School, Shaoxing University, Shaoxing, Zhejiang, China
| | - Mengfei Ye
- Department of Psychiatry, Shaoxing Seventh People's Hospital (Affiliated Mental Health Center, Medical College of Shaoxing University), Shaoxing, Zhejiang, China
| | - Xiuqin Lin
- Clinical Laboratory, Shaoxing Seventh People's Hospital (Affiliated Mental Health Center, Medical College of Shaoxing University), Shaoxing, Zhejiang, China.
| | - Zheng Liu
- Department of Behavioral Neurosciences, Science Research Center of Medical School, Shaoxing University, Shaoxing, Zhejiang, China; Department of Pharmacology, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China.
| |
Collapse
|
13
|
Arshavsky YI. Autoimmune hypothesis of Alzheimer's disease: unanswered question. J Neurophysiol 2024; 132:929-942. [PMID: 39163023 DOI: 10.1152/jn.00259.2024] [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: 06/18/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/21/2024] Open
Abstract
Alzheimer's disease (AD) was described more than a century ago. However, there are still no effective approaches to its treatment, which may suggest that the search for the cure is not being conducted in the most productive direction. AD begins as selective impairments of declarative memory with no deficits in other cognitive functions. Therefore, understanding of the AD pathogenesis has to include the understanding of this selectivity. Currently, the main efforts aimed at prevention and treatment of AD are based on the dominating hypothesis for the AD pathogenesis: the amyloid hypothesis. But this hypothesis does not explain selective memory impairments since β-amyloid accumulates extracellularly and should be toxic to all types of cerebral neurons, not only to "memory engram neurons." To explain selective memory impairment, I propose the autoimmune hypothesis of AD, based on the analysis of risk factors for AD and molecular mechanisms of memory formation. Memory formation is associated with epigenetic modifications of chromatin in memory engram neurons and, therefore, might be accompanied by the expression of memory-specific proteins recognized by the adaptive immune system as "non-self" antigens. Normally, the brain is protected by the blood-brain barrier (BBB). All risk factors for AD provoke BBB disruptions, possibly leading to an autoimmune reaction against memory engram neurons. This reaction would make them selectively sensitive to tauopathy. If this hypothesis is confirmed, the strategies for AD prevention and treatment would be radically changed.
Collapse
Affiliation(s)
- Yuri I Arshavsky
- BioCircuits Institute, University of California, San Diego, La Jolla, California, United States
| |
Collapse
|
14
|
Gao Y, Zhao P, Wang C, Fang K, Pan Y, Zhang Y, Miao Z, Wang M, Wei M, Zou W, Liu M, Peng K. Buqi Huoxue Tongnao prescription protects against chronic cerebral hypoperfusion via regulating PI3K/AKT and LXRα/CYP7A1 signaling pathways. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155844. [PMID: 38959552 DOI: 10.1016/j.phymed.2024.155844] [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: 02/22/2024] [Revised: 06/01/2024] [Accepted: 06/24/2024] [Indexed: 07/05/2024]
Abstract
BACKGROUND Chronic cerebral hypoperfusion (CCH) has been confirmed as one of the pathogenesis underlying vascular cognitive impairment. A series of pathological changes, including inflammation, oxidative stress, and apoptosis, are involved in this pathophysiology and contribute to cognitive impairment and neuropathological alterations. The traditional Chinese medicine (TCM) of Buqi Huoxue Tongnao (BQHXTN) prescription possesses a remarkable clinical efficacy for treating patients with CCH, but still lacks a scientific foundation for its pharmacological mechanisms. PURPOSE To investigate the role and underlying mechanism of the effects of BQHXTN on CCH both in vitro and in vivo. METHODS In this study, we established a two-vessel occlusion (2-VO) induced CCH model in Sprague-Dawley rats, an oxygen-glucose deprivation model in BV2 cells, and a steatosis cell model in L02 cells to reveal the underlying mechanisms of BQHXTN by behavioral test, histopathological analysis and the detection of pro-inflammatory cytokine, apoptotic factors and reactive oxide species. Donepezil hydrochloride and Buyang Huanwu decoction were used as positive drugs. RESULTS Compared with the 2-VO group, BQHXTN treatment at three doses significantly enhanced the memory and learning abilities in the Y-maze and novel object recognition tests. The hematoxylin-eosin staining indicated that BQHXTN protected against hippocampal injury induced by CCH. Of note, in both in vivo and in vitro experiments, BQHXTN prominently inhibited the production of IL-1β, TNF-α, cleaved-caspase 3, and iNOS by regulating the PI3K/AKT pathway, consequently exerting anti-inflammatory, anti-apoptotic, and antioxidant effects. Moreover, it provided the first initial evidence that BQHXTN treatment mitigated dyslipidemia by increasing the LXRα/CYP7A1 expression, thereby delaying the neuropathological process. CONCLUSION In summary, these findings firstly revealed the pharmacodynamics and mechanism of BQHXTN, that is, BQHXTN could alleviate cognitive impairment, neuropathological alterations and dyslipidemia in CCH rats by activating PI3K/AKT and LXRα/CYP7A1 signaling pathways, as well as providing a TCM treatment strategy for CCH.
Collapse
Affiliation(s)
- Yinhuang Gao
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Peng Kang National Famous Traditional Chinese Medicine Expert Inheritance Studio, Southern Medicine University, Guangzhou 510315, China; Key Laboratory of Drug Metabolism Research and Evaluation of the State Drug Administration, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Peng Zhao
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Peng Kang National Famous Traditional Chinese Medicine Expert Inheritance Studio, Southern Medicine University, Guangzhou 510315, China
| | - Chunyan Wang
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Peng Kang National Famous Traditional Chinese Medicine Expert Inheritance Studio, Southern Medicine University, Guangzhou 510315, China
| | - Keren Fang
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Peng Kang National Famous Traditional Chinese Medicine Expert Inheritance Studio, Southern Medicine University, Guangzhou 510315, China
| | - Yueqing Pan
- Key Laboratory of Drug Metabolism Research and Evaluation of the State Drug Administration, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yan Zhang
- Key Laboratory of Drug Metabolism Research and Evaluation of the State Drug Administration, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhishuo Miao
- Key Laboratory of Drug Metabolism Research and Evaluation of the State Drug Administration, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Meirong Wang
- Key Laboratory of Drug Metabolism Research and Evaluation of the State Drug Administration, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Minlong Wei
- Key Laboratory of Drug Metabolism Research and Evaluation of the State Drug Administration, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Wei Zou
- Changsha Research and Development Center on Obstetric and Gynecologic Traditional Chinese Medicine Preparation, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan 410008, China
| | - Menghua Liu
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Peng Kang National Famous Traditional Chinese Medicine Expert Inheritance Studio, Southern Medicine University, Guangzhou 510315, China; Key Laboratory of Drug Metabolism Research and Evaluation of the State Drug Administration, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China.
| | - Kang Peng
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Peng Kang National Famous Traditional Chinese Medicine Expert Inheritance Studio, Southern Medicine University, Guangzhou 510315, China.
| |
Collapse
|
15
|
Di Domenico F, Lanzillotta C, Perluigi M. Redox imbalance and metabolic defects in the context of Alzheimer disease. FEBS Lett 2024; 598:2047-2066. [PMID: 38472147 DOI: 10.1002/1873-3468.14840] [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: 10/17/2023] [Revised: 02/12/2024] [Accepted: 02/12/2024] [Indexed: 03/14/2024]
Abstract
Redox reactions play a critical role for intracellular processes, including pathways involved in metabolism and signaling. Reactive oxygen species (ROS) act either as second messengers or generators of protein modifications, fundamental mechanisms for signal transduction. Disturbance of redox homeostasis is associated with many disorders. Among these, Alzheimer's disease is a neurodegenerative pathology that presents hallmarks of oxidative damage such as increased ROS production, decreased activity of antioxidant enzymes, oxidative modifications of macromolecules, and changes in mitochondrial homeostasis. Interestingly, alteration of redox homeostasis is closely associated with defects of energy metabolism, involving both carbohydrates and lipids, the major energy fuels for the cell. As the brain relies exclusively on glucose metabolism, defects of glucose utilization represent a harmful event for the brain. During aging, a progressive perturbation of energy metabolism occurs resulting in brain hypometabolism. This condition contributes to increase neuronal cell vulnerability ultimately resulting in cognitive impairment. The current review discusses the crosstalk between alteration of redox homeostasis and brain energy defects that seems to act in concert in promoting Alzheimer's neurodegeneration.
Collapse
Affiliation(s)
- Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Italy
| | - Chiara Lanzillotta
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Italy
| | - Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Italy
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| |
Collapse
|
16
|
Lu Y, Xu K, Maydanchik N, Kang B, Pierce BL, Yang F, Chen LS. An integrative multi-context Mendelian randomization method for identifying risk genes across human tissues. Am J Hum Genet 2024; 111:1736-1749. [PMID: 39053459 PMCID: PMC11339623 DOI: 10.1016/j.ajhg.2024.06.012] [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: 02/03/2024] [Revised: 06/11/2024] [Accepted: 06/24/2024] [Indexed: 07/27/2024] Open
Abstract
Mendelian randomization (MR) provides valuable assessments of the causal effect of exposure on outcome, yet the application of conventional MR methods for mapping risk genes encounters new challenges. One of the issues is the limited availability of expression quantitative trait loci (eQTLs) as instrumental variables (IVs), hampering the estimation of sparse causal effects. Additionally, the often context- or tissue-specific eQTL effects challenge the MR assumption of consistent IV effects across eQTL and GWAS data. To address these challenges, we propose a multi-context multivariable integrative MR framework, mintMR, for mapping expression and molecular traits as joint exposures. It models the effects of molecular exposures across multiple tissues in each gene region, while simultaneously estimating across multiple gene regions. It uses eQTLs with consistent effects across more than one tissue type as IVs, improving IV consistency. A major innovation of mintMR involves employing multi-view learning methods to collectively model latent indicators of disease relevance across multiple tissues, molecular traits, and gene regions. The multi-view learning captures the major patterns of disease relevance and uses these patterns to update the estimated tissue relevance probabilities. The proposed mintMR iterates between performing a multi-tissue MR for each gene region and joint learning the disease-relevant tissue probabilities across gene regions, improving the estimation of sparse effects across genes. We apply mintMR to evaluate the causal effects of gene expression and DNA methylation for 35 complex traits using multi-tissue QTLs as IVs. The proposed mintMR controls genome-wide inflation and offers insights into disease mechanisms.
Collapse
Affiliation(s)
- Yihao Lu
- Department of Public Health Sciences, The University of Chicago, Chicago, IL, USA
| | - Ke Xu
- Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, Notre Dame, IN, USA
| | - Nathaniel Maydanchik
- Department of Public Health Sciences, The University of Chicago, Chicago, IL, USA
| | - Bowei Kang
- Department of Public Health Sciences, The University of Chicago, Chicago, IL, USA
| | - Brandon L Pierce
- Department of Public Health Sciences, The University of Chicago, Chicago, IL, USA
| | - Fan Yang
- Yau Mathematical Sciences Center, Tsinghua University, Beijing, China; Yanqi Lake Beijing Institute of Mathematical Sciences and Applications, Beijing, China.
| | - Lin S Chen
- Department of Public Health Sciences, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
17
|
Schultz C, Wegner T, Heusel C, Gallagher T, Zheng Y, Werner M, Wegner SV, Meyer-Zedler T, Werz O, Schmitt M, Popp J, Glorius F. Alkyne-tagged imidazolium-based membrane cholesterol analogs for Raman imaging applications. Chem Sci 2024:d4sc03155e. [PMID: 39156930 PMCID: PMC11325193 DOI: 10.1039/d4sc03155e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 07/24/2024] [Indexed: 08/20/2024] Open
Abstract
Cholesterol is an important lipid playing a crucial role in mediating essential cellular processes as well as maintaining the basic structural integrity of biological membranes. Given its vast biological importance, there is an unabated need for sophisticated strategies to investigate cholesterol-mediated biological processes. Raman-tagged sterol analogs offer the advantage of being visualizable without the need for a bulky dye that potentially affects natural membrane integration and cellular interactions as it is the case for many conventionally used fluorescent analogs. Herein, we report a series of alkyne-tagged imidazolium-based cholesterol analogs (CHIMs) with large Raman scattering cross-sections that readily integrate into HEK cells and primary monocyte-derived macrophages and allow (multiplexed) cellular Raman imaging. We envision Raman-tagged CHIM analogs to be a powerful platform for the investigation of cholesterol-mediated cellular processes complementary to other established methods, such as the use of fluorescent analogs.
