1
|
Lindner K, Gavin AC. Isoform- and cell-state-specific APOE homeostasis and function. Neural Regen Res 2024; 19:2456-2466. [PMID: 38526282 PMCID: PMC11090418 DOI: 10.4103/nrr.nrr-d-23-01470] [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: 08/31/2023] [Revised: 11/17/2023] [Accepted: 12/26/2023] [Indexed: 03/26/2024] Open
Abstract
Apolipoprotein E is the major lipid transporter in the brain and an important player in neuron-astrocyte metabolic coupling. It ensures the survival of neurons under stressful conditions and hyperactivity by nourishing and detoxifying them. Apolipoprotein E polymorphism, combined with environmental stresses and/or age-related alterations, influences the risk of developing late-onset Alzheimer's disease. In this review, we discuss our current knowledge of how apolipoprotein E homeostasis, i.e. its synthesis, secretion, degradation, and lipidation, is affected in Alzheimer's disease.
Collapse
Affiliation(s)
- Karina Lindner
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Anne-Claude Gavin
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
2
|
Deza‐Lougovski YI, Weiss LM, Horton HM, Sun A, Borbye‐Lorenzen N, Skogstrand K, Holmgaard S, Andersen‐Ranberg K, Lundmark VP, Börsch‐Supan A, Börsch‐Supan M, Rieckmann A. Circulating apoE4 protein levels from dried blood spots predict cognitive function in a large population-based survey setting. Alzheimers Dement 2024; 20:7613-7623. [PMID: 39234633 PMCID: PMC11567846 DOI: 10.1002/alz.14224] [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: 03/06/2024] [Revised: 07/30/2024] [Accepted: 08/07/2024] [Indexed: 09/06/2024]
Abstract
INTRODUCTION The apolipoprotein E (APOE) ε4 allele carries risk for cognitive impairment, but whether the level of circulating apoE4 protein in carriers affects cognition is unclear, as is how health and lifestyle impact circulating apoE4 levels. METHODS We assayed apoE4 protein levels in dried blood spots of 12,532 adults aged 50+. Regression analyses tested the likelihood of cognitive impairment between groups and within those with detected apoE4 protein. Predictors of circulating apoE4 were assessed. RESULTS We detected protein binding that indicates the presence of an APOE ε4 allele in 28.4% of this group. This group was more likely to have cognitive impairment, and this risk increases with age. However, higher apoE4 levels were associated with less likelihood of cognitive impairment within this group. Antihypertensive medication predicted apoE4 protein levels. DISCUSSION The apoE4 isoform is associated with a deficient protein and worse cognition. This association is modulated by the level of circulating apoE4 protein in ε4 carriers. HIGHLIGHTS An assay to quantify apoE4 levels from dried blood spot samples was applied. The apoE4 protein was detected as specific binding at ≥30,000 pg/mL in 28.4% of samples. Having the apoE4 protein was associated with worse cognitive performance. Higher apoE4 protein levels in those who have it were associated with better cognition. Cardiovascular factors influenced levels of apoE4 protein.
Collapse
Affiliation(s)
- Yacila I. Deza‐Lougovski
- Institute of Psychology, University of the Bundeswehr MünchenNeubibergGermany
- Max Planck Institute for Social Law and Social PolicyMunichGermany
| | - Luzia M. Weiss
- Max Planck Institute for Social Law and Social PolicyMunichGermany
| | - Hannah M. Horton
- Max Planck Institute for Social Law and Social PolicyMunichGermany
- Munich Research Institute for the Economics of Aging and SHARE Analyses (MEA)MunichGermany
| | - Aijing Sun
- Max Planck Institute for Social Law and Social PolicyMunichGermany
- Munich Research Institute for the Economics of Aging and SHARE Analyses (MEA)MunichGermany
| | - Nis Borbye‐Lorenzen
- Department for Congenital DisordersCenter for Neonatal Screening, Statens Serum InstitutCopenhagenDenmark
| | - Kristin Skogstrand
- Department for Congenital DisordersCenter for Neonatal Screening, Statens Serum InstitutCopenhagenDenmark
| | - Solveig Holmgaard
- Department for Congenital DisordersCenter for Neonatal Screening, Statens Serum InstitutCopenhagenDenmark
| | - Karen Andersen‐Ranberg
- Department of Public Health Epidemiology, Biostatistics and BiodemographyUniversity of Southern DenmarkOdenseDenmark
- Department of Clinical Research Geriatric Research UnitUniversity of Southern DenmarkOdenseDenmark
| | - Vania Panes Lundmark
- Department of Integrative Medical BiologyUmeå UniversityUmeåSweden
- Umeå Center for Functional Brain ImagingUmeå UniversityUmeåSweden
| | - Axel Börsch‐Supan
- Max Planck Institute for Social Law and Social PolicyMunichGermany
- Munich Research Institute for the Economics of Aging and SHARE Analyses (MEA)MunichGermany
| | - Martina Börsch‐Supan
- Munich Research Institute for the Economics of Aging and SHARE Analyses (MEA)MunichGermany
- Survey of Health, Ageing and Retirement in Europe (SHARE Biomarker Project)MunichGermany
| | - Anna Rieckmann
- Institute of Psychology, University of the Bundeswehr MünchenNeubibergGermany
- Max Planck Institute for Social Law and Social PolicyMunichGermany
| |
Collapse
|
3
|
