1
|
Wang L, Zhang X, Yang Z, Wang B, Gong H, Zhang K, Lin Y, Sun M. Extracellular vesicles: biological mechanisms and emerging therapeutic opportunities in neurodegenerative diseases. Transl Neurodegener 2024; 13:60. [PMID: 39643909 PMCID: PMC11622582 DOI: 10.1186/s40035-024-00453-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 11/14/2024] [Indexed: 12/09/2024] Open
Abstract
Extracellular vesicles (EVs) are membrane vesicles originating from different cells within the brain. The pathophysiological role of EVs in neurodegenerative diseases is progressively acknowledged. This field has advanced from basic biological research to essential clinical significance. The capacity to selectively enrich specific subsets of EVs from biofluids via distinctive surface markers has opened new avenues for molecular understandings across various tissues and organs, notably in the brain. In recent years, brain-derived EVs have been extensively investigated as biomarkers, therapeutic targets, and drug-delivery vehicles for neurodegenerative diseases. This review provides a brief overview of the characteristics and physiological functions of the various classes of EVs, focusing on the biological mechanisms by which various types of brain-derived EVs mediate the occurrence and development of neurodegenerative diseases. Concurrently, novel therapeutic approaches and challenges for the use of EVs as delivery vehicles are delineated.
Collapse
Affiliation(s)
- Ling Wang
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Xiaoyan Zhang
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Ziyi Yang
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Binquan Wang
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Hongyang Gong
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Ke Zhang
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Yi Lin
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Mingkuan Sun
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
2
|
Allen MI, Johnson BN, Kumar A, Su Y, Singh S, Deep G, Nader MA. Behavioral and neuronal extracellular vesicle biomarkers associated with nicotine's enhancement of the reinforcing strength of cocaine in female and male monkeys. ADDICTION NEUROSCIENCE 2024; 11:100151. [PMID: 38911873 PMCID: PMC11192513 DOI: 10.1016/j.addicn.2024.100151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/25/2024]
Abstract
While the majority of people with cocaine use disorders (CUD) also co-use tobacco/nicotine, most preclinical cocaine research does not include nicotine. The present study examined nicotine and cocaine co-use under several conditions of intravenous drug self-administration in monkeys, as well as potential peripheral biomarkers associated with co-use. In Experiment 1, male rhesus monkeys (N = 3) self-administered cocaine (0.001-0.1 mg/kg/injection) alone and with nicotine (0.01-0.03 mg/kg/injection) under a progressive-ratio schedule of reinforcement. When nicotine was added to cocaine, there was a significant leftward/upward shift in the number of injections received. In Experiment 2, socially housed female and male cynomolgus monkeys (N = 14) self-administered cocaine under a concurrent drug-vs-food choice schedule of reinforcement. Adding nicotine to the cocaine solution shifted the cocaine dose-response curves to the left, with more robust shifts noted in the female animals. There was no evidence of social rank differences. To assess reinforcing strength, delays were added to the presentation of drug; the co-use of nicotine and cocaine required significantly longer delays to decrease drug choice, compared with cocaine alone. Blood samples obtained post-session were used to analyze concentrations of neuronally derived small extracellular vesicles (NDE); significant differences in NDE profile were observed for kappa-opioid receptors when nicotine and cocaine were co-used compared with each drug alone and controls. These results suggest that drug interactions involving the co-use of nicotine and cocaine are not simply changing potency, but rather resulting in changes in reinforcing strength that should be utilized to better understand the neuropharmacology of CUD and the evaluation of potential treatments.