Collapse
Affiliation(s)
- Constanze Schultz
- Leibniz Institute of Photonic Technology (Leibniz-IPHT), Member of Leibniz Health Technologies, Member of the Leibniz Center for Photonics in Infection Research (LPI) Albert-Einstein-Str. 9 07745 Jena Germany
| | - Tristan Wegner
- University of Münster, Institute of Organic Chemistry Corrensstraße 40 48149 Münster Germany
| | - Corinna Heusel
- University of Münster, Institute of Organic Chemistry Corrensstraße 40 48149 Münster Germany
| | - Tim Gallagher
- University of Münster, Institute of Organic Chemistry Corrensstraße 40 48149 Münster Germany
| | - Yanjun Zheng
- University of Münster, Institute of Physiological Chemistry and Pathobiochemistry Waldeyerstraße 15 48149 Münster Germany
| | - Markus Werner
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena Philosophenweg 14 07743 Jena Germany
| | - Seraphine V Wegner
- University of Münster, Institute of Physiological Chemistry and Pathobiochemistry Waldeyerstraße 15 48149 Münster Germany
| | - Tobias Meyer-Zedler
- Leibniz Institute of Photonic Technology (Leibniz-IPHT), Member of Leibniz Health Technologies, Member of the Leibniz Center for Photonics in Infection Research (LPI) Albert-Einstein-Str. 9 07745 Jena Germany
- Institute of Physical Chemistry (IPC) and Abbe Center of Photonics (ACP), Member of the Leibniz Center for Photonics in Infection Research (LPI), Friedrich Schiller University Jena Helmholtzweg 4 07743 Jena Germany
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena Philosophenweg 14 07743 Jena Germany
| | - Michael Schmitt
- Institute of Physical Chemistry (IPC) and Abbe Center of Photonics (ACP), Member of the Leibniz Center for Photonics in Infection Research (LPI), Friedrich Schiller University Jena Helmholtzweg 4 07743 Jena Germany
| | - Juergen Popp
- Leibniz Institute of Photonic Technology (Leibniz-IPHT), Member of Leibniz Health Technologies, Member of the Leibniz Center for Photonics in Infection Research (LPI) Albert-Einstein-Str. 9 07745 Jena Germany
- Institute of Physical Chemistry (IPC) and Abbe Center of Photonics (ACP), Member of the Leibniz Center for Photonics in Infection Research (LPI), Friedrich Schiller University Jena Helmholtzweg 4 07743 Jena Germany
| | - Frank Glorius
- University of Münster, Institute of Organic Chemistry Corrensstraße 40 48149 Münster Germany
| |
Collapse
|
18
|
Mirdha L. Aggregation Behavior of Amyloid Beta Peptide Depends Upon the Membrane Lipid Composition. J Membr Biol 2024; 257:151-164. [PMID: 38888760 DOI: 10.1007/s00232-024-00314-3] [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: 03/07/2024] [Accepted: 05/15/2024] [Indexed: 06/20/2024]
Abstract
Protein aggregation plays a crucial role in the development of several neurodegenerative diseases. It is important to understand the aggregation process for the detection of the onset of these diseases. Alzheimer's Disease (AD) is one of the most prevalent neurodegenerative diseases caused by the aggregation of Aβ-40 and Aβ-42 peptides. The smaller oligomers lead to the formation of protein plaque at the neural membranes leading to memory loss and other disorders. Interestingly, aggregation takes place at the neural membranes, therefore the membrane composition seems to play an important role in the aggregation process. Despite a large number of literatures on the effect of lipid composition on protein aggregation, there are very few concise reviews that highlight the role of membrane composition in protein aggregation. In this review, we have discussed the implication of membrane composition on the aggregation of amyloid beta peptide with a special emphasis on cholesterol. We have further discussed the role of the degree of unsaturation of fatty acids and the participation of apolipoprotein E4 (ApoE4) in the onset of AD.
Collapse
Affiliation(s)
- Lipika Mirdha
- School of Chemistry, Sambalpur University, Jyoti Vihar, Burla, Odisha, 768 019, India.
| |
Collapse
|
19
|
Ha J, Kwon GE, Son Y, Jang SA, Cho SY, Park SJ, Kim H, Lee J, Lee J, Seo D, Lee M, Lee DY, Choi MH, Kim E. Cholesterol profiling reveals 7β-hydroxycholesterol as a pathologically relevant peripheral biomarker of Alzheimer's disease. Psychiatry Clin Neurosci 2024; 78:473-481. [PMID: 38923201 PMCID: PMC11488599 DOI: 10.1111/pcn.13706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 04/22/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024]
Abstract
AIM Cholesterol homeostasis is associated with Alzheimer's disease (AD). Despite the multitude of cholesterol metabolites, little is known about which metabolites are directly involved in AD pathogenesis and can serve as its potential biomarkers. METHODS To identify "hit" metabolites, steroid profiling was conducted in mice with different age, diet, and genotype and also in humans with normal cognition, mild cognitive impairment, and AD using gas chromatography-mass spectrometry. Then, using one of the "hit" molecules (7β-hydroxycholesterol; OHC), molecular and histopathological experiment and behavioral testing were conducted in normal mice following its intracranial stereotaxic injection to see whether this molecule drives AD pathogenesis and causes cognitive impairment. RESULTS The serum levels of several metabolites, including 7β-OHC, were increased by aging in the 3xTg-AD unlike normal mice. Consistently, the levels of 7β-OHC were increased in the hairs of patients with AD and were correlated with clinical severity. We found that 7β-OHC directly affects AD-related pathophysiology; intrahippocampal injection of 7β-OHC induced astrocyte and microglial cell activation, increased the levels of pro-inflammatory cytokines (TNF-alpha, IL-1β, IL-6), and enhanced amyloidogenic pathway. Mice treated with 7β-OHC also exhibited deficits in memory and frontal/executive functions assessed by object recognition and 5-choice serial reaction time task, respectively. CONCLUSIONS Our results suggest that 7β-OHC could serve as a convenient, peripheral biomarker of AD. As directly involved in AD pathogenesis, 7β-OHC assay may help actualize personalized medicine in a way to identify an at-risk subgroup as a candidate population for statin-based AD treatment.
Collapse
Affiliation(s)
- Junghee Ha
- Department of Psychiatry, Laboratory for Alzheimer's Molecular Psychiatry, Institute of Behavioral Science in MedicineYonsei University College of MedicineSeoulRepublic of Korea
| | - Go Eun Kwon
- Molecular Recognition Research CenterKorea Institute of Science and TechnologySeoulRepublic of Korea
| | - Yumi Son
- Department of Psychiatry, Laboratory for Alzheimer's Molecular Psychiatry, Institute of Behavioral Science in MedicineYonsei University College of MedicineSeoulRepublic of Korea
- Graduate School of Medical Science, Brain Korea 21 PLUS Project for Medical ScienceYonsei University College of MedicineSeoulRepublic of Korea
| | - Soo Ah Jang
- Department of Psychiatry, Laboratory for Alzheimer's Molecular Psychiatry, Institute of Behavioral Science in MedicineYonsei University College of MedicineSeoulRepublic of Korea
| | - So Yeon Cho
- Department of Psychiatry, Laboratory for Alzheimer's Molecular Psychiatry, Institute of Behavioral Science in MedicineYonsei University College of MedicineSeoulRepublic of Korea
- Graduate School of Medical Science, Brain Korea 21 PLUS Project for Medical ScienceYonsei University College of MedicineSeoulRepublic of Korea
| | - Soo Jin Park
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, Research Institute for Agricultural and Life SciencesSeoul National UniversitySeoulRepublic of Korea
| | - Hyunjeong Kim
- Department of Psychiatry, Laboratory for Alzheimer's Molecular Psychiatry, Institute of Behavioral Science in MedicineYonsei University College of MedicineSeoulRepublic of Korea
- Metabolism‐Dementia Research InstituteYonsei University College of MedicineSeoulRepublic of Korea
| | - Jimin Lee
- Department of Psychiatry, Laboratory for Alzheimer's Molecular Psychiatry, Institute of Behavioral Science in MedicineYonsei University College of MedicineSeoulRepublic of Korea
| | - Juseok Lee
- Department of MedicineYonsei University College of MedicineSeoulRepublic of Korea
| | - Dongryul Seo
- Department of MedicineYonsei University College of MedicineSeoulRepublic of Korea
| | - Myeongjee Lee
- Biostatistics Collaboration Unit, Department of Biomedical Systems InformaticsYonsei University College of MedicineSeoulKorea
| | - Do Yup Lee
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, Research Institute for Agricultural and Life SciencesSeoul National UniversitySeoulRepublic of Korea
| | - Man Ho Choi
- Molecular Recognition Research CenterKorea Institute of Science and TechnologySeoulRepublic of Korea
| | - Eosu Kim
- Department of Psychiatry, Laboratory for Alzheimer's Molecular Psychiatry, Institute of Behavioral Science in MedicineYonsei University College of MedicineSeoulRepublic of Korea
- Graduate School of Medical Science, Brain Korea 21 PLUS Project for Medical ScienceYonsei University College of MedicineSeoulRepublic of Korea
- Metabolism‐Dementia Research InstituteYonsei University College of MedicineSeoulRepublic of Korea
| |
Collapse
|
20
|
Selmi H, Walker A, Debarbieux L, Schmitt-Kopplin P. Improving the intestinal lipidome coverage in a gnotobiotic mouse model using UHPLC-MS-based approach through optimization of mobile phase modifiers and column selection. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1242:124188. [PMID: 38901159 DOI: 10.1016/j.jchromb.2024.124188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/21/2024] [Accepted: 05/30/2024] [Indexed: 06/22/2024]
Abstract
Lipidomics is focusing on the screening of lipid species in complex mixtures using mass spectrometry-based approaches. In this work, we aim to enhance the intestinal lipidome coverage within the Oligo-Mouse-Microbiota (OMM12) colonized mouse model by testing eight mobile phase conditions on five reversed-phase columns. Our selected mobile phase modifiers included two ammonium salts, two concentrations, and the addition of respective acids at 0.1 %. We compared two columns with hybrid surface technology, two with ethylene bridged hybrid technology and one with core-shell particles. Best performance was attained for standards and intestinal lipidome, using either ammonium formate or acetate in ESI(+) or ammonium acetate in ESI(-) for all column technologies. Notably, a concentration of 5 mM ammonium salt showed optimal results for both modes, while the addition of acids had a negligible effect on lipid ionization efficiency. The HST BEH C18 column improved peak width and tailing factor parameters compared to other technologies. We achieved the highest lipid count in colon and ileum content, including ceramides, phosphatidylethanolamines and phosphatidylcholines, when using 5 mM ammonium acetate in ESI(-). Conversely, in ESI(+) 5 mM ammonium formate demonstrated superior coverage for diacylglycerols and triacylglycerols.
Collapse
Affiliation(s)
- Habiba Selmi
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, Neuherberg, Germany
| | - Alesia Walker
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, Neuherberg, Germany.
| | - Laurent Debarbieux
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Bacteriophage Bacterium Host, Paris, France
| | - Philippe Schmitt-Kopplin
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, Neuherberg, Germany; Chair of Analytical Food Chemistry, Technical University of Munich, Freising, Germany
| |
Collapse
|
21
|
Uchiumi O, Zou J, Yamaki S, Hori Y, Ono M, Yamamoto R, Kato N. Disruption of sphingomyelin synthase 2 gene alleviates cognitive impairment in a mouse model of Alzheimer's disease. Brain Res 2024; 1835:148934. [PMID: 38609029 DOI: 10.1016/j.brainres.2024.148934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/28/2024] [Accepted: 04/09/2024] [Indexed: 04/14/2024]
Abstract
The membrane raft accommodates the key enzymes synthesizing amyloid β (Aβ). One of the two characteristic components of the membrane raft, cholesterol, is well known to promote the key enzymes that produce amyloid-β (Aβ) and exacerbate Alzheimer's disease (AD) pathogenesis. Given that the raft is a physicochemical platform for the sound functioning of embedded bioactive proteins, the other major lipid component sphingomyelin may also be involved in AD. Here we knocked out the sphingomyelin synthase 2 gene (SMS2) in 3xTg AD model mice by hybridization, yielding SMS2KO mice (4S mice). The novel object recognition test in 9/10-month-old 4S mice showed that cognitive impairment in 3xTg mice was alleviated by SMS2KO, though performance in the Morris water maze (MWM) was not improved. The tail suspension test detected a depressive trait in 4S mice, which may have hindered the manifestation of performance in the wet, stressful environment of MWM. In the hippocampal CA1, hyperexcitability in 3xTg was also found alleviated by SMS2KO. In the hippocampal dentate gyrus of 4S mice, the number of neurons positive with intracellular Aβ or its precursor proteins, the hallmark of young 3xTg mice, is reduced to one-third, suggesting an SMS2KO-led suppression of syntheses of those peptides in the dentate gyrus. Although we previously reported that large-conductance calcium-activated potassium (BK) channels are suppressed in 3xTg mice and their recovery relates to cognitive amelioration, no changes occurred by hybridization. Sphingomyelin in the membrane raft may serve as a novel target for AD drugs.