Reis ALG, Maximino JR, Lage LADPC, Gomes HR, Pereira J, Brofman PRS, Senegaglia AC, Rebelatto CLK, Daga DR, Paiva WS, Chadi G. Proteomic analysis of cerebrospinal fluid of amyotrophic lateral sclerosis patients in the presence of autologous bone marrow derived mesenchymal stem cells. Stem Cell Res Ther 2024; 15:301. [PMID: 39278909 PMCID: PMC11403799 DOI: 10.1186/s13287-024-03820-2] [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: 11/24/2023] [Accepted: 06/27/2024] [Indexed: 09/18/2024] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a fatal and rapidly progressive motoneuron degenerative disorder. There are still no drugs capable of slowing disease evolution or improving life quality of ALS patients. Thus, autologous stem cell therapy has emerged as an alternative treatment regime to be investigated in clinical ALS. METHOD Using Proteomics and Protein-Protein Interaction Network analyses combined with bioinformatics, the possible cellular mechanisms and molecular targets related to mesenchymal stem cells (MSCs, 1 × 106 cells/kg, intrathecally in the lumbar region of the spine) were investigated in cerebrospinal fluid (CSF) of ALS patients who received intrathecal infusions of autologous bone marrow-derived MSCs thirty days after cell therapy. Data are available via ProteomeXchange with identifier PXD053129. RESULTS Proteomics revealed 220 deregulated proteins in CSF of ALS subjects treated with MSCs compared to CSF collected from the same patients prior to MSCs infusion. Bioinformatics enriched analyses highlighted events of Extracellular matrix and Cell adhesion molecules as well as related key targets APOA1, APOE, APP, C4A, C5, FGA, FGB, FGG and PLG in the CSF of cell treated ALS subjects. CONCLUSIONS Extracellular matrix and cell adhesion molecules as well as their related highlighted components have emerged as key targets of autologous MSCs in CSF of ALS patients. TRIAL REGISTRATION Clinicaltrial.gov identifier NCT0291768. Registered 28 September 2016.
Collapse
Affiliation(s)
- Ana Luiza Guimarães Reis
- Laboratorio de Neurologia Translacional, Departamento de Neurologia, Hospital das Clinicas, Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, SP, 01246-903, Brazil
| | - Jessica Ruivo Maximino
- Laboratorio de Neurologia Translacional, Departamento de Neurologia, Hospital das Clinicas, Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, SP, 01246-903, Brazil
| | | | - Hélio Rodrigues Gomes
- Departamento de Neurologia, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, SP, 01246-903, Brazil
| | - Juliana Pereira
- LIM-31, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Paulo Roberto Slud Brofman
- Core for Cell Technology, School of Medicine and Life Sciences, Pontifícia Universidade Catolica do Parana, Curitiba, PR, 80215-901, Brazil
| | - Alexandra Cristina Senegaglia
- Core for Cell Technology, School of Medicine and Life Sciences, Pontifícia Universidade Catolica do Parana, Curitiba, PR, 80215-901, Brazil
| | - Carmen Lúcia Kuniyoshi Rebelatto
- Core for Cell Technology, School of Medicine and Life Sciences, Pontifícia Universidade Catolica do Parana, Curitiba, PR, 80215-901, Brazil
| | - Debora Regina Daga
- Core for Cell Technology, School of Medicine and Life Sciences, Pontifícia Universidade Catolica do Parana, Curitiba, PR, 80215-901, Brazil
| | - Wellingson Silva Paiva
- Departamento de Neurologia, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, SP, 01246-903, Brazil
| | - Gerson Chadi
- Laboratorio de Neurologia Translacional, Departamento de Neurologia, Hospital das Clinicas, Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, SP, 01246-903, Brazil.
- Departamento de Neurologia, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, SP, 01246-903, Brazil.
| |
Collapse
|
4
|
Christensen A, McGill CJ, Qian W, Pike CJ. Effects of obesogenic diet and 17β-estradiol in female mice with APOE 3/3, 3/4, and 4/4 genotypes. Front Aging Neurosci 2024; 16:1415072. [PMID: 39347015 PMCID: PMC11427389 DOI: 10.3389/fnagi.2024.1415072] [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: 04/09/2024] [Accepted: 08/26/2024] [Indexed: 10/01/2024] Open
Abstract
The main genetic risk factor for Alzheimer's disease (AD) is the apolipoprotein E ε4 allele (APOE4). AD risk associated with APOE4 disproportionately affects women. Furthermore, human and rodent studies indicate that the cognitive deficits associated with APOE4 are greater in females. One modifiable AD risk factor is obesity during middle age. Given that approximately two-thirds of US adults are overweight, it is important to understand how obesity affects AD risk, how it interacts with APOE4, and the extent to which its detrimental effects can be mitigated with therapeutics. One intervention study for women is estrogen-based hormone therapy, which can exert numerous health benefits when administered in early middle age. No experimental studies have examined the interactions among APOE4, obesity, and hormone therapy in aging females. To begin to explore these issues, we considered how obesity outcomes are affected by treatment with estradiol at the onset of middle age in female mice with human APOE3 and APOE4. Furthermore, to explore how gene dosage affects outcomes, we compared mice homozygous for APOE3 (3/3) and homozygous (4/4) or hemizygous (3/4) for APOE4. Mice were examined over a 4-month period that spans the transition into reproductive senescence, a normal age-related change that models many aspects of human perimenopause. Beginning at 5 months of age, mice were maintained on a control diet (10% fat) or high-fat diet (HFD; 60% fat). After 8 weeks, by which time obesity was present in all HFD groups, mice were implanted with an estradiol or vehicle capsule that was maintained for the final 8 weeks. Animals were assessed on a range of metabolic and neural measures. Overall, APOE4 was associated with poorer metabolic function and cognitive performance. However, an obesogenic diet induced relatively greater impairments in metabolic function and cognitive performance in APOE3/3 mice. Estradiol treatment improved metabolic and cognitive outcomes across all HFD groups, with APOE4/4 generally exhibiting the greatest benefit. APOE3/4 mice were intermediate to the homozygous genotypes on many measures but also exhibited unique profiles. Together, these findings highlight the importance of the APOE genotype as a modulator of the risks associated with obesity and the beneficial outcomes of estradiol.