Collapse
Affiliation(s)
- Mia I. Allen
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC, United States
- Center for Addiction Research, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Bernard N. Johnson
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC, United States
- Center for Addiction Research, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Ashish Kumar
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Yixin Su
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Sangeeta Singh
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Gagan Deep
- Center for Addiction Research, Wake Forest University School of Medicine, Winston-Salem, NC, United States
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
- J Paul Sticht Center for Healthy Aging and Alzheimer’s Prevention, School of Medicine, Wake Forest University, Winston-Salem, NC, United States
- Department of Cancer Biology, School of Medicine, Wake Forest University, Winston-Salem, NC, United States
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, United States
| | - Michael A. Nader
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC, United States
- Center for Addiction Research, Wake Forest University School of Medicine, Winston-Salem, NC, United States
- Department of Radiology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
3
|
Kumar A, Nader MA, Deep G. Emergence of Extracellular Vesicles as "Liquid Biopsy" for Neurological Disorders: Boom or Bust. Pharmacol Rev 2024; 76:199-227. [PMID: 38351075 PMCID: PMC10877757 DOI: 10.1124/pharmrev.122.000788] [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: 11/23/2022] [Revised: 11/11/2023] [Accepted: 11/27/2023] [Indexed: 02/16/2024] Open
Abstract
Extracellular vesicles (EVs) have emerged as an attractive liquid biopsy approach in the diagnosis and prognosis of multiple diseases and disorders. The feasibility of enriching specific subpopulations of EVs from biofluids based on their unique surface markers has opened novel opportunities to gain molecular insight from various tissues and organs, including the brain. Over the past decade, EVs in bodily fluids have been extensively studied for biomarkers associated with various neurological disorders, such as Alzheimer's disease, Parkinson's disease, schizophrenia, bipolar disorder, major depressive disorders, substance use disorders, human immunodeficiency virus-associated neurocognitive disorder, and cancer/treatment-induced neurodegeneration. These studies have focused on the isolation and cargo characterization of either total EVs or brain cells, such as neuron-, astrocyte-, microglia-, oligodendrocyte-, pericyte-, and endothelial-derived EVs from biofluids to achieve early diagnosis and molecular characterization and to predict the treatment and intervention outcomes. The findings of these studies have demonstrated that EVs could serve as a repetitive and less invasive source of valuable molecular information for these neurological disorders, supplementing existing costly neuroimaging techniques and relatively invasive measures, like lumbar puncture. However, the initial excitement surrounding blood-based biomarkers for brain-related diseases has been tempered by challenges, such as lack of central nervous system specificity in EV markers, lengthy protocols, and the absence of standardized procedures for biological sample collection, EV isolation, and characterization. Nevertheless, with rapid advancements in the EV field, supported by improved isolation methods and sensitive assays for cargo characterization, brain cell-derived EVs continue to offer unparallel opportunities with significant translational implications for various neurological disorders. SIGNIFICANCE STATEMENT: Extracellular vesicles present a less invasive liquid biopsy approach in the diagnosis and prognosis of various neurological disorders. Characterizing these vesicles in biofluids holds the potential to yield valuable molecular information, thereby significantly impacting the development of novel biomarkers for various neurological disorders. This paper has reviewed the methodology employed to isolate extracellular vesicles derived from various brain cells in biofluids, their utility in enhancing the molecular understanding of neurodegeneration, and the potential challenges in this research field.
Collapse
Affiliation(s)
- Ashish Kumar
- Departments of Cancer Biology (A.K., G.D.), Physiology and Pharmacology (M.A.N.), Radiology (M.A.N.), and Center for Addiction Research (M.A.N., G.D.), Wake Forest University School of Medicine, Winston-Salem, North Carolina; Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, North Carolina (G.D.); and Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest School of Medicine, Winston-Salem, North Carolina (G.D.)
| | - Michael A Nader
- Departments of Cancer Biology (A.K., G.D.), Physiology and Pharmacology (M.A.N.), Radiology (M.A.N.), and Center for Addiction Research (M.A.N., G.D.), Wake Forest University School of Medicine, Winston-Salem, North Carolina; Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, North Carolina (G.D.); and Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest School of Medicine, Winston-Salem, North Carolina (G.D.)