Collapse
Affiliation(s)
- Osamu Uchiumi
- Department of Physiology, Kanazawa Medical University, Ishikawa 920-0293, Japan
| | - Jingyu Zou
- Department of Physiology, Kanazawa Medical University, Ishikawa 920-0293, Japan; First Affiliated Hospital, China Medical University, Shenyang 110001, China
| | - Sachiko Yamaki
- Department of Physiology, Kanazawa Medical University, Ishikawa 920-0293, Japan
| | - Yoshie Hori
- Department of Physiology, Kanazawa Medical University, Ishikawa 920-0293, Japan
| | - Munenori Ono
- Department of Physiology, Kanazawa Medical University, Ishikawa 920-0293, Japan
| | - Ryo Yamamoto
- Department of Physiology, Kanazawa Medical University, Ishikawa 920-0293, Japan
| | - Nobuo Kato
- Department of Physiology, Kanazawa Medical University, Ishikawa 920-0293, Japan.
| |
Collapse
|
22
|
Feringa FM, Hertog SJKD, Wang L, Derks RJE, Kruijff I, Erlebach L, Heijneman J, Miramontes R, Pömpner N, Blomberg N, Olivier-Jimenez D, Johansen LE, Cammack AJ, Giblin A, Toomey CE, Rose IVL, Yuan H, Ward M, Isaacs AM, Kampmann M, Kronenberg-Versteeg D, Lashley T, Thompson LM, Ori A, Mohammed Y, Giera M, van der Kant R. The Neurolipid Atlas: a lipidomics resource for neurodegenerative diseases uncovers cholesterol as a regulator of astrocyte reactivity impaired by ApoE4. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.01.601474. [PMID: 39005258 PMCID: PMC11244892 DOI: 10.1101/2024.07.01.601474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Lipid changes in the brain have been implicated in many neurodegenerative diseases including Alzheimer's Disease (AD), Parkinson's disease and Amyotrophic Lateral Sclerosis. To facilitate comparative lipidomic research across brain-diseases we established a data commons named the Neurolipid Atlas, that we have pre-populated with novel human, mouse and isogenic induced pluripotent stem cell (iPSC)-derived lipidomics data for different brain diseases. We show that iPSC-derived neurons, microglia and astrocytes display distinct lipid profiles that recapitulate in vivo lipotypes. Leveraging multiple datasets, we show that the AD risk gene ApoE4 drives cholesterol ester (CE) accumulation in human astrocytes recapitulating CE accumulation measured in the human AD brain. Multi-omic interrogation of iPSC-derived astrocytes revealed that cholesterol plays a major role in astrocyte interferon-dependent pathways such as the immunoproteasome and major histocompatibility complex (MHC) class I antigen presentation. We show that through enhanced cholesterol esterification ApoE4 suppresses immune activation of astrocytes. Our novel data commons, available at neurolipidatlas.com, provides a user-friendly tool and knowledge base for a better understanding of lipid dyshomeostasis in neurodegenerative diseases.
Collapse
Affiliation(s)
- Femke M Feringa
- Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Sascha J Koppes-den Hertog
- Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, the Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Lian Wang
- Leiden University Medical Center, Center for Proteomics and Metabolomics, Leiden, the Netherlands
| | - Rico J E Derks
- Leiden University Medical Center, Center for Proteomics and Metabolomics, Leiden, the Netherlands
| | - Iris Kruijff
- Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, the Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Lena Erlebach
- German Center for Neurodegenerative Diseases (DZNE) Tübingen, Tübingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Jorin Heijneman
- Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, the Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Ricardo Miramontes
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Nadine Pömpner
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany
| | - Niek Blomberg
- Leiden University Medical Center, Center for Proteomics and Metabolomics, Leiden, the Netherlands
| | - Damien Olivier-Jimenez
- Leiden University Medical Center, Center for Proteomics and Metabolomics, Leiden, the Netherlands
| | - Lill Eva Johansen
- Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Alexander J Cammack
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute at UCL, University College London, London, UK
| | - Ashling Giblin
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute at UCL, University College London, London, UK
| | - Christina E Toomey
- Department of Clinical and Molecular Neuroscience, Queen Square Institute of Neurology, University College London, London, UK
| | - Indigo V L Rose
- Institute for Neurodegenerative Diseases and Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Hebao Yuan
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Michael Ward
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Adrian M Isaacs
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute at UCL, University College London, London, UK
| | - Martin Kampmann
- Department of Biochemistry and Biophysics, Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
| | - Deborah Kronenberg-Versteeg
- German Center for Neurodegenerative Diseases (DZNE) Tübingen, Tübingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Tammaryn Lashley
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Leslie M Thompson
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Alessandro Ori
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany
| | - Yassene Mohammed
- Leiden University Medical Center, Center for Proteomics and Metabolomics, Leiden, the Netherlands
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC H3A 0G4, Canada
| | - Martin Giera
- Leiden University Medical Center, Center for Proteomics and Metabolomics, Leiden, the Netherlands
| | - Rik van der Kant
- Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, the Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, the Netherlands
| |
Collapse
|
23
|
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
|
24
|
Wang W, Huang J, Qian S, Zheng Y, Yu X, Jiang T, Ai R, Hou J, Ma E, Cai J, He H, Wang X, Xie C. Amyloid-β but not tau accumulation is strongly associated with longitudinal cognitive decline. CNS Neurosci Ther 2024; 30:e14860. [PMID: 39014268 PMCID: PMC11251873 DOI: 10.1111/cns.14860] [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: 12/13/2023] [Revised: 06/11/2024] [Accepted: 07/03/2024] [Indexed: 07/18/2024] Open
Abstract
OBJECTIVE Alzheimer's disease (AD) pathology is featured by the extracellular accumulation of amyloid-β (Aβ) plaques and intracellular tau neurofibrillary tangles in the brain. We studied whether Aβ and tau accumulation are independently associated with future cognitive decline in the AD continuum. METHODS Data were acquired from the Alzheimer's Disease Neuroimaging Initiative (ADNI) public database. A total of 1272 participants were selected based on the availability of Aβ-PET and CSF tau at baseline and of those 777 participants with follow-up visits. RESULTS We found that Aβ-PET and CSF tau pathology were related to cognitive decline across the AD clinical spectrum, both as potential predictors for dementia progression. Among them, Aβ-PET (A + T- subjects) is an independent reliable predictor of longitudinal cognitive decline in terms of ADAS-13, ADNI-MEM, and MMSE scores rather than tau pathology (A - T+ subjects), indicating tau accumulation is not closely correlated with future cognitive impairment without being driven by Aβ deposition. Of note, a high percentage of APOE ε4 carriers with Aβ pathology (A+) develop poor memory and learning capacity. Interestingly, this condition is not recurrence in terms of the ADNI-MEM domain when adding APOE ε4 status. Finally, the levels of Aβ-PET SUVR related to glucose hypometabolism more strongly in subjects with A + T- than A - T+ both happen at baseline and longitudinal changes. CONCLUSIONS In conclusion, Aβ-PET alone without tau pathology (A + T-) measure is an independent reliable predictor of longitudinal cognitive decline but may nonetheless forecast different status of dementia progression. However, tau accumulation alone without Aβ pathology background (A - T+) was not enough to be an independent predictor of cognitive worsening.
Collapse
Affiliation(s)
- Wenwen Wang
- The Center of Traditional Chinese Medicine, The Second Affiliated HospitalYuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Jiani Huang
- Department of NeurologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Shuangjie Qian
- Department of NeurologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Yi Zheng
- Department of NeurologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Xinyue Yu
- Alberta InstituteWenzhou Medical UniversityWenzhouZhejiangChina
| | - Tao Jiang
- Department of NeurologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Ruixue Ai
- Department of Clinical Molecular Biology, Akershus University HospitalUniversity of OsloLørenskogNorway
| | - Jialong Hou
- Department of NeurologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Enzi Ma
- Department of NeurologyTraditional Chinese and Western Medicine Hospital of WenzhouWenzhouZhejiangChina
| | - Jinlai Cai
- Department of NeurologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Haijun He
- Department of NeurologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - XinShi Wang
- Department of NeurologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Chenglong Xie
- Department of NeurologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
- Oujiang LaboratoryWenzhouZhejiangChina
- Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of AgingWenzhou Medical UniversityWenzhouZhejiangChina
- Department of Geriatrics, Geriatric Medical CenterThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| |
Collapse
|
25
|
Singh MK, Shin Y, Ju S, Han S, Kim SS, Kang I. Comprehensive Overview of Alzheimer's Disease: Etiological Insights and Degradation Strategies. Int J Mol Sci 2024; 25:6901. [PMID: 39000011 PMCID: PMC11241648 DOI: 10.3390/ijms25136901] [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: 05/21/2024] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 07/14/2024] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder and affects millions of individuals globally. AD is associated with cognitive decline and memory loss that worsens with aging. A statistical report using U.S. data on AD estimates that approximately 6.9 million individuals suffer from AD, a number projected to surge to 13.8 million by 2060. Thus, there is a critical imperative to pinpoint and address AD and its hallmark tau protein aggregation early to prevent and manage its debilitating effects. Amyloid-β and tau proteins are primarily associated with the formation of plaques and neurofibril tangles in the brain. Current research efforts focus on degrading amyloid-β and tau or inhibiting their synthesis, particularly targeting APP processing and tau hyperphosphorylation, aiming to develop effective clinical interventions. However, navigating this intricate landscape requires ongoing studies and clinical trials to develop treatments that truly make a difference. Genome-wide association studies (GWASs) across various cohorts identified 40 loci and over 300 genes associated with AD. Despite this wealth of genetic data, much remains to be understood about the functions of these genes and their role in the disease process, prompting continued investigation. By delving deeper into these genetic associations, novel targets such as kinases, proteases, cytokines, and degradation pathways, offer new directions for drug discovery and therapeutic intervention in AD. This review delves into the intricate biological pathways disrupted in AD and identifies how genetic variations within these pathways could serve as potential targets for drug discovery and treatment strategies. Through a comprehensive understanding of the molecular underpinnings of AD, researchers aim to pave the way for more effective therapies that can alleviate the burden of this devastating disease.
Collapse
Affiliation(s)
- Manish Kumar Singh
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yoonhwa Shin
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Songhyun Ju
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sunhee Han
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sung Soo Kim
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Insug Kang
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
26
|
Mast N, Butts M, Pikuleva IA. Unbiased insights into the multiplicity of the CYP46A1 brain effects in 5XFAD mice treated with low dose-efavirenz. J Lipid Res 2024; 65:100555. [PMID: 38719151 PMCID: PMC11176809 DOI: 10.1016/j.jlr.2024.100555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/12/2024] [Accepted: 05/01/2024] [Indexed: 05/30/2024] Open
Abstract
Cytochrome P450 46A1 (CYP46A1) is the CNS-specific cholesterol 24-hydroxylase that controls cholesterol elimination and turnover in the brain. In mouse models, pharmacologic CYP46A1 activation with low-dose efavirenz or by gene therapy mitigates the manifestations of various brain disorders, neurologic, and nonneurologic, by affecting numerous, apparently unlinked biological processes. Accordingly, CYP46A1 is emerging as a promising therapeutic target; however, the mechanisms underlying the multiplicity of the brain CYP46A1 activity effects are currently not understood. We proposed the chain reaction hypothesis, according to which CYP46A1 is important for the three primary (unifying) processes in the brain (sterol flux through the plasma membranes, acetyl-CoA, and isoprenoid production), which in turn affect a variety of secondary processes. We already identified several processes secondary to changes in sterol flux and herein undertook a multiomics approach to compare the brain proteome, acetylproteome, and metabolome of 5XFAD mice (an Alzheimer's disease model), control and treated with low-dose efavirenz. We found that the latter had increased production of phospholipids from the corresponding lysophospholipids and a globally increased protein acetylation (including histone acetylation). Apparently, these effects were secondary to increased acetyl-CoA production. Signaling of small GTPases due to their altered abundance or abundance of their regulators could be affected as well, potentially via isoprenoid biosynthesis. In addition, the omics data related differentially abundant molecules to other biological processes either reported previously or new. Thus, we obtained unbiased mechanistic insights and identified potential players mediating the multiplicity of the CYP46A1 brain effects and further detailed our chain reaction hypothesis.