Collapse
Affiliation(s)
| | | | | | - Christian J. Pike
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
5
|
Chen F, Zhao J, Meng F, He F, Ni J, Fu Y. The vascular contribution of apolipoprotein E to Alzheimer's disease. Brain 2024; 147:2946-2965. [PMID: 38748848 DOI: 10.1093/brain/awae156] [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: 12/24/2023] [Revised: 03/23/2024] [Accepted: 04/21/2024] [Indexed: 09/04/2024] Open
Abstract
Alzheimer's disease, the most prevalent form of dementia, imposes a substantial societal burden. The persistent inadequacy of disease-modifying drugs targeting amyloid plaques and neurofibrillary tangles suggests the contribution of alternative pathogenic mechanisms. A frequently overlooked aspect is cerebrovascular dysfunction, which may manifest early in the progression of Alzheimer's disease pathology. Mounting evidence underscores the pivotal role of the apolipoprotein E gene, particularly the apolipoprotein ε4 allele as the strongest genetic risk factor for late-onset Alzheimer's disease, in the cerebrovascular pathology associated with Alzheimer's disease. In this review, we examine the evidence elucidating the cerebrovascular impact of both central and peripheral apolipoprotein E on the pathogenesis of Alzheimer's disease. We present a novel three-hit hypothesis, outlining potential mechanisms that shed light on the intricate relationship among different pathogenic events. Finally, we discuss prospective therapeutics targeting the cerebrovascular pathology associated with apolipoprotein E and explore their implications for future research endeavours.
Collapse
Affiliation(s)
- Feng Chen
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jing Zhao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, China
| | - Fanxia Meng
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Fangping He
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jie Ni
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yuan Fu
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| |
Collapse
|
6
|
Burton EA, Argenziano M, Cook K, Ridler M, Lu S, Su C, Manduchi E, Littleton SH, Leonard ME, Hodge KM, Wang LS, Schellenberg GD, Johnson ME, Pahl MC, Pippin JA, Wells AD, Anderson SA, Brown CD, Grant SF, Chesi A. Variant-to-function mapping of late-onset Alzheimer's disease GWAS signals in human microglial cell models implicates RTFDC1 at the CASS4 locus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.22.609230. [PMID: 39229212 PMCID: PMC11370593 DOI: 10.1101/2024.08.22.609230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Late-onset Alzheimer's disease (LOAD) research has principally focused on neurons over the years due to their known role in the production of amyloid beta plaques and neurofibrillary tangles. In contrast, recent genomic studies of LOAD have implicated microglia as culprits of the prolonged inflammation exacerbating the neurodegeneration observed in patient brains. Indeed, recent LOAD genome-wide association studies (GWAS) have reported multiple loci near genes related to microglial function, including TREM2, ABI3, and CR1. However, GWAS alone cannot pinpoint underlying causal variants or effector genes at such loci, as most signals reside in non-coding regions of the genome and could presumably confer their influence frequently via long-range regulatory interactions. We elected to carry out a combination of ATAC-seq and high-resolution promoter-focused Capture-C in two human microglial cell models (iPSC-derived microglia and HMC3) in order to physically map interactions between LOAD GWAS-implicated candidate causal variants and their corresponding putative effector genes. Notably, we observed consistent evidence that rs6024870 at the GWAS CASS4 locus contacted the promoter of nearby gene, RTFDC1. We subsequently observed a directionallly consistent decrease in RTFDC1 expression with the the protective minor A allele of rs6024870 via both luciferase assays in HMC3 cells and expression studies in primary human microglia. Through CRISPR-Cas9-mediated deletion of the putative regulatory region harboring rs6024870 in HMC3 cells, we observed increased pro-inflammatory cytokine secretion and decreased DNA double strand break repair related, at least in part, to RTFDC1 expression levels. Our variant-to-function approach therefore reveals that the rs6024870-harboring regulatory element at the LOAD 'CASS4' GWAS locus influences both microglial inflammatory capacity and DNA damage resolution, along with cumulative evidence implicating RTFDC1 as a novel candidate effector gene.