| | - Gagan Deep
- Departments of Cancer Biology (A.K., G.D.), Physiology and Pharmacology (M.A.N.), Radiology (M.A.N.), and Center for Addiction Research (M.A.N., G.D.), Wake Forest University School of Medicine, Winston-Salem, North Carolina; Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, North Carolina (G.D.); and Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest School of Medicine, Winston-Salem, North Carolina (G.D.)
| |
Collapse
|
4
|
Zhuang H, Cao X, Tang X, Zou Y, Yang H, Liang Z, Yan X, Chen X, Feng X, Shen L. Investigating metabolic dysregulation in serum of triple transgenic Alzheimer's disease male mice: implications for pathogenesis and potential biomarkers. Amino Acids 2024; 56:10. [PMID: 38315232 PMCID: PMC10844422 DOI: 10.1007/s00726-023-03375-1] [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: 05/15/2023] [Accepted: 11/11/2023] [Indexed: 02/07/2024]
Abstract
Alzheimer's disease (AD) is a multifactorial neurodegenerative disease that lacks convenient and accessible peripheral blood diagnostic markers and effective drugs. Metabolic dysfunction is one of AD risk factors, which leaded to alterations of various metabolites in the body. Pathological changes of the brain can be reflected in blood metabolites that are expected to explain the disease mechanisms or be candidate biomarkers. The aim of this study was to investigate the changes of targeted metabolites within peripheral blood of AD mouse model, with the purpose of exploring the disease mechanism and potential biomarkers. Targeted metabolomics was used to quantify 256 metabolites in serum of triple transgenic AD (3 × Tg-AD) male mice. Compared with controls, 49 differential metabolites represented dysregulation in purine, pyrimidine, tryptophan, cysteine and methionine and glycerophospholipid metabolism. Among them, adenosine, serotonin, N-acetyl-5-hydroxytryptamine, and acetylcholine play a key role in regulating neural transmitter network. The alteration of S-adenosine-L-homocysteine, S-adenosine-L-methionine, and trimethylamine-N-oxide in AD mice serum can served as indicator of AD risk. The results revealed the changes of metabolites in serum, suggesting that metabolic dysregulation in periphery in AD mice may be related to the disturbances in neuroinhibition, the serotonergic system, sleep function, the cholinergic system, and the gut microbiota. This study provides novel insights into the dysregulation of several key metabolites and metabolic pathways in AD, presenting potential avenues for future research and the development of peripheral biomarkers.
Collapse
Affiliation(s)
- Hongbin Zhuang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Xueshan Cao
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Xiaoxiao Tang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Yongdong Zou
- Center for Instrumental Analysis, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Hongbo Yang
- Center for Instrumental Analysis, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Zhiyuan Liang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Xi Yan
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, People's Republic of China
| | - Xiaolu Chen
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, People's Republic of China
| | - Xingui Feng
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China
| | - Liming Shen
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, 518071, People's Republic of China.
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, People's Republic of China.
| |
Collapse
|
5
|
Ribeiro DE, Petiz LL, Glaser T, Oliveira-Giacomelli Á, Andrejew R, Saab FDAR, Milanis MDS, Campos HC, Sampaio VFA, La Banca S, Longo BM, Lameu C, Tang Y, Resende RR, Ferreira ST, Ulrich H. Purinergic signaling in cognitive impairment and neuropsychiatric symptoms of Alzheimer's disease. Neuropharmacology 2023; 226:109371. [PMID: 36502867 DOI: 10.1016/j.neuropharm.2022.109371] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/04/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022]
Abstract
About 10 million new cases of dementia develop worldwide each year, of which up to 70% are attributable to Alzheimer's disease (AD). In addition to the widely known symptoms of memory loss and cognitive impairment, AD patients frequently develop non-cognitive symptoms, referred to as behavioral and psychological symptoms of dementia (BPSDs). Sleep disorders are often associated with AD, but mood alterations, notably depression and apathy, comprise the most frequent class of BPSDs. BPSDs negatively affect the lives of AD patients and their caregivers, and have a significant impact on public health systems and the economy. Because treatments currently available for AD are not disease-modifying and mainly aim to ameliorate some of the cognitive symptoms, elucidating the mechanisms underlying mood alterations and other BPSDs in AD may reveal novel avenues for progress in AD therapy. Purinergic signaling is implicated in the pathophysiology of several central nervous system (CNS) disorders, such as AD, depression and sleep disorders. Here, we review recent findings indicating that purinergic receptors, mainly the A1, A2A, and P2X7 subtypes, are associated with the development/progression of AD. Current evidence suggests that targeting purinergic signaling may represent a promising therapeutic approach in AD and related conditions. This article is part of the Special Issue on "Purinergic Signaling: 50 years".