Collapse
Affiliation(s)
- Natalia Mast
- Department of Ophthalmology and Visual Science, Case Western Reserve University, Cleveland, OH, USA
| | - Makaya Butts
- Department of Ophthalmology and Visual Science, Case Western Reserve University, Cleveland, OH, USA
| | - Irina A Pikuleva
- Department of Ophthalmology and Visual Science, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
27
|
Ahmed H, Wang Y, Griffiths WJ, Levey AI, Pikuleva I, Liang SH, Haider A. Brain cholesterol and Alzheimer's disease: challenges and opportunities in probe and drug development. Brain 2024; 147:1622-1635. [PMID: 38301270 PMCID: PMC11068113 DOI: 10.1093/brain/awae028] [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: 03/04/2023] [Revised: 12/20/2023] [Accepted: 01/13/2024] [Indexed: 02/03/2024] Open
Abstract
Cholesterol homeostasis is impaired in Alzheimer's disease; however, attempts to modulate brain cholesterol biology have not translated into tangible clinical benefits for patients to date. Several recent milestone developments have substantially improved our understanding of how excess neuronal cholesterol contributes to the pathophysiology of Alzheimer's disease. Indeed, neuronal cholesterol was linked to the formation of amyloid-β and neurofibrillary tangles through molecular pathways that were recently delineated in mechanistic studies. Furthermore, remarkable advances in translational molecular imaging have now made it possible to probe cholesterol metabolism in the living human brain with PET, which is an important prerequisite for future clinical trials that target the brain cholesterol machinery in Alzheimer's disease patients-with the ultimate aim being to develop disease-modifying treatments. This work summarizes current concepts of how the biosynthesis, transport and clearance of brain cholesterol are affected in Alzheimer's disease. Further, current strategies to reverse these alterations by pharmacotherapy are critically discussed in the wake of emerging translational research tools that support the assessment of brain cholesterol biology not only in animal models but also in patients with Alzheimer's disease.
Collapse
Affiliation(s)
- Hazem Ahmed
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, Yale University, New Haven, CT 06510, USA
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, 8093 Zurich, Switzerland
| | - Yuqin Wang
- Institute of Life Science, Swansea University Medical School, Swansea SA2 8PP, UK
| | - William J Griffiths
- Institute of Life Science, Swansea University Medical School, Swansea SA2 8PP, UK
| | - Allan I Levey
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Irina Pikuleva
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Steven H Liang
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ahmed Haider
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| |
Collapse
|
28
|
Xu Y, Zhang J, Li X. Erjingwan and Alzheimer's disease: research based on network pharmacology and experimental confirmation. Front Pharmacol 2024; 15:1328334. [PMID: 38741585 PMCID: PMC11089143 DOI: 10.3389/fphar.2024.1328334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 04/15/2024] [Indexed: 05/16/2024] Open
Abstract
Background Alzheimer's disease (AD), a challenging neurodegenerative condition, has emerged as a significant global public health concern. The Chinese medicine decoction Erjingwan (EJW) has shown promising efficacy in AD treatment, though its mechanism remains unclear. Objective This study aims to elucidate the mechanism by which EJW treats AD through network pharmacology analysis and in vivo experiments. Methods We identified EJW's components using the Traditional Chinese Medicine Systems Pharmacology (TCMSP) database and determined AD-related targets from various databases. A network comprising herbs-compounds-targets was established, and EJW's core targets were ascertained through protein-protein interaction (PPI) analysis. This study assessed the cognitive abilities of APP/PS1 mice using Morris water mazes and Y mazes, in addition to analyzing blood samples for triglyceride (TG), total cholesterol (TC), low-density lipoprotein (LDL), and high-density lipoprotein (HDL) levels. Brain tissues were examined histologically with HE staining, Nissl staining, and immunohistochemistry (IHC) for amyloid β-protein (Aβ) detection. Superoxide dismutase (SOD), reactive oxygen species (ROS), Interleukin-1β (IL-1β), and Interleukin-6 (IL-6) levels in the hippocampal region were measured by ELISA. mRNA expression of apolipoprotein A-I (APOA-I), apolipoprotein B (APOB), apolipoprotein E4 (APOE4), advanced glycation end products (AGE), the receptor for AGE (RAGE), and nuclear factor kappa-B (NF-κB) was evaluated by quantitative PCR (q-PCR). Western blotting was used to detect the expression of AGE, RAGE, NF-κB, and Tau protein. Results Screening identified 57 chemical components and 222 potential targets of EJW. Ten core targets for AD treatment were identified, with enrichment analysis suggesting EJW's effects are related to lipid metabolism and AGEs/RAGE pathways. EJW enhanced learning and memory in APP/PS1 mice, protected neuronal structure in the hippocampal region, reduced Aβ deposition, and altered levels of TG, TC, LDL, IL-1β, and IL-6, and the expression of APOE4, AGEs, RAGE, NF-κB, and Tau protein, while increasing SOD, APOA-I, and APOB mRNA expression. Conclusion The study identified four core components of EJW-iosgenin, baicalein, beta-sitosterol, quercetin-and ten core targets including AKT1, IL6, VEGFA, TP53, CASP3, for treating AD. Experimental results demonstrate EJW's capacity to modulate lipid profiles, reduce pathological markers such as Aβ1-42, Tau, IL-6, IL-1β, reactive oxygen species, SOD, and enhance cognitive functions in APP/PS1 mice, potentially through inhibiting the AGEs/RAGE/NF-κB pathway.
Collapse
Affiliation(s)
- Yuya Xu
- Department of Neurology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | - Jian Zhang
- School of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Xuling Li
- Department of Neurology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
29
|
Luo YX, Yang LL, Yao XQ. Gut microbiota-host lipid crosstalk in Alzheimer's disease: implications for disease progression and therapeutics. Mol Neurodegener 2024; 19:35. [PMID: 38627829 PMCID: PMC11020986 DOI: 10.1186/s13024-024-00720-0] [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] [Accepted: 03/18/2024] [Indexed: 04/19/2024] Open
Abstract
Trillions of intestinal bacteria in the human body undergo dynamic transformations in response to physiological and pathological changes. Alterations in their composition and metabolites collectively contribute to the progression of Alzheimer's disease. The role of gut microbiota in Alzheimer's disease is diverse and complex, evidence suggests lipid metabolism may be one of the potential pathways. However, the mechanisms that gut microbiota mediate lipid metabolism in Alzheimer's disease pathology remain unclear, necessitating further investigation for clarification. This review highlights the current understanding of how gut microbiota disrupts lipid metabolism and discusses the implications of these discoveries in guiding strategies for the prevention or treatment of Alzheimer's disease based on existing data.
Collapse
Affiliation(s)
- Ya-Xi Luo
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ling-Ling Yang
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiu-Qing Yao
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
- Chongqing Municipality Clinical Research Center for Geriatric Medicine, Chongqing, China.
- Department of Rehabilitation Therapy, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
30
|
Oka T, Matsuzawa Y, Tsuneyoshi M, Nakamura Y, Aoshima K, Tsugawa H. Multiomics analysis to explore blood metabolite biomarkers in an Alzheimer's Disease Neuroimaging Initiative cohort. Sci Rep 2024; 14:6797. [PMID: 38565541 PMCID: PMC10987653 DOI: 10.1038/s41598-024-56837-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 03/12/2024] [Indexed: 04/04/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that commonly causes dementia. Identifying biomarkers for the early detection of AD is an emerging need, as brain dysfunction begins two decades before the onset of clinical symptoms. To this end, we reanalyzed untargeted metabolomic mass spectrometry data from 905 patients enrolled in the AD Neuroimaging Initiative (ADNI) cohort using MS-DIAL, with 1,304,633 spectra of 39,108 unique biomolecules. Metabolic profiles of 93 hydrophilic metabolites were determined. Additionally, we integrated targeted lipidomic data (4873 samples from 1524 patients) to explore candidate biomarkers for predicting progressive mild cognitive impairment (pMCI) in patients diagnosed with AD within two years using the baseline metabolome. Patients with lower ergothioneine levels had a 12% higher rate of AD progression with the significance of P = 0.012 (Wald test). Furthermore, an increase in ganglioside (GM3) and decrease in plasmalogen lipids, many of which are associated with apolipoprotein E polymorphism, were confirmed in AD patients, and the higher levels of lysophosphatidylcholine (18:1) and GM3 d18:1/20:0 showed 19% and 17% higher rates of AD progression, respectively (Wald test: P = 3.9 × 10-8 and 4.3 × 10-7). Palmitoleamide, oleamide, diacylglycerols, and ether lipids were also identified as significantly altered metabolites at baseline in patients with pMCI. The integrated analysis of metabolites and genomics data showed that combining information on metabolites and genotypes enhances the predictive performance of AD progression, suggesting that metabolomics is essential to complement genomic data. In conclusion, the reanalysis of multiomics data provides new insights to detect early development of AD pathology and to partially understand metabolic changes in age-related onset of AD.
Collapse
Affiliation(s)
- Takaki Oka
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Yuki Matsuzawa
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Momoka Tsuneyoshi
- Human Biology Integration Foundation, Eisai Co., Ltd., Ibaraki, Japan
| | | | - Ken Aoshima
- Microbes & Host Defense Domain, Eisai Co., Ltd., Ibaraki, Japan
- School of Integrative and Global Majors, University of Tsukuba, Ibaraki, Japan
| | - Hiroshi Tsugawa
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, Tokyo, Japan.
- RIKEN Center for Sustainable Resource Science, Yokohama, Japan.
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan.
| |
Collapse
|
31
|
Toral-Rios D, Long JM, Ulrich JD, Yu J, Strickland MR, Han X, Holtzman DM, Cashikar AG, Paul SM. Cholesterol 25-hydroxylase mediates neuroinflammation and neurodegeneration in a mouse model of tauopathy. J Exp Med 2024; 221:e20232000. [PMID: 38442267 PMCID: PMC10908359 DOI: 10.1084/jem.20232000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/03/2024] [Accepted: 02/01/2024] [Indexed: 03/07/2024] Open
Abstract
Alzheimer's disease (AD) is characterized by amyloid plaques and neurofibrillary tangles, in addition to neuroinflammation and changes in brain lipid metabolism. 25-Hydroxycholesterol (25-HC), a known modulator of both inflammation and lipid metabolism, is produced by cholesterol 25-hydroxylase encoded by Ch25h expressed as a "disease-associated microglia" signature gene. However, whether Ch25h influences tau-mediated neuroinflammation and neurodegeneration is unknown. Here, we show that in the absence of Ch25h and the resultant reduction in 25-HC, there is strikingly reduced age-dependent neurodegeneration and neuroinflammation in the hippocampus and entorhinal/piriform cortex of PS19 mice, which express the P301S mutant human tau transgene. Transcriptomic analyses of bulk hippocampal tissue and single nuclei revealed that Ch25h deficiency in PS19 mice strongly suppressed proinflammatory signaling in microglia. Our results suggest a key role for Ch25h/25-HC in potentiating proinflammatory signaling to promote tau-mediated neurodegeneration. Ch25h may represent a novel therapeutic target for primary tauopathies, AD, and other neuroinflammatory diseases.