Collapse
Affiliation(s)
- Elizabeth A. Burton
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- CAMB Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mariana Argenziano
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kieona Cook
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Molly Ridler
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sumei Lu
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Chun Su
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Elisabetta Manduchi
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sheridan H. Littleton
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- CAMB Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michelle E. Leonard
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Li-San Wang
- Department of Pathology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gerard D. Schellenberg
- Department of Pathology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Matthew E. Johnson
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Matthew C. Pahl
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - James A. Pippin
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Andrew D. Wells
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stewart A. Anderson
- Department of Child and Adolescent Psychiatry and Behavioral Services, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher D. Brown
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Struan F.A. Grant
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division Endocrinology and Diabetes, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alessandra Chesi
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
7
|
Moon HJ, Luo Y, Chugh D, Zhao L. Human apolipoprotein E glycosylation and sialylation: from structure to function. Front Mol Neurosci 2024; 17:1399965. [PMID: 39169951 PMCID: PMC11335735 DOI: 10.3389/fnmol.2024.1399965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/28/2024] [Indexed: 08/23/2024] Open
Abstract
Human apolipoprotein E (ApoE) was first identified as a polymorphic gene in the 1970s; however, the genetic association of ApoE genotypes with late-onset sporadic Alzheimer's disease (sAD) was only discovered 20 years later. Since then, intensive research has been undertaken to understand the molecular effects of ApoE in the development of sAD. Despite three decades' worth of effort and over 10,000 papers published, the greatest mystery in the ApoE field remains: human ApoE isoforms differ by only one or two amino acid residues; what is responsible for their significantly distinct roles in the etiology of sAD, with ApoE4 conferring the greatest genetic risk for sAD whereas ApoE2 providing exceptional neuroprotection against sAD. Emerging research starts to point to a novel and compelling hypothesis that the sialoglycans posttranslationally appended to human ApoE may serve as a critical structural modifier that alters the biology of ApoE, leading to the opposing impacts of ApoE isoforms on sAD and likely in the peripheral systems as well. ApoE has been shown to be posttranslationally glycosylated in a species-, tissue-, and cell-specific manner. Human ApoE, particularly in brain tissue and cerebrospinal fluid (CSF), is highly glycosylated, and the glycan chains are exclusively attached via an O-linkage to serine or threonine residues. Moreover, studies have indicated that human ApoE glycans undergo sialic acid modification or sialylation, a structural alteration found to be more prominent in ApoE derived from the brain and CSF than plasma. However, whether the sialylation modification of human ApoE has a biological role is largely unexplored. Our group recently first reported that the three major isoforms of human ApoE in the brain undergo varying degrees of sialylation, with ApoE2 exhibiting the most abundant sialic acid modification, whereas ApoE4 is the least sialylated. Our findings further indicate that the sialic acid moiety on human ApoE glycans may serve as a critical modulator of the interaction of ApoE with amyloid β (Aβ) and downstream Aβ pathogenesis, a prominent pathologic feature in AD. In this review, we seek to provide a comprehensive summary of this exciting and rapidly evolving area of ApoE research, including the current state of knowledge and opportunities for future exploration.
Collapse
Affiliation(s)
- Hee-Jung Moon
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
| | - Yan Luo
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
| | - Diksha Chugh
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
| | - Liqin Zhao
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
- Neuroscience Graduate Program, University of Kansas, Lawrence, KS, United States
| |
Collapse
|
8
|
Mdivnishvili M, Mdinaradze D, Virkovi K, Gogashvili G. A Case Report of Long COVID or Post-COVID-19 Symptoms and Characteristics. Cureus 2024; 16:e63876. [PMID: 39099965 PMCID: PMC11298016 DOI: 10.7759/cureus.63876] [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] [Accepted: 07/04/2024] [Indexed: 08/06/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has led to a global health crisis. Long COVID refers to a debilitating condition characterized by severe symptoms that may arise after the initial acute phase of COVID-19. Significant attention has been directed toward the acute phase of the respiratory system while overshadowing the understanding and management of long-term complications, often referred to as "long COVID." This case focuses on a 19-year-old female who experienced the multisystemic manifestation of COVID-19 syndrome several months after the initial infection, spanning cardiovascular, respiratory, endocrine, central nervous system, and multi-skeletal domains. This study aims to describe the patient's experience and recovery process with a specific emphasis on the long COVID experience.
Collapse
Affiliation(s)
| | | | - Ketino Virkovi
- Medicine, Caucasus's International University, Tbilisi, GEO
| | - George Gogashvili
- Emergency Department, Simon Khechinashvili University Clinic, Tbilisi, GEO
| |
Collapse
|
9
|
Alpers CE. Seeing through the density of dense deposit disease. Kidney Int 2024; 105:929-931. [PMID: 38642992 DOI: 10.1016/j.kint.2024.02.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 02/28/2024] [Indexed: 04/22/2024]
Abstract
Little is known about what constitutes the dense deposits of dense deposit disease (DDD), apart from components of the complement pathway. This study presents the novel finding that large accumulations of apolipoprotein E are present in the deposits of DDD, as revealed by mass spectroscopy and confirmed by both confocal microscopy and immunohistochemistry. The findings suggest a new modality for diagnosis of DDD and introduce potential new mechanisms for understanding DDD pathophysiology.
Collapse
Affiliation(s)
- Charles E Alpers
- Department of Laboratory Medicine and Pathology, University of Washington Medical Center, Seattle, Washington, USA.