Collapse
Affiliation(s)
- Deidiane Elisa Ribeiro
- Department of Biochemistry, Chemistry Institute, University of São Paulo (USP), São Paulo, Brazil.
| | - Lyvia Lintzmaier Petiz
- Department of Biochemistry and Molecular Biology, Universidade Federal do Paraná, Curitiba, Brazil
| | - Talita Glaser
- Department of Biochemistry, Chemistry Institute, University of São Paulo (USP), São Paulo, Brazil
| | | | - Roberta Andrejew
- Department of Biochemistry, Chemistry Institute, University of São Paulo (USP), São Paulo, Brazil
| | | | - Milena da Silva Milanis
- Department of Biochemistry, Chemistry Institute, University of São Paulo (USP), São Paulo, Brazil
| | - Henrique Correia Campos
- Laboratory of Neurophysiology, Department of Physiology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | | | - Sophia La Banca
- Department of Biochemistry, Chemistry Institute, University of São Paulo (USP), São Paulo, Brazil
| | - Beatriz Monteiro Longo
- Laboratory of Neurophysiology, Department of Physiology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Claudiana Lameu
- Department of Biochemistry, Chemistry Institute, University of São Paulo (USP), São Paulo, Brazil
| | - Yong Tang
- International Collaborative Centre on Big Science Plan for Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China; Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, 610075, China
| | - Rodrigo Ribeiro Resende
- Department of Biochemistry and Immunology, Federal University of Minas Gerais Belo Horizonte, MG, Brazil
| | - Sergio T Ferreira
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Henning Ulrich
- Department of Biochemistry, Chemistry Institute, University of São Paulo (USP), São Paulo, Brazil; International Collaborative Centre on Big Science Plan for Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
| |
Collapse
|
6
|
Bagó-Mas A, Korimová A, Deulofeu M, Verdú E, Fiol N, Svobodová V, Dubový P, Boadas-Vaello P. Polyphenolic grape stalk and coffee extracts attenuate spinal cord injury-induced neuropathic pain development in ICR-CD1 female mice. Sci Rep 2022; 12:14980. [PMID: 36056079 PMCID: PMC9440260 DOI: 10.1038/s41598-022-19109-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 08/24/2022] [Indexed: 11/09/2022] Open
Abstract
More than half of spinal cord injury (SCI) patients develop central neuropathic pain (CNP), which is largely refractory to current treatments. Considering the preclinical evidence showing that polyphenolic compounds may exert antinociceptive effects, the present work aimed to study preventive effects on SCI-induced CNP development by repeated administration of two vegetal polyphenolic extracts: grape stalk extract (GSE) and coffee extract (CE). Thermal hyperalgesia and mechanical allodynia were evaluated at 7, 14 and 21 days postinjury. Then, gliosis, ERK phosphorylation and the expression of CCL2 and CX3CL1 chemokines and their receptors, CCR2 and CX3CR1, were analyzed in the spinal cord. Gliosis and CX3CL1/CX3CR1 expression were also analyzed in the anterior cingulate cortex (ACC) and periaqueductal gray matter (PAG) since they are supraspinal structures involved in pain perception and modulation. GSE and CE treatments modulated pain behaviors accompanied by reduced gliosis in the spinal cord and both treatments modulated neuron-glia crosstalk-related biomolecules expression. Moreover, both extracts attenuated astrogliosis in the ACC and PAG as well as microgliosis in the ACC with an increased M2 subpopulation of microglial cells in the PAG. Finally, GSE and CE prevented CX3CL1/CX3CR1 upregulation in the PAG, and modulated their expression in ACC. These findings suggest that repeated administrations of either GSE or CE after SCI may be suitable pharmacologic strategies to attenuate SCI-induced CNP development by means of spinal and supraspinal neuroinflammation modulation.