Collapse
Affiliation(s)
- Danira Toral-Rios
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
| | - Justin M. Long
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO, USA
| | - Jason D. Ulrich
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, MO, USA
| | - Jinsheng Yu
- Department of Genetics, Genome Technology Access Center at the McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
| | - Michael R. Strickland
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - Xianlin Han
- Department of Medicine, Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - David M. Holtzman
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO, USA
| | - Anil G. Cashikar
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, MO, USA
- Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St Louis, MO, USA
| | - Steven M. Paul
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, MO, USA
- Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
32
|
Zhou J, Ji J, Li X, Zhang Y, Gu L, Zheng X, Li Y, He J, Yang C, Xiao K, Gong Q, Gu Z, Luo K. Homomultivalent Polymeric Nanotraps Disturb Lipid Metabolism Homeostasis and Tune Pyroptosis in Cancer. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2312528. [PMID: 38240412 DOI: 10.1002/adma.202312528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/16/2024] [Indexed: 01/30/2024]
Abstract
Genetic manipulations and pharmaceutical interventions to disturb lipid metabolism homeostasis have emerged as an attractive approach for the management of cancer. However, the research on the utilization of bioactive materials to modulate lipid metabolism homeostasis remains constrained. In this study, heptakis (2,3,6-tri-O-methyl)-β-cyclodextrin (TMCD) is utilized to fabricate homomultivalent polymeric nanotraps, and surprisingly, its unprecedented ability to perturb lipid metabolism homeostasis and induce pyroptosis in tumor cells is found. Through modulation of the density of TMCD arrayed on the polymers, one top-performing nanotrap, PTMCD4, exhibits the most powerful cholesterol-trapping and depletion capacity, thus achieving prominent cytotoxicity toward different types of tumor cells and encouraging antitumor effects in vivo. The interactions between PTMCD4 and biomembranes of tumor cells effectively enable the reduction of cellular phosphatidylcholine and cholesterol levels, thus provoking damage to the biomembrane integrity and perturbation of lipid metabolism homeostasis. Additionally, the interplays between PTMCD4 and lysosomes also induce lysosomal stress, activate the nucleotide-binding oligomerization domain-like receptor protein 3 inflammasomes, and subsequently trigger tumor cell pyroptosis. To sum up, this study first introduces dendronized bioactive polymers to manipulate lipid metabolism and has shed light on another innovative insight for cancer therapy.
Collapse
Affiliation(s)
- Jie Zhou
- Department of Radiology, and Department of Pharmacy, Huaxi MR Research Center (HMRRC), Laboratory of Stem Cell Biology, and Laboratory of Precision Cancer Therapeutics, Precision Medicine Research Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiecheng Ji
- Department of Radiology, and Department of Pharmacy, Huaxi MR Research Center (HMRRC), Laboratory of Stem Cell Biology, and Laboratory of Precision Cancer Therapeutics, Precision Medicine Research Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xue Li
- Department of Radiology, and Department of Pharmacy, Huaxi MR Research Center (HMRRC), Laboratory of Stem Cell Biology, and Laboratory of Precision Cancer Therapeutics, Precision Medicine Research Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuxin Zhang
- Department of Radiology, and Department of Pharmacy, Huaxi MR Research Center (HMRRC), Laboratory of Stem Cell Biology, and Laboratory of Precision Cancer Therapeutics, Precision Medicine Research Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lei Gu
- Department of Radiology, and Department of Pharmacy, Huaxi MR Research Center (HMRRC), Laboratory of Stem Cell Biology, and Laboratory of Precision Cancer Therapeutics, Precision Medicine Research Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiuli Zheng
- Department of Radiology, and Department of Pharmacy, Huaxi MR Research Center (HMRRC), Laboratory of Stem Cell Biology, and Laboratory of Precision Cancer Therapeutics, Precision Medicine Research Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yunkun Li
- Department of Radiology, and Department of Pharmacy, Huaxi MR Research Center (HMRRC), Laboratory of Stem Cell Biology, and Laboratory of Precision Cancer Therapeutics, Precision Medicine Research Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jinhan He
- Department of Radiology, and Department of Pharmacy, Huaxi MR Research Center (HMRRC), Laboratory of Stem Cell Biology, and Laboratory of Precision Cancer Therapeutics, Precision Medicine Research Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Cheng Yang
- Key Laboratory of Green Chemistry & Technology, College of Chemistry, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610064, China
| | - Kai Xiao
- Department of Radiology, and Department of Pharmacy, Huaxi MR Research Center (HMRRC), Laboratory of Stem Cell Biology, and Laboratory of Precision Cancer Therapeutics, Precision Medicine Research Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qiyong Gong
- Department of Radiology, and Department of Pharmacy, Huaxi MR Research Center (HMRRC), Laboratory of Stem Cell Biology, and Laboratory of Precision Cancer Therapeutics, Precision Medicine Research Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
- Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen, 361000, China
| | - Zhongwei Gu
- Department of Radiology, and Department of Pharmacy, Huaxi MR Research Center (HMRRC), Laboratory of Stem Cell Biology, and Laboratory of Precision Cancer Therapeutics, Precision Medicine Research Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Kui Luo
- Department of Radiology, and Department of Pharmacy, Huaxi MR Research Center (HMRRC), Laboratory of Stem Cell Biology, and Laboratory of Precision Cancer Therapeutics, Precision Medicine Research Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| |
Collapse
|
33
|
Tang X, Walter E, Wohleb E, Fan Y, Wang C. ATG5 (autophagy related 5) in microglia controls hippocampal neurogenesis in Alzheimer disease. Autophagy 2024; 20:847-862. [PMID: 37915255 PMCID: PMC11062374 DOI: 10.1080/15548627.2023.2277634] [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: 08/28/2023] [Revised: 10/19/2023] [Accepted: 10/26/2023] [Indexed: 11/03/2023] Open
Abstract
Macroautophagy/autophagy is the intracellular degradation process of cytoplasmic content and damaged organelles. Autophagy is strongly associated with the progression of Alzheimer disease (AD). Microglia are brain-resident macrophages, and recent studies indicate that autophagy in microglia protects neurons from neurodegeneration. Postnatal neurogenesis, the generation of new neurons from adult neural stem cells (NSCs), is impaired in AD patients as well as in AD animal models. However, the extent to which microglial autophagy influences adult NSCs and neurogenesis in AD animal models has not been studied. Here, we showed that conditional knock out (cKO) of Atg5 (autophagy related 5) in microglia inhibited postnatal neurogenesis in the dentate gyrus (DG) of the hippocampus, but not in the subventricular zone (SVZ) of a 5×FAD mouse model. Interestingly, the protection of neurogenesis by Atg5 in microglia was only observed in female AD mice. To confirm the roles of autophagy in microglia for postnatal hippocampal neurogenesis, we generated additional cKO mice to delete autophagy essential genes Rb1cc1 or Atg14 in microglia. However, these rb1cc1 cKO and atg14 cKO mice did not exhibit neurogenesis defects in the context of a female AD mouse model. Last, we used the CSF1R antagonist to deplete ATG5-deficient microglia and this intervention restored neurogenesis in the hippocampus of 5×FAD mice. These results indicate that microglial ATG5 is essential to maintain postnatal hippocampal neurogenesis in a mouse model of AD. Our findings further support the notion that ATG5 in microglia supports NSC health and may prevent neurodegeneration.Abbreviations: 5×FAD: familial Alzheimer disease; Aβ: β-amyloid; AD: Alzheimer disease; AIF1: allograft inflammatory factor 1; ATG: autophagy related; BrdU: 5-bromo-2'-deoxyuridine; CA: Cornu Ammonis; cKO: conditional knock out; CSF1R: colony stimulating factor 1 receptor; Ctrl: control; DCX: doublecortin; DG: dentate gyrus; GFAP: glial fibrillary acidic protein; GZ: granular zone; H&E: hematoxylin and eosin; IF: immunofluorescence; LD: lipid droplet; LDAM: lipid droplets accumulated microglia; LPS: lipopolysaccharides; MAP1LC3B/LC3: microtubule-associated protein 1 light chain 3 beta; NSCs: neural stem cells; RB1CC1: RB1-inducible coiled-coil 1; SOX2: SRY (sex determining region Y)-box 2; SGZ: subgranular zone; SVZ: subventricular zone; WT: wild type.
Collapse
Affiliation(s)
- Xin Tang
- Department of Cancer Biology, University of Cincinnati College Medicine, Cincinnati, OH, USA
| | - Ellen Walter
- Department of Cancer Biology, University of Cincinnati College Medicine, Cincinnati, OH, USA
| | - Eric Wohleb
- Department of Pharmacology & Systems Physiology, University of Cincinnati College Medicine, Cincinnati, OH, USA
| | - Yanbo Fan
- Department of Cancer Biology, University of Cincinnati College Medicine, Cincinnati, OH, USA
| | - Chenran Wang
- Department of Cancer Biology, University of Cincinnati College Medicine, Cincinnati, OH, USA
| |
Collapse
|
34
|
Pang KS, Peng HB, Li BP, Wen B, Noh K, Xia R, Toscan A, Serson S, Fraser PE, Tirona RG, de Lannoy IAM. Aging and brain free cholesterol concentration on amyloid-β peptide accumulation in guinea pigs. Biopharm Drug Dispos 2024; 45:93-106. [PMID: 38488691 DOI: 10.1002/bdd.2386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 01/21/2024] [Accepted: 02/08/2024] [Indexed: 04/19/2024]
Abstract
Alzheimer's disease is a complex multifactorial neurodegenerative disorder wherein age is a major risk factor. The appropriateness of the Hartley guinea pig (GP), which displays high sequence homologies of its amyloid-β (Aβ40 and Aβ42) peptides, Mdr1 and APP (amyloid precursor protein) and similarity in lipid handling to humans, was appraised among 9-40 weeks old guinea pigs. Protein expression levels of P-gp (Abcb1) and Cyp46a1 (24(S)-hydroxylase) for Aβ40, and Aβ42 efflux and cholesterol metabolism, respectively, were decreased with age, whereas those for Lrp1 (low-density lipoprotein receptor related protein 1), Rage (receptor for advanced glycation endproducts) for Aβ efflux and influx, respectively, and Abca1 (the ATP binding cassette subfamily A member 1) for cholesterol efflux, were unchanged among the ages examined. There was a strong, negative correlation of the brain Aβ peptide concentrations and Abca1 protein expression levels with free cholesterol. The correlation of Aβ peptide concentrations with Cyp46a1 was, however, not significant, and concentrations of the 24(S)-hydroxycholesterol metabolite revealed a decreasing trend from 20 weeks old toward 40 weeks old guinea pigs. The composite data suggest a role for free cholesterol on brain Aβ accumulation. The decreases in P-gp and Lrp1 protein levels should further exacerbate the accumulation of Aβ peptides in guinea pig brain.
Collapse
Affiliation(s)
- K Sandy Pang
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - H Benson Peng
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Betty P Li
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Binyu Wen
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Keumhan Noh
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Runyu Xia
- Department of Pharmacology and Toxicology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Anja Toscan
- Transpharmation Canada, Mississauga, Ontario, Canada
| | - Sylvia Serson
- Transpharmation Canada, Mississauga, Ontario, Canada
| | - Paul E Fraser
- Tanz Centre for Research in Neurodegenerative Diseases and Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Rommel G Tirona
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | | |
Collapse
|
35
|
Garcia CS, da Rocha MJ, Presa MH, Pires CS, Besckow EM, Penteado F, Gomes CS, Lenardão EJ, Bortolatto CF, Brüning CA. Exploring the antioxidant potential of chalcogen-indolizines throughout in vitro assays. PeerJ 2024; 12:e17074. [PMID: 38500528 PMCID: PMC10946399 DOI: 10.7717/peerj.17074] [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: 09/26/2023] [Accepted: 02/18/2024] [Indexed: 03/20/2024] Open
Abstract
Reactive oxygen species (ROS) and reactive nitrogen species (RNS) are highly reactive molecules produced naturally by the body and by external factors. When these species are generated in excessive amounts, they can lead to oxidative stress, which in turn can cause cellular and tissue damage. This damage is known to contribute to the aging process and is associated with age-related conditions, including cardiovascular and neurodegenerative diseases. In recent years, there has been an increased interest in the development of compounds with antioxidant potential to assist in the treatment of disorders related to oxidative stress. In this way, compounds containing sulfur (S) and/or selenium (Se) have been considered promising due to the relevant role of these elements in the biosynthesis of antioxidant enzymes and essential proteins with physiological functions. In this context, studies involving heterocyclic nuclei have significantly increased, notably highlighting the indolizine nucleus, given that compounds containing this nucleus have been demonstrating considerable pharmacological properties. Thus, the objective of this research was to evaluate the in vitro antioxidant activity of eight S- and Se-derivatives containing indolizine nucleus and different substituents. The in vitro assays 1,1-diphenyl-2-picryl-hydrazil (DPPH) scavenger activity, ferric ion (Fe3+) reducing antioxidant power (FRAP), thiobarbituric acid reactive species (TBARS), and protein carbonylation (PC) were used to access the antioxidant profile of the compounds. Our findings demonstrated that all the compounds showed FRAP activity and reduced the levels of TBARS and PC in mouse brains homogenates. Some compounds were also capable of acting as DPPH scavengers. In conclusion, the present study demonstrated that eight novel organochalcogen compounds exhibit antioxidant activity.