| |
Collapse
|
10
|
Krishnamurthy HK, Rajavelu I, Reddy S, Pereira M, Jayaraman V, Krishna K, Song Q, Wang T, Bei K, Rajasekaran JJ. Association of Apolipoprotein E (APOE) Polymorphisms With Serological Lipid and Inflammatory Markers. Cureus 2024; 16:e60721. [PMID: 38903305 PMCID: PMC11187349 DOI: 10.7759/cureus.60721] [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] [Accepted: 05/13/2024] [Indexed: 06/22/2024] Open
Abstract
Background The study aims to assess the association of apolipoprotein E (APOE) gene polymorphisms with serological lipid and inflammatory markers to determine their potential role in predicting the risk of cardiovascular diseases (CVDs) and Alzheimer's disease (AD). Methodology A total of 915 individuals underwent testing for lipid and inflammatory biomarkers at Vibrant America Clinical Laboratory. Clinical data, blood lipid and inflammatory profiles, and APOE genotyping were analyzed using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). Results Compared to the E3/E3 genotype, individuals with E2/E3 genotypes showed higher levels of high-density lipoprotein (HDL), triglycerides, apolipoprotein A (APOA), high-sensitivity C-reactive protein (hs-CRP), and myeloperoxidase (MPO). E2/E4 genotype carriers had higher levels of HDL, triglycerides, Lp(a), and N-terminal pro b-type natriuretic peptide (BNPNT). E3/E4 genotypes were associated with elevated levels of total cholesterol, LDL, Lp(a), hs-CRP, small-density low-density lipoprotein (SDLDL), oxidized LDL (OXLDL), MPO, LDL-CAL, PLAC, and APOB. The E4/E4 group displayed higher concentrations of total cholesterol, LDL, APOB, Lp(a), hs-CRP, SDLDL, OXLDL, MPO, LDLCAL, and PLAC compared to E3/E3 carriers. These findings highlight the potential atherogenic effect of the ε4 allele and the protective effect of the ε2 allele based on lipid and inflammatory marker profiles. Conclusions This study provides strong evidence linking APOE gene polymorphism to abnormal serum lipid and inflammatory profiles. Individuals carrying the ε4 alleles exhibited dysregulated lipid metabolism and abnormal inflammatory markers, increasing their risk of CVD and AD. Early detection and prompt diagnosis are crucial for implementing therapeutic, dietary, and lifestyle interventions to mitigate risks and prevent or delay lipid and inflammation-related disorders.
Collapse
Affiliation(s)
| | | | | | | | | | - Karthik Krishna
- Research & Development, Vibrant Sciences LLC, San Carlos, USA
| | - Qi Song
- Data Acquisition and Analysis, Vibrant America LLC, San Carlos, USA
| | - Tianhao Wang
- Data Acquisition and Analysis, Vibrant Sciences LLC, San Carlos, USA
| | - Kang Bei
- Data Acquisition and Analysis, Vibrant Sciences LLC, San Carlos, USA
| | | |
Collapse
|
11
|
McGill CJ, Christensen A, Qian W, Thorwald MA, Lugo JG, Namvari S, White OS, Finch CE, Benayoun BA, Pike CJ. Protection against APOE4 -associated aging phenotypes with the longevity-promoting intervention 17α-estradiol in male mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.12.584678. [PMID: 38559059 PMCID: PMC10980056 DOI: 10.1101/2024.03.12.584678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The apolipoprotein ε4 allele ( APOE4 ) is associated with decreased longevity, increased vulnerability to age-related declines, and disorders across multiple systems. Interventions that promote healthspan and lifespan represent a promising strategy to attenuate the development of APOE4 -associated aging phenotypes. Here we studied the ability of the longevity-promoting intervention 17α-estradiol (17αE2) to protect against age-related impairments in APOE4 versus the predominant APOE3 genotype using early middle-aged mice with knock-in of human APOE alleles. Beginning at age 10 months, male APOE3 or APOE4 mice were treated for 20 weeks with 17αE2 or vehicle then compared for indices of aging phenotypes body-wide. Across peripheral and neural measures, APOE4 was associated with poorer outcomes. Notably, 17αE2 treatment improved outcomes in a genotype-dependent manner favoring APOE4 mice. These data demonstrate a positive APOE4 bias in 17αE2-mediated healthspan actions, suggesting that longevity-promoting interventions may be useful in mitigating deleterious age-related risks associated with APOE4 genotype.
Collapse
|
12
|
Nepomuceno M, Monllor P, Cardells MJ, Ftara A, Magallon M, Dasí F, Badia MC, Viña J, Lloret A. Redox-associated changes in healthy individuals at risk of Alzheimer's disease. A ten-year follow-up study. Free Radic Biol Med 2024; 215:56-63. [PMID: 38417685 DOI: 10.1016/j.freeradbiomed.2024.02.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 02/16/2024] [Accepted: 02/26/2024] [Indexed: 03/01/2024]
Abstract
Carrying an allele 4 of the apolipoprotein E (ApoE) is the best-established genetic risk factor to develop Alzheimer's disease (AD). Fifty percent of ApoE4/4 individuals develop the disease at 70 years of age. ApoE3/4 carriers have a lower risk of developing the disease, still 50% of them suffer AD at around 80 years. In a previous study we showed that healthy young individuals, who had a parent with AD and were carriers of at least one ApoE4 allele displayed reductive stress. This was evidenced as a decrease in oxidative markers, such as oxidized glutathione, p-p38, and NADP+/NADPH ratio, and an increase of antioxidant enzymes, such as glutathione peroxidase (Gpx1) and both the catalytic and regulatory subunits of glutamyl-cysteinyl (GCLM and GCLC). Moreover, we found an increase in stress-related proteins involved in tau physiopathology. Now, 10 years later, we have conducted a follow-up study measuring the same parameters in the same cohort. Our results show that reductive stress has reversed, as we could now observe an increase in lipid peroxidation and in the oxidation of glutathione along with a decrease in the expression of Gpx1 and SOD1 antioxidant enzymes in ApoE4 carriers. Furthermore, we found an increase in plasma levels of IL1β levels and in PKR (eukaryotic translation initiation factor 2 alpha kinase 2) gene expression in isolated lymphocytes. Altogether, our results suggest that, in the continuum of Alzheimer's disease, people at risk of developing the disease go through different redox phases, from stablished reductive stress to oxidative stress.