Collapse
Affiliation(s)
- Anna Bagó-Mas
- Research Group of Clinical Anatomy, Embryology and Neuroscience (NEOMA), Department of Medical Sciences, University of Girona, Girona, Spain
| | - Andrea Korimová
- Department of Anatomy, Division of Neuroanatomy, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Meritxell Deulofeu
- Research Group of Clinical Anatomy, Embryology and Neuroscience (NEOMA), Department of Medical Sciences, University of Girona, Girona, Spain
| | - Enrique Verdú
- Research Group of Clinical Anatomy, Embryology and Neuroscience (NEOMA), Department of Medical Sciences, University of Girona, Girona, Spain
| | - Núria Fiol
- Department of Chemical Engineering, Agriculture and Food Technology, Polytechnic School, University of Girona, Girona, Spain
| | - Viktorie Svobodová
- Department of Anatomy, Division of Neuroanatomy, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Petr Dubový
- Department of Anatomy, Division of Neuroanatomy, Faculty of Medicine, Masaryk University, Brno, Czechia.
| | - Pere Boadas-Vaello
- Research Group of Clinical Anatomy, Embryology and Neuroscience (NEOMA), Department of Medical Sciences, University of Girona, Girona, Spain.
| |
Collapse
|
7
|
Neuroprotective Effects of Resveratrol by Modifying Cholesterol Metabolism and Aβ Processing in SAMP8 Mice. Int J Mol Sci 2022; 23:ijms23147580. [PMID: 35886936 PMCID: PMC9324102 DOI: 10.3390/ijms23147580] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/04/2022] [Accepted: 07/07/2022] [Indexed: 11/17/2022] Open
Abstract
Cholesterol metabolism seems dysregulated and linked to amyloid-β (Aβ) formation in neurodegeneration, but the underlying mechanisms are poorly known. Resveratrol (RSV) is a polyphenol with antioxidant activity and neuroprotective properties. Here, we analyzed the effect of age and RSV supplementation on cholesterol metabolism in the brain and blood serum, and its potential link to Aβ processing, in SAMP8 mice—an animal model of aging and Alzheimer’s disease. In the brain, our results revealed an age-related increase in ApoE and unesterified cholesterol in the plasma membrane whereas LDL receptor, HMG-CoA reductase, HMG-CoA-C1 synthase, and ABCA1 transporter remained unaltered. Furthermore, BACE-1 and APP gene expression was decreased. This dysregulation could be involved in the amyloidogenic processing pathway of APP towards Aβ formation. In turn, RSV exhibited an age-dependent effect. While levels of unesterified cholesterol in the plasma membrane were not affected by RSV, several participants in cholesterol uptake, release, and de novo synthesis differed, depending on age. Thus, RSV supplementation exhibited a different neuroprotective effect acting on Aβ processing or cholesterol metabolism in the brain at earlier or later ages, respectively. In blood serum, HDL lipoprotein and free cholesterol were increased by age, whereas VLDL and LDL lipoproteins remained unaltered. Again, the protective effect of RSV by decreasing the LDL or increasing the HDL levels also seems to depend on the intervention’s moment. In conclusion, age is a prominent factor for cholesterol metabolism dysregulation in the brain of SAMP8 mice and influences the protective effects of RSV through cholesterol metabolism and Aβ processing.
Collapse
|