Collapse
Affiliation(s)
- Cleisson Schossler Garcia
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Chemical, Pharmaceutical and Food Sciences Center (CCQFA), Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Marcia Juciele da Rocha
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Chemical, Pharmaceutical and Food Sciences Center (CCQFA), Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Marcelo Heinemann Presa
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Chemical, Pharmaceutical and Food Sciences Center (CCQFA), Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Camila Simões Pires
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Chemical, Pharmaceutical and Food Sciences Center (CCQFA), Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Evelyn Mianes Besckow
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Chemical, Pharmaceutical and Food Sciences Center (CCQFA), Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Filipe Penteado
- Department of Chemistry, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Caroline Signorini Gomes
- Laboratory of Clean Organic Synthesis (LASOL), Chemical, Pharmaceutical and Food Sciences Center (CCQFA), Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Eder João Lenardão
- Laboratory of Clean Organic Synthesis (LASOL), Chemical, Pharmaceutical and Food Sciences Center (CCQFA), Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Cristiani Folharini Bortolatto
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Chemical, Pharmaceutical and Food Sciences Center (CCQFA), Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - César Augusto Brüning
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Chemical, Pharmaceutical and Food Sciences Center (CCQFA), Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| |
Collapse
|
36
|
Borrell-Pages M, Luquero A, Vilahur G, Padró T, Badimon L. Canonical Wnt pathway and the LDL receptor superfamily in neuronal cholesterol homeostasis and function. Cardiovasc Res 2024; 120:140-151. [PMID: 37882606 DOI: 10.1093/cvr/cvad159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/13/2023] [Accepted: 09/04/2023] [Indexed: 10/27/2023] Open
Abstract
AIMS There is little information on the regulation of cholesterol homeostasis in the brain. Whether cholesterol crosses the blood-brain barrier is under investigation, but the present understanding is that cholesterol metabolism in the brain is independent from that in peripheral tissues. Lipoprotein receptors from the LDL receptor family (LRPs) have key roles in lipid particle accumulation in cells involved in vascular and cardiac pathophysiology; however, their function on neural cells is unknown. METHODS AND RESULTS The expression of LRP5 and the components and targets of its downstream signalling pathway, the canonical Wnt pathway, including β-catenin, LEF1, VEGF, OPN, MMP7, and ADAM10, is analysed in the brains of Wt and Lrp5-/- mice and in a neuroblastoma cell line. LRP5 expression is increased in a time- and dose-dependent manner after lipid loading in neuronal cells; however, it does not participate in cholesterol homeostasis as shown by intracellular lipid accumulation analyses. Neurons challenged with staurosporin and H2O2 display an anti-apoptotic protective role for LRP5. CONCLUSIONS For the first time, it has been shown that neurons can accumulate intracellular lipids and lipid uptake is performed mainly by the LDLR, while CD36, LRP1, and LRP5 do not play a major role. In addition, it has been shown that LRP5 triggers the canonical Wnt pathway in neuronal cells to generate pro-survival signals. Finally, Lrp5-/- mice have maintained expression of LRP5 only in the brain supporting the biological plausible concept of the need of brain LRP5 to elicit pro-survival processes and embryonic viability.
Collapse
Affiliation(s)
- Maria Borrell-Pages
- Cardiovascular Program ICCC, Sant Pau Institute for Biomedical Research (IIB-Sant Pau), C/Sant Antoni Maria Claret 167, Barcelona 08025, Spain
- CIBER-CV, Av. Monforte de Lemos, 3-5, 28029 Madrid, Spain
| | - Aureli Luquero
- Cardiovascular Program ICCC, Sant Pau Institute for Biomedical Research (IIB-Sant Pau), C/Sant Antoni Maria Claret 167, Barcelona 08025, Spain
- CIBER-CV, Av. Monforte de Lemos, 3-5, 28029 Madrid, Spain
| | - Gemma Vilahur
- Cardiovascular Program ICCC, Sant Pau Institute for Biomedical Research (IIB-Sant Pau), C/Sant Antoni Maria Claret 167, Barcelona 08025, Spain
- CIBER-CV, Av. Monforte de Lemos, 3-5, 28029 Madrid, Spain
| | - Teresa Padró
- Cardiovascular Program ICCC, Sant Pau Institute for Biomedical Research (IIB-Sant Pau), C/Sant Antoni Maria Claret 167, Barcelona 08025, Spain
- CIBER-CV, Av. Monforte de Lemos, 3-5, 28029 Madrid, Spain
| | - Lina Badimon
- Cardiovascular Program ICCC, Sant Pau Institute for Biomedical Research (IIB-Sant Pau), C/Sant Antoni Maria Claret 167, Barcelona 08025, Spain
- CIBER-CV, Av. Monforte de Lemos, 3-5, 28029 Madrid, Spain
- Cardiovascular Research Chair, Universitat Autònoma de Barcelona, Plaça Cívica, 08193 Bellaterra, Barcelona, Spain
| |
Collapse
|
37
|
Nabizadeh F, Valizadeh P, Fallahi MS. Bile acid profile associated with CSF and PET biomarkers in Alzheimer's disease. Aging Clin Exp Res 2024; 36:62. [PMID: 38451317 PMCID: PMC10920417 DOI: 10.1007/s40520-024-02729-3] [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: 10/04/2023] [Accepted: 02/23/2024] [Indexed: 03/08/2024]
Abstract
BACKGROUND Recent studies have shown that gut microbiota can affect the development of Alzheimer's disease (AD) through various mechanisms. Bile acids (BAs), which are the final byproducts of cholesterol metabolism created through both the human body and gut microbiome, appear to be influenced by gut microbiota and may impact AD pathological characteristics such as the accumulation of tau and amyloid-β. We aimed to investigate the associations between various serum BAs and CSF biomarkers (including Aβ, total tau, and p-tau). Additionally, we sought to examine the longitudinal changes in brain Aβ and tau through PET imaging in relation to BAs profile. METHODS The data of 828 subjects including 491 diagnosed with mild cognitive impairment (MCI), 119 patients diagnosed with AD, and 267 cognitively normal (CN) participants were obtained from ADNI. The baseline and longitudinal [18F] florbetapir and [18F] flortaucipir PET standard uptake value ratios (SUVR) measures were obtained to assess the accumulation of tau and Aβ. Moreover, baseline levels of serum BAs and CSF Aβ1-42, tau, and p-tau were used. RESULTS After FDR correction we observed that five BAs level and relevant calculated ratios were associated with CSF p-tau and tau, three with CSF Aβ1-42. Furthermore, three BAs level and relevant calculated ratios were associated with the tau-PET rate of change, and two with the Aβ rate of change. CONCLUSION The findings from our study suggest a correlation between altered profiles of BAs and CSF and imaging biomarkers associated with AD. These results provide supporting evidence for the link between the gut microbiome and the pathological features of AD.
Collapse
Affiliation(s)
- Fardin Nabizadeh
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Parya Valizadeh
- School of Medicine, Tehran University of Medical Science, Tehran, Iran
| | | |
Collapse
|
38
|
Lu Y, Xu K, Kang B, Pierce BL, Yang F, Chen LS. An integrative multi-context Mendelian randomization method for identifying risk genes across human tissues. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.03.04.24303731. [PMID: 38496462 PMCID: PMC10942526 DOI: 10.1101/2024.03.04.24303731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Mendelian randomization (MR) provides valuable assessments of the causal effect of exposure on outcome, yet the application of conventional MR methods for mapping risk genes encounters new challenges. One of the issues is the limited availability of expression quantitative trait loci (eQTLs) as instrumental variables (IVs), hampering the estimation of sparse causal effects. Additionally, the often context/tissue-specific eQTL effects challenge the MR assumption of consistent IV effects across eQTL and GWAS data. To address these challenges, we propose a multi-context multivariable integrative MR framework, mintMR, for mapping expression and molecular traits as joint exposures. It models the effects of molecular exposures across multiple tissues in each gene region, while simultaneously estimating across multiple gene regions. It uses eQTLs with consistent effects across more than one tissue type as IVs, improving IV consistency. A major innovation of mintMR involves employing multi-view learning methods to collectively model latent indicators of disease relevance across multiple tissues, molecular traits, and gene regions. The multi-view learning captures the major patterns of disease-relevance and uses these patterns to update the estimated tissue relevance probabilities. The proposed mintMR iterates between performing a multi-tissue MR for each gene region and joint learning the disease-relevant tissue probabilities across gene regions, improving the estimation of sparse effects across genes. We apply mintMR to evaluate the causal effects of gene expression and DNA methylation for 35 complex traits using multi-tissue QTLs as IVs. The proposed mintMR controls genome-wide inflation and offers new insights into disease mechanisms.
Collapse
|
39
|
Tang X, Yan T, Wang S, Liu Q, Yang Q, Zhang Y, Li Y, Wu Y, Liu S, Ma Y, Yang L. Treatment with β-sitosterol ameliorates the effects of cerebral ischemia/reperfusion injury by suppressing cholesterol overload, endoplasmic reticulum stress, and apoptosis. Neural Regen Res 2024; 19:642-649. [PMID: 37721296 PMCID: PMC10581587 DOI: 10.4103/1673-5374.380904] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 03/30/2023] [Accepted: 05/27/2023] [Indexed: 09/19/2023] Open
Abstract
β-Sitosterol is a type of phytosterol that occurs naturally in plants. Previous studies have shown that it has anti-oxidant, anti-hyperlipidemic, anti-inflammatory, immunomodulatory, and anti-tumor effects, but it is unknown whether β-sitosterol treatment reduces the effects of ischemic stroke. Here we found that, in a mouse model of ischemic stroke induced by middle cerebral artery occlusion, β-sitosterol reduced the volume of cerebral infarction and brain edema, reduced neuronal apoptosis in brain tissue, and alleviated neurological dysfunction; moreover, β-sitosterol increased the activity of oxygen- and glucose-deprived cerebral cortex neurons and reduced apoptosis. Further investigation showed that the neuroprotective effects of β-sitosterol may be related to inhibition of endoplasmic reticulum stress caused by intracellular cholesterol accumulation after ischemic stroke. In addition, β-sitosterol showed high affinity for NPC1L1, a key transporter of cholesterol, and antagonized its activity. In conclusion, β-sitosterol may help treat ischemic stroke by inhibiting neuronal intracellular cholesterol overload/endoplasmic reticulum stress/apoptosis signaling pathways.
Collapse
Affiliation(s)
- Xiuling Tang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi’an, Shaanxi Province, China
| | - Tao Yan
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi’an, Shaanxi Province, China
| | - Saiying Wang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi’an, Shaanxi Province, China
| | - Qingqing Liu
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi’an, Shaanxi Province, China
| | - Qi Yang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi’an, Shaanxi Province, China
| | - Yongqiang Zhang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi’an, Shaanxi Province, China
| | - Yujiao Li
- Department of Clinical Pharmacy, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu Province, China
| | - Yumei Wu
- Department of Pharmacology, School of Pharmacy, Air Force Medical University, Xi’an, Shaanxi Province, China
| | - Shuibing Liu
- Department of Pharmacology, School of Pharmacy, Air Force Medical University, Xi’an, Shaanxi Province, China
| | - Yulong Ma
- Department of Anesthesiology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Le Yang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi’an, Shaanxi Province, China
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, Air Force Medical University, Xi’an, Shaanxi Province, China
| |
Collapse
|
40
|
Jha D, Blennow K, Zetterberg H, Savas JN, Hanrieder J. Spatial neurolipidomics-MALDI mass spectrometry imaging of lipids in brain pathologies. JOURNAL OF MASS SPECTROMETRY : JMS 2024; 59:e5008. [PMID: 38445816 DOI: 10.1002/jms.5008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/03/2024] [Accepted: 01/25/2024] [Indexed: 03/07/2024]
Abstract
Given the complexity of nervous tissues, understanding neurochemical pathophysiology puts high demands on bioanalytical techniques with respect to specificity and sensitivity. Mass spectrometry imaging (MSI) has evolved to become an important, biochemical imaging technology for spatial biology in biological and translational research. The technique facilitates comprehensive, sensitive elucidation of the spatial distribution patterns of drugs, lipids, peptides, and small proteins in situ. Matrix-assisted laser desorption ionization (MALDI)-based MSI is the dominating modality due to its broad applicability and fair compromise of selectivity, sensitivity price, throughput, and ease of use. This is particularly relevant for the analysis of spatial lipid patterns, where no other comparable spatial profiling tools are available. Understanding spatial lipid biology in nervous tissue is therefore a key and emerging application area of MSI research. The aim of this review is to give a concise guide through the MSI workflow for lipid imaging in central nervous system (CNS) tissues and essential parameters to consider while developing and optimizing MSI assays. Further, this review provides a broad overview of key developments and applications of MALDI MSI-based spatial neurolipidomics to map lipid dynamics in neuronal structures, ultimately contributing to a better understanding of neurodegenerative disease pathology.