Collapse
Affiliation(s)
- Mariana Nepomuceno
- Department of Physiology, Faculty of Medicine, CIBERFES, Fundación Investigación Hospital Clínico Universitario/INCLIVA, University of Valencia, Valencia, Spain
| | - Paloma Monllor
- Department of Physiology, Faculty of Medicine, CIBERFES, Fundación Investigación Hospital Clínico Universitario/INCLIVA, University of Valencia, Valencia, Spain; Internal Medicine Department, University Hospital of La Plana, Vila-Real, Spain
| | - Maria Jose Cardells
- Department of Physiology, Faculty of Medicine, CIBERFES, Fundación Investigación Hospital Clínico Universitario/INCLIVA, University of Valencia, Valencia, Spain
| | - Artemis Ftara
- Department of Physiology, Faculty of Medicine, CIBERFES, Fundación Investigación Hospital Clínico Universitario/INCLIVA, University of Valencia, Valencia, Spain
| | - Maria Magallon
- Department of Physiology, Faculty of Medicine, CIBERFES, Fundación Investigación Hospital Clínico Universitario/INCLIVA, University of Valencia, Valencia, Spain
| | - Francisco Dasí
- Department of Physiology, Faculty of Medicine, CIBERFES, Fundación Investigación Hospital Clínico Universitario/INCLIVA, University of Valencia, Valencia, Spain
| | | | - Jose Viña
- Department of Physiology, Faculty of Medicine, CIBERFES, Fundación Investigación Hospital Clínico Universitario/INCLIVA, University of Valencia, Valencia, Spain.
| | - Ana Lloret
- Department of Physiology, Faculty of Medicine, CIBERFES, Fundación Investigación Hospital Clínico Universitario/INCLIVA, University of Valencia, Valencia, Spain.
| |
Collapse
|
13
|
Giil LM, Hanseth S, Bojovic O, Nordrehaug JE, Ueland PM, Meyer K, Tell GS. The inverse association between the apolipoprotein E ε4 allele and C-reactive protein levels is stronger in persons with obesity and diabetes. Scand J Immunol 2024; 99:e13323. [PMID: 37801334 DOI: 10.1111/sji.13323] [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: 07/17/2022] [Revised: 08/09/2023] [Accepted: 08/10/2023] [Indexed: 10/07/2023]
Abstract
BACKGROUND C-reactive protein (CRP) is lower in patients who carry the apolipoprotein E epsilon 4 allele variant (APOEε4) of the APOE gene. This could however be explained by other factors observed in APOEε4 carriers, such as lower body mass index (BMI), possibly less diabetes and more use of statins, all associated with CRP concentrations. OBJECTIVES To assess the association between CRP and APOEε4 stratified by BMI, statin use and diabetes. METHODS We included 2700 community-dwelling older adults from the Hordaland health study with genotyping of the APOE gene by a one-step polymerase chain reaction and CRP measured using immuno-MALDI-TOF MS. Differences in CRP concentrations by APOE (ε4 vs no ε4) were assessed using the Mann-Whitney U tests, also stratified by statin use, diabetes and BMI categories. Finally, we performed linear regression with log (CRP) as the outcome and APOEε4 together with statin use, diabetes, BMI and their respective interactions. RESULTS CRP was higher in APOEε4 carriers irrespective of BMI, diabetes and statin use. In APOEε4 non-carriers, CRP was elevated with diabetes and obesity as expected. However, this was attenuated or even reversed in APOEε4 carriers. Such differences were not observed for statin use. CONCLUSIONS Statin use, obesity or diabetes did not confound the known association between the APOEε4 allele and lower CRP. Our data suggest that CRP is less responsive to inflammatory cues involved in diabetes and obesity in APOEε4 carriers. Epidemiological studies should take note of these relationships, as CRP, APOEε4, diabetes and obesity are both linked to neurodegenerative and cardiovascular disease.
Collapse
Affiliation(s)
- Lasse Melvaer Giil
- Neuro-SysMed, Department of Internal Medicine, Haraldsplass Deaconess Hospital, Bergen, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Silja Hanseth
- Neuro-SysMed, Department of Internal Medicine, Haraldsplass Deaconess Hospital, Bergen, Norway
| | - Ognjen Bojovic
- Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | | | | | | | - Grethe S Tell
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
| |
Collapse
|
14
|
Nickhah Klashami Z, Yaghoobi A, Panahi N, Amoli MM. Association of the APOE gene variants with depression in type 2 diabetes. J Diabetes Metab Disord 2023; 22:1481-1487. [PMID: 37975117 PMCID: PMC10638337 DOI: 10.1007/s40200-023-01271-9] [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: 07/06/2023] [Accepted: 07/27/2023] [Indexed: 11/19/2023]
Abstract
Background The risk of depression among patients with diabetes is higher than the general population. The exact mechanisms linking these two diseases are mostly unknown. Energy metabolism disorders seem to be a shared pathway. One of the key genes playing important roles in energy metabolism-related pathways is the APOE gene. We aimed to investigate the association of the APOE gene variants with depression among Iranian patients with type 2 diabetes (T2DM). Methods Three APOE gene alleles and genotypes frequencies (E2, E3, E4) were determined in 244 patients with T2DM (114 with depression and 130 without depression) using the high-resolution melting (HRM) method on the genomic DNA extracted from the patient's peripheral blood. Results Apoe4 allele frequency was significantly higher in T2DM patients without depression compared with those with depression (11.9 vs. 2.2%, p-value < 0.0001 and p-value = 0.001, respectively). Conversely, the wild allele apoe3 frequency was significantly higher in T2DM patients with depression (86% vs., 69%, p-value < 0.0001). Apoe4 carrier status was associated with decreased risk of depression in patients with T2DM [OR: 0.19 (0.07-0.53)]. Conclusion Our results showed that the apoe4 allele and apoe4 carrier status significantly reduced the risk of depression among patients with T2DM. Further studies are needed to unravel the complex role of the APOE gene in depression among patients with diabetes.
Collapse
Affiliation(s)
- Zeynab Nickhah Klashami
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Arash Yaghoobi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Nekoo Panahi
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa M. Amoli
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Liemisa B, Newbury SF, Novy MJ, Pasato JA, Morales-Corraliza J, Peng KY, Mathews PM. Brain apolipoprotein E levels in mice challenged by a Western diet increase in an allele-dependent manner. AGING BRAIN 2023; 4:100102. [PMID: 38058491 PMCID: PMC10696459 DOI: 10.1016/j.nbas.2023.100102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/05/2023] [Accepted: 11/17/2023] [Indexed: 12/08/2023] Open
Abstract
Human apolipoprotein E (APOE) is the greatest determinant of genetic risk for memory deficits and Alzheimer's disease (AD). While APOE4 drives memory loss and high AD risk, APOE2 leads to healthy brain aging and reduced AD risk compared to the common APOE3 variant. We examined brain APOE protein levels in humanized mice homozygous for these alleles and found baseline levels to be age- and isoform-dependent: APOE2 levels were greater than APOE3, which were greater than APOE4. Despite the understanding that APOE lipoparticles do not traverse the blood-brain barrier, we show that brain APOE levels are responsive to dietary fat intake. Challenging mice for 6 months on a Western diet high in fat and cholesterol increased APOE protein levels in an allele-dependent fashion with a much greater increase within blood plasma than within the brain. In the brain, APOE2 levels responded most to the Western diet challenge, increasing by 20 % to 30 %. While increased lipoparticles are generally deleterious in the periphery, we propose that higher brain APOE2 levels may represent a readily available pool of beneficial lipid particles for neurons.
Collapse
Affiliation(s)
- Braison Liemisa
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
| | - Samantha F. Newbury
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
| | - Mariah J. Novy
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
| | - Jonathan A. Pasato
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
| | - Jose Morales-Corraliza
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Katherine Y. Peng
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Paul M. Mathews
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY 10016, USA
- NYU Neuroscience Institute, New York University Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
16
|
Balu D, Valencia-Olvera AC, Islam Z, Mielczarek C, Hansen A, Perez Ramos TM, York J, LaDu MJ, Tai LM. APOE genotype and sex modulate Alzheimer's disease pathology in aged EFAD transgenic mice. Front Aging Neurosci 2023; 15:1279343. [PMID: 38020764 PMCID: PMC10644540 DOI: 10.3389/fnagi.2023.1279343] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 10/10/2023] [Indexed: 12/01/2023] Open
Abstract
Increasing evidence supports that age, APOE and sex interact to modulate Alzheimer's disease (AD) risk, however the underlying pathways are unclear. One way that AD risk factors may modulate cognition is by impacting amyloid beta (Aβ) accumulation as plaques, and/or neuroinflammation Therefore, the goal of the present study was to evaluate the extent to which age, APOE and sex modulate Aβ pathology, neuroinflammation and behavior in vivo. To achieve this goal, we utilized the EFAD mice, which express human APOE3 or APOE4 and have five familial AD mutations (FAD) that result in Aβ42 overproduction. We assessed Aβ levels, reactive glia and Morris water maze performance in 6-, 10-, 14-, and 18-month-old EFAD mice. Female APOE4 mice had the highest Aβ deposition, fibrillar amyloid deposits and neuroinflammation as well as earlier behavior deficits. Interestingly, we found that female APOE3 mice and male APOE4 mice had similar levels of pathology. Collectively our data support that the combination of APOE4 and female sex is the most detrimental combination for AD, and that at older ages, female sex may be equivalent to APOE4 genotype.