Collapse
Affiliation(s)
- Durga Jha
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, Mölndal, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, Mölndal, Sweden
- Clinical Neurochemistry Lab, Sahlgrenska University Hospital, Mölndal, Sweden
- Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, China
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, Mölndal, Sweden
- Clinical Neurochemistry Lab, Sahlgrenska University Hospital, Mölndal, Sweden
- UK Dementia Research Institute at UCL, London, UK
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Jeffrey N Savas
- Department of Neurology, Northwestern University Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Jörg Hanrieder
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, Mölndal, Sweden
- Clinical Neurochemistry Lab, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London, UK
| |
Collapse
|
41
|
Aldworth H, Hooper NM. Post-translational regulation of the low-density lipoprotein receptor provides new targets for cholesterol regulation. Biochem Soc Trans 2024; 52:431-440. [PMID: 38329179 PMCID: PMC10903450 DOI: 10.1042/bst20230918] [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: 11/14/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 02/09/2024]
Abstract
The amount of the low-density lipoprotein receptor (LDLR) on the surface of hepatocytes is the primary determinant of plasma low-density lipoprotein (LDL)-cholesterol level. Although the synthesis and cellular trafficking of the LDLR have been well-documented, there is growing evidence of additional post-translational mechanisms that regulate or fine tune the surface availability of the LDLR, thus modulating its ability to bind and internalise LDL-cholesterol. Proprotein convertase subtilisin/kexin type 9 and the asialoglycoprotein receptor 1 both independently interact with the LDLR and direct it towards the lysosome for degradation. While ubiquitination by the E3 ligase inducible degrader of the LDLR also targets the receptor for lysosomal degradation, ubiquitination of the LDLR by a different E3 ligase, RNF130, redistributes the receptor away from the plasma membrane. The activity of the LDLR is also regulated by proteolysis. Proteolytic cleavage of the transmembrane region of the LDLR by γ-secretase destabilises the receptor, directing it to the lysosome for degradation. Shedding of the extracellular domain of the receptor by membrane-type 1 matrix metalloprotease and cleavage of the receptor in its LDL-binding domain by bone morphogenetic protein-1 reduces the ability of the LDLR to bind and internalise LDL-cholesterol at the cell surface. A better understanding of how the activity of the LDLR is regulated will not only unravel the complex biological mechanisms controlling LDL-cholesterol metabolism but also could help inform the development of alternative pharmacological intervention strategies for the treatment of hypercholesterolaemia.
Collapse
Affiliation(s)
- Harry Aldworth
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, U.K
| | - Nigel M. Hooper
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, U.K
| |
Collapse
|
42
|
Ortlund E, Chen CY, Maner-Smith K, Khadka M, Ahn J, Gulbin X, Ivanova A, Dammer E, Seyfried N, Bennett D, Hajjar I. Integrative brain omics approach reveals key role for sn-1 lysophosphatidylethanolamine in Alzheimer's dementia. RESEARCH SQUARE 2024:rs.3.rs-3973736. [PMID: 38464293 PMCID: PMC10925467 DOI: 10.21203/rs.3.rs-3973736/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
The biology of individual lipid species and their relevance in Alzheimer's disease (AD) remains incompletely understood. We utilized non-targeted mass spectrometry to examine brain lipids variations across 316 post-mortem brains from participants in the Religious Orders Study (ROS) or Rush Memory and Aging Project (MAP) cohorts classified as either control, asymptomatic AD (AAD), or symptomatic AD (SAD) and integrated the lipidomics data with untargeted proteomic characterization on the same individuals. Lipid enrichment analysis and analysis of variance identified significantly lower abundance of lysophosphatidylethanolamine (LPE) and lysophosphatidylcholine (LPC) species in SAD than controls or AAD. Lipid-protein co-expression network analyses revealed that lipid modules consisting of LPE and LPC exhibited a significant association to protein modules associated with MAPK/metabolism, post-synaptic density, and Cell-ECM interaction pathways and were associated with better antemortem cognition and with neuropathological changes seen in AD. Particularly, LPE 22:6 [sn-1] levels are significantly decreased across AD cases (SAD) and show the most influence on protein changes compared to other lysophospholipid species. LPE 22:6 may be a lipid signature for AD and could be leveraged as potential therapeutic or dietary targets for AD.
Collapse
|
43
|
Michno W, Bowman A, Jha D, Minta K, Ge J, Koutarapu S, Zetterberg H, Blennow K, Lashley T, Heeren RMA, Hanrieder J. Spatial Neurolipidomics at the Single Amyloid-β Plaque Level in Postmortem Human Alzheimer's Disease Brain. ACS Chem Neurosci 2024; 15:877-888. [PMID: 38299453 PMCID: PMC10885149 DOI: 10.1021/acschemneuro.4c00006] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 02/02/2024] Open
Abstract
Lipid dysregulations have been critically implicated in Alzheimer's disease (AD) pathology. Chemical analysis of amyloid-β (Aβ) plaque pathology in transgenic AD mouse models has demonstrated alterations in the microenvironment in the direct proximity of Aβ plaque pathology. In mouse studies, differences in lipid patterns linked to structural polymorphism among Aβ pathology, such as diffuse, immature, and mature fibrillary aggregates, have also been reported. To date, no comprehensive analysis of neuronal lipid microenvironment changes in human AD tissue has been performed. Here, for the first time, we leverage matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) through a high-speed and spatial resolution commercial time-of-light instrument, as well as a high-mass-resolution in-house-developed orbitrap system to characterize the lipid microenvironment in postmortem human brain tissue from AD patients carrying Presenilin 1 mutations (PSEN1) that lead to familial forms of AD (fAD). Interrogation of the spatially resolved MSI data on a single Aβ plaque allowed us to verify nearly 40 sphingolipid and phospholipid species from diverse subclasses being enriched and depleted, in relation to the Aβ deposits. This included monosialo-gangliosides (GM), ceramide monohexosides (HexCer), ceramide-1-phosphates (CerP), ceramide phosphoethanolamine conjugates (PE-Cer), sulfatides (ST), as well as phosphatidylinositols (PI), phosphatidylethanolamines (PE), and phosphatidic acid (PA) species (including Lyso-forms). Indeed, many of the sphingolipid species overlap with the species previously seen in transgenic AD mouse models. Interestingly, in comparison to the animal studies, we observed an increased level of localization of PE and PI species containing arachidonic acid (AA). These findings are highly relevant, demonstrating for the first time Aβ plaque pathology-related alteration in the lipid microenvironment in humans. They provide a basis for the development of potential lipid biomarkers for AD characterization and insight into human-specific molecular pathway alterations.
Collapse
Affiliation(s)
- Wojciech Michno
- Department
of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Mölndal 43180, Sweden
- Department
of Neuroscience, Physiology and Pharmacology, University College London, London WC1E6BT, United
Kingdom
- Department
of Public Health and Caring Sciences, Uppsala
University, Uppsala 75237, Sweden
- Science
for Life Laboratory (SciLife), Uppsala University, Uppsala 75237, Sweden
| | - Andrew Bowman
- Maastricht
MultiModal Molecular Imaging Institute (M4I), Maastricht University, Maastricht 6229 ER, The Netherlands
| | - Durga Jha
- Department
of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Mölndal 43180, Sweden
| | - Karolina Minta
- Department
of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Mölndal 43180, Sweden
| | - Junyue Ge
- Department
of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Mölndal 43180, Sweden
| | - Srinivas Koutarapu
- Department
of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Mölndal 43180, Sweden
| | - Henrik Zetterberg
- Department
of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Mölndal 43180, Sweden
- Clinical
Neurochemistry Laboratory, Sahlgrenska University
Hospital, Mölndal 43180, Sweden
- Department
of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, Queen Square, London WC1N 3BG, United
Kingdom
- UK
Dementia Research Institute at UCL, London WC1E 6BT, United Kingdom
- Hong
Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong 999077, China
- Wisconsin
Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University
of Wisconsin-Madison, Madison, Wisconsin 53726, United States
| | - Kaj Blennow
- Department
of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Mölndal 43180, Sweden
- Clinical
Neurochemistry Laboratory, Sahlgrenska University
Hospital, Mölndal 43180, Sweden
- Paris Brain
Institute, ICM, Pitié-Salpêtrière
Hospital, Sorbonne University, Paris 75005, France
- Neurodegenerative
Disorder Research Center, Division of Life Sciences
and Medicine, Department of Neurology, Institute on Aging and Brain
Disorders, University of Science and Technology
of China and First Affiliated Hospital of USTC, Hefei 230001, P. R. China
| | - Tammaryn Lashley
- Department
of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, Queen Square, London WC1N 3BG, United
Kingdom
- Queen Square Brain Bank for Neurological
Disorders, Department of
Clinical and Movement Neurosciences, Institute of Neurology, University College London, London WC1N 1PJ, United Kingdom
| | - Ron M. A. Heeren
- Maastricht
MultiModal Molecular Imaging Institute (M4I), Maastricht University, Maastricht 6229 ER, The Netherlands
| | - Jörg Hanrieder
- Department
of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Mölndal 43180, Sweden
- Clinical
Neurochemistry Laboratory, Sahlgrenska University
Hospital, Mölndal 43180, Sweden
- Department
of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, Queen Square, London WC1N 3BG, United
Kingdom
- Science for Life
Laboratory (SciLife), University of Gothenburg, Gothenburg 40530, Sweden
| |
Collapse
|
44
|
Wang Y, Gao L. Cholesterol: A friend to viruses. Int Rev Immunol 2024; 43:248-262. [PMID: 38372266 DOI: 10.1080/08830185.2024.2314577] [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: 09/17/2023] [Accepted: 01/28/2024] [Indexed: 02/20/2024]
Abstract
Cholesterol is a key life-sustaining molecule which regulates membrane fluidity and serves as a signaling mediator. Cholesterol homeostasis is closely related to various pathological conditions including tumor, obesity, atherosclerosis, Alzheimer's disease and viral infection. Viral infection disrupts host cholesterol homeostasis, facilitating their own survival. Meanwhile, the host cells strive to reduce cholesterol accessibility to limit viral infection. This review focuses on the regulation of cholesterol metabolism and the role of cholesterol in viral infection, specifically providing an overview of cholesterol as a friend to promote viral entry, replication, assembly, release and immune evasion, which might inspire valuable thinking for pathogenesis and intervention of viral infection.
Collapse
Affiliation(s)
- Yingchun Wang
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Infection and Immunity, and Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
| | - Lifen Gao
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Infection and Immunity, and Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
| |
Collapse
|
45
|
Wang X, Li H, Sheng Y, He B, Liu Z, Li W, Yu S, Wang J, Zhang Y, Chen J, Qin L, Meng X. The function of sphingolipids in different pathogenesis of Alzheimer's disease: A comprehensive review. Biomed Pharmacother 2024; 171:116071. [PMID: 38183741 DOI: 10.1016/j.biopha.2023.116071] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 12/11/2023] [Accepted: 12/21/2023] [Indexed: 01/08/2024] Open
Abstract
Sphingolipids (SPLs) represent a highly diverse and structurally complex lipid class. The discussion of SPL metabolism-related issues is of importance in understanding the neuropathological progression of Alzheimer's disease (AD). AD is characterized by the accumulation of extracellular deposits of the amyloid β-peptide (Aβ) and intraneuronal aggregates of the microtubule-associated protein tau. Critical roles of Aβ oligomer deposited and ganglioside GM1 could be formed as "seed" from insoluble GAβ polymer in initiating the pathogenic process, while tau might also mediate SPLs and their toxicity. The interaction between ceramide and α-Synuclein (α-Syn) accelerates the aggregation of ferroptosis and exacerbates the pathogenesis of AD. For instance, reducing the levels of SPLs can mitigate α-Syn accumulation and inhibit AD progression. Meanwhile, loss of SPLs may inhibit the expression of APOE4 and confer protection against AD, while the loss of APOE4 expression also disrupts SPLs homeostasis. Moreover, the heightened activation of sphingomyelinase promotes the ferroptosis signaling pathway, leading to exacerbated AD symptoms. Ferroptosis plays a vital role in the pathological progression of AD by influencing Aβ, tau, APOE, and α-Syn. Conversely, the development of AD also exacerbates the manifestation of ferroptosis and SPLs. We are compiling the emerging techniques (Derivatization and IM-MS) of sphingolipidomics, to overcome the challenges of AD diagnosis and treatment. In this review, we examined the intricate neuro-mechanistic interactions between SPLs and Aβ, tau, α-Syn, APOE, and ferroptosis, mediating the onset of AD. Furthermore, our findings highlight the potential of targeting SPLs as underexplored avenue for devising innovative therapeutic strategies against AD.
Collapse
Affiliation(s)
- Xinyi Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Huaqiang Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Yunjie Sheng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Bingqian He
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Zeying Liu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Wanli Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Shujie Yu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Jiajing Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Yixin Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Jianyu Chen
- Fujian University of Traditional Chinese Medicine, School of Pharmacy, Fuzhou, Fujian 350122, PR China.
| | - Luping Qin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China.
| | - Xiongyu Meng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China.
| |
Collapse
|
46
|
Kawade N, Yamanaka K. Novel insights into brain lipid metabolism in Alzheimer's disease: Oligodendrocytes and white matter abnormalities. FEBS Open Bio 2024; 14:194-216. [PMID: 37330425 PMCID: PMC10839347 DOI: 10.1002/2211-5463.13661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/07/2023] [Accepted: 06/14/2023] [Indexed: 06/19/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. A genome-wide association study has shown that several AD risk genes are involved in lipid metabolism. Additionally, epidemiological studies have indicated that the levels of several lipid species are altered in the AD brain. Therefore, lipid metabolism is likely changed in the AD brain, and these alterations might be associated with an exacerbation of AD pathology. Oligodendrocytes are glial cells that produce the myelin sheath, which is a lipid-rich insulator. Dysfunctions of the myelin sheath have been linked to white matter abnormalities observed in the AD brain. Here, we review the lipid composition and metabolism in the brain and myelin and the association between lipidic alterations and AD pathology. We also present the abnormalities in oligodendrocyte lineage cells and white matter observed in AD. Additionally, we discuss metabolic disorders, including obesity, as AD risk factors and the effects of obesity and dietary intake of lipids on the brain.
Collapse
Affiliation(s)
- Noe Kawade
- Department of Neuroscience and Pathobiology, Research Institute of Environmental MedicineNagoya UniversityJapan
- Department of Neuroscience and Pathobiology, Nagoya University Graduate School of MedicineNagoya UniversityJapan
| | - Koji Yamanaka
- Department of Neuroscience and Pathobiology, Research Institute of Environmental MedicineNagoya UniversityJapan
- Department of Neuroscience and Pathobiology, Nagoya University Graduate School of MedicineNagoya UniversityJapan
- Institute for Glyco‐core Research (iGCORE)Nagoya UniversityJapan
- Center for One Medicine Innovative Translational Research (COMIT)Nagoya UniversityJapan
| |
Collapse
|
47
|
Alavez-Rubio JS, Juarez-Cedillo T. Microglia as a Possible Alternative Therapeutic for Dementia. J Alzheimers Dis Rep 2024; 8:43-56. [PMID: 38229830 PMCID: PMC10789290 DOI: 10.3233/adr-230112] [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: 08/21/2023] [Accepted: 12/04/2023] [Indexed: 01/18/2024] Open
Abstract
Dementia is a syndrome in which there is deterioration in memory, behavior, and the ability to perform everyday activities. Alzheimer's disease and vascular dementia are the most common forms of dementia. There is evidence supporting the hypothesis that inflammatory and immune mechanisms are involved in dementia. Microglia, the resident macrophage tissues in the central nervous system, play a significant role in neuroinflammation and play an important role in amyloid-β clearance in the brain, and impaired microglial clearance of amyloid-β has also been shown to be involved in the pathogenesis of Alzheimer's disease. However, there is also abundant evidence that microglia have harmful actions in dementia. Once activated, they can mediate uptake at neuronal synapses. They can also exacerbate tau pathology and secrete deleterious inflammatory factors that can directly or indirectly damage neurons. Thus, depending on the stage of the disease, microglia can act both protectively and detrimentally. Therefore, it is still necessary to continue with studies to better understand the role of microglia in the pathology of dementia. Currently available drugs can only improve cognitive symptoms, have no impact on progression and are not curative, so identifying and studying new therapeutic approaches is important. Considering the role played by microglia in this pathology, it has been pointed out as a possible therapeutic approach. This manuscript aims to address the relationship between microglia and dementia and how this relationship could be used for therapeutic purposes.
Collapse
Affiliation(s)
| | - Teresa Juarez-Cedillo
- Unidad de Investigación Epidemiológica y en Servicios de Salud, Área de Envejecimiento, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico
| |
Collapse
|
48
|
Area-Gomez E, Schon EA. Towards a Unitary Hypothesis of Alzheimer's Disease Pathogenesis. J Alzheimers Dis 2024; 98:1243-1275. [PMID: 38578892 DOI: 10.3233/jad-231318] [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: 04/07/2024]
Abstract
The "amyloid cascade" hypothesis of Alzheimer's disease (AD) pathogenesis invokes the accumulation in the brain of plaques (containing the amyloid-β protein precursor [AβPP] cleavage product amyloid-β [Aβ]) and tangles (containing hyperphosphorylated tau) as drivers of pathogenesis. However, the poor track record of clinical trials based on this hypothesis suggests that the accumulation of these peptides is not the only cause of AD. Here, an alternative hypothesis is proposed in which the AβPP cleavage product C99, not Aβ, is the main culprit, via its role as a regulator of cholesterol metabolism. C99, which is a cholesterol sensor, promotes the formation of mitochondria-associated endoplasmic reticulum (ER) membranes (MAM), a cholesterol-rich lipid raft-like subdomain of the ER that communicates, both physically and biochemically, with mitochondria. We propose that in early-onset AD (EOAD), MAM-localized C99 is elevated above normal levels, resulting in increased transport of cholesterol from the plasma membrane to membranes of intracellular organelles, such as ER/endosomes, thereby upregulating MAM function and driving pathology. By the same token, late-onset AD (LOAD) is triggered by any genetic variant that increases the accumulation of intracellular cholesterol that, in turn, boosts the levels of C99 and again upregulates MAM function. Thus, the functional cause of AD is upregulated MAM function that, in turn, causes the hallmark disease phenotypes, including the plaques and tangles. Accordingly, the MAM hypothesis invokes two key interrelated elements, C99 and cholesterol, that converge at the MAM to drive AD pathogenesis. From this perspective, AD is, at bottom, a lipid disorder.
Collapse
Affiliation(s)
- Estela Area-Gomez
- Department of Neurology, Columbia University, New York, NY, USA
- Centro de Investigaciones Biológicas "Margarita Salas", Spanish National Research Council, Madrid, Spain
| | - Eric A Schon
- Department of Neurology, Columbia University, New York, NY, USA
- Department of Genetics and Development>, Columbia University, New York, NY, USA
| |
Collapse
|
49
|
Moreno-Rodriguez M, Perez SE, Martinez-Gardeazabal J, Manuel I, Malek-Ahmadi M, Rodriguez-Puertas R, Mufson EJ. Frontal Cortex Lipid Alterations During the Onset of Alzheimer's Disease. J Alzheimers Dis 2024; 98:1515-1532. [PMID: 38578893 DOI: 10.3233/jad-231485] [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: 04/07/2024]
Abstract
Background Although sporadic Alzheimer's disease (AD) is a neurodegenerative disorder of unknown etiology, familial AD is associated with specific gene mutations. A commonality between these forms of AD is that both display multiple pathogenic events including cholinergic and lipid dysregulation. Objective We aimed to identify the relevant lipids and the activity of their related receptors in the frontal cortex and correlating them with cognition during the progression of AD. Methods MALDI-mass spectrometry imaging (MSI) and functional autoradiography was used to evaluate the distribution of phospholipids/sphingolipids and the activity of cannabinoid 1 (CB1), sphingosine 1-phosphate 1 (S1P1), and muscarinic M2/M4 receptors in the frontal cortex (FC) of people that come to autopsy with premortem clinical diagnosis of AD, mild cognitive impairment (MCI), and no cognitive impairment (NCI). Results MALDI-MSI revealed an increase in myelin-related lipids, such as diacylglycerol (DG) 36:1, DG 38:5, and phosphatidic acid (PA) 40:6 in the white matter (WM) in MCI compared to NCI, and a downregulation of WM phosphatidylinositol (PI) 38:4 and PI 38:5 levels in AD compared to NCI. Elevated levels of phosphatidylcholine (PC) 32:1, PC 34:0, and sphingomyelin 38:1 were observed in discrete lipid accumulations in the FC supragranular layers during disease progression. Muscarinic M2/M4 receptor activation in layers V-VI decreased in AD compared to MCI. CB1 receptor activity was upregulated in layers V-VI, while S1P1 was downregulated within WM in AD relative to NCI. Conclusions FC WM lipidomic alterations are associated with myelin dyshomeostasis in prodromal AD, suggesting WM lipid maintenance as a potential therapeutic target for dementia.
Collapse
Affiliation(s)
- Marta Moreno-Rodriguez
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Sylvia E Perez
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ, USA
| | | | - Ivan Manuel
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country, Leioa, Spain
- Neurodegenerative Diseases, BioBizkaia Health Research Institute, Barakaldo, Spain
| | | | - Rafael Rodriguez-Puertas
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country, Leioa, Spain
- Neurodegenerative Diseases, BioBizkaia Health Research Institute, Barakaldo, Spain
| | - Elliott J Mufson
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ, USA
| |
Collapse
|
50
|
Perluigi M, Di Domenico F, Butterfield DA. Oxidative damage in neurodegeneration: roles in the pathogenesis and progression of Alzheimer disease. Physiol Rev 2024; 104:103-197. [PMID: 37843394 PMCID: PMC11281823 DOI: 10.1152/physrev.00030.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 03/30/2023] [Accepted: 05/24/2023] [Indexed: 10/17/2023] Open
Abstract
Alzheimer disease (AD) is associated with multiple etiologies and pathological mechanisms, among which oxidative stress (OS) appears as a major determinant. Intriguingly, OS arises in various pathways regulating brain functions, and it seems to link different hypotheses and mechanisms of AD neuropathology with high fidelity. The brain is particularly vulnerable to oxidative damage, mainly because of its unique lipid composition, resulting in an amplified cascade of redox reactions that target several cellular components/functions ultimately leading to neurodegeneration. The present review highlights the "OS hypothesis of AD," including amyloid beta-peptide-associated mechanisms, the role of lipid and protein oxidation unraveled by redox proteomics, and the antioxidant strategies that have been investigated to modulate the progression of AD. Collected studies from our groups and others have contributed to unraveling the close relationships between perturbation of redox homeostasis in the brain and AD neuropathology by elucidating redox-regulated events potentially involved in both the pathogenesis and progression of AD. However, the complexity of AD pathological mechanisms requires an in-depth understanding of several major intracellular pathways affecting redox homeostasis and relevant for brain functions. This understanding is crucial to developing pharmacological strategies targeting OS-mediated toxicity that may potentially contribute to slow AD progression as well as improve the quality of life of persons with this severe dementing disorder.
Collapse
Affiliation(s)
- Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi Fanelli," Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi Fanelli," Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - D Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States
| |
Collapse
|