Collapse
Affiliation(s)
- Deebika Balu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Ana C. Valencia-Olvera
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Zarak Islam
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
- University of Illinois College of Medicine, Chicago, IL, United States
| | - Clare Mielczarek
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Allison Hansen
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
- University of Illinois College of Medicine, Peoria, IL, United States
| | - Tamara M. Perez Ramos
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
- School of Medicine, St. George’s University, St. George’s, Grenada
| | - Jason York
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Mary Jo LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Leon M. Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
17
|
Balu D, Valencia-Olvera AC, Nguyen A, Patnam M, York J, Peri F, Neumann F, LaDu MJ, Tai LM. A small-molecule TLR4 antagonist reduced neuroinflammation in female E4FAD mice. Alzheimers Res Ther 2023; 15:181. [PMID: 37858252 PMCID: PMC10585767 DOI: 10.1186/s13195-023-01330-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 10/10/2023] [Indexed: 10/21/2023]
Abstract
BACKGROUND APOE genotype is the greatest genetic risk factor for sporadic Alzheimer's disease (AD). APOE4 increases AD risk up to 12-fold compared to APOE3, an effect that is greater in females. Evidence suggests that one-way APOE could modulate AD risk and progression through neuroinflammation. Indeed, APOE4 is associated with higher glial activation and cytokine levels in AD patients and mice. Therefore, identifying pathways that contribute to APOE4-associated neuroinflammation is an important approach for understanding and treating AD. Human and in vivo evidence suggests that TLR4, one of the key receptors involved in the innate immune system, could be involved in APOE-modulated neuroinflammation. Consistent with that idea, we previously demonstrated that the TLR4 antagonist IAXO-101 can reduce LPS- and Aβ-induced cytokine secretion in APOE4 glial cultures. Therefore, the goal of this study was to advance these findings and determine whether IAXO-101 can modulate neuroinflammation, Aβ pathology, and behavior in mice that express APOE4. METHODS We used mice that express five familial AD mutations and human APOE3 (E3FAD) or APOE4 (E4FAD). Female and male E4FAD mice and female E3FAD mice were treated with vehicle or IAXO-101 in two treatment paradigms: prevention from 4 to 6 months of age or reversal from 6 to 7 months of age. Learning and memory were assessed by modified Morris water maze. Aβ deposition, fibrillar amyloid deposition, astrogliosis, and microgliosis were assessed by immunohistochemistry. Soluble levels of Aβ and apoE, insoluble levels of apoE and Aβ, and IL-1β were measured by ELISA. RESULTS IAXO-101 treatment resulted in lower Iba-1 coverage, lower number of reactive microglia, and improved memory in female E4FAD mice in both prevention and reversal paradigms. IAXO-101-treated male E4FAD mice also had lower Iba-1 coverage and reactivity in the RVS paradigm, but there was no effect on behavior. There was also no effect of IAXO-101 treatment on neuroinflammation and behavior in female E3FAD mice. CONCLUSION Our data supports that TLR4 is a potential mechanistic therapeutic target for modulating neuroinflammation and cognition in APOE4 females.
Collapse
Affiliation(s)
- Deebika Balu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Ana C Valencia-Olvera
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Austin Nguyen
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Mehul Patnam
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Jason York
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Francesco Peri
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | | | - Mary Jo LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Leon M Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
18
|
Eren N, Gerike S, Üsekes B, Peters O, Cosma NC, Hellmann-Regen J. Effects of autologous serum on TREM2 and APOE in a personalized monocyte-derived macrophage assay of late-onset Alzheimer's patients. Immun Ageing 2023; 20:52. [PMID: 37833781 PMCID: PMC10576307 DOI: 10.1186/s12979-023-00376-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 09/14/2023] [Indexed: 10/15/2023]
Abstract
BACKGROUND Age-associated deterioration of the immune system contributes to a chronic low-grade inflammatory state known as "inflammaging" and is implicated in the pathogenesis of late-onset Alzheimer's disease (LOAD). Whether changes in the tissue environment caused by circulatory factors associated with aging may alter the innate immune response is unknown. Monocyte-derived macrophages (Mo-MФs) infiltrating the brain alongside microglia are postulated to play a modulatory role in LOAD and both express triggering receptor expressed on myeloid cells 2 (TREM2). Apolipoprotein E (APOE) acts as a ligand for TREM2, and their role in amyloid beta (Aβ) clearance highlights their importance in LOAD. However, the influence of the patient's own milieu (autologous serum) on the synthesis of TREM2 and APOE in infiltrating macrophages remains unknown. OBJECTIVES To functionally assess patient-specific TREM2 and APOE synthesis, we designed a personalized assay based on Mo-MФs using monocytes from LOAD patients and matched controls (CO). We assessed the influence of each participant's own milieu, by examining the effect of short- (1 day) and long- (10 days) term differentiation of the cells in the presence of the donor´s autologous serum (AS) into M1-, M2- or M0-macrophages. Additionally, sex differences and Aβ-uptake ability in short- and long-term differentiated Mo-MФs were assessed. RESULTS We showed a time-dependent increase in TREM2 and APOE protein levels in LOAD- and CO-derived cells. While AS did not differentially modulate TREM2 compared to standard fetal calf serum (FCS), AS decreased APOE levels in M2 macrophages but increased levels in M1 macrophages. Interestingly, higher levels of TREM2 and lower levels of APOE were detected in female- than in male- LOAD patients. Finally, we report decreased Aβ-uptake in long-term differentiated CO- and LOAD-derived cells, particularly in APOEε4(+) carriers. CONCLUSIONS We demonstrate for the first time the suitability of a personalized Mo-MФ cell culture-based assay for studying functional TREM2 and APOE synthesis in a patient's own aged milieu. Our strategy may thus provide a useful tool for future research on diagnostic and therapeutic aspects of personalized medicine.
Collapse
Affiliation(s)
- Neriman Eren
- Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Section Clinical Neurobiology, Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany.
| | - Susanna Gerike
- Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Section Clinical Neurobiology, Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Berk Üsekes
- Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Section Clinical Neurobiology, Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Oliver Peters
- Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Section Clinical Neurobiology, Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
- German Center for Mental Health (DZPG) Partner Site Berlin, Berlin, Germany
| | - Nicoleta-Carmen Cosma
- Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Section Clinical Neurobiology, Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
- BIH Biomedical Innovation Academy, BIH Charité Clinician Scientist Program, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, Berlin, 10117, Germany
| | - Julian Hellmann-Regen
- Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Section Clinical Neurobiology, Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
- BIH Biomedical Innovation Academy, BIH Charité Clinician Scientist Program, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, Berlin, 10117, Germany
| |
Collapse
|
19
|
Marottoli FM, Zhang H, Flores-Barrera E, Artur de la Villarmois E, Damen FC, Miguelez Fernández AM, Blesson HV, Chaudhary R, Nguyen AL, Nwokeji AE, Talati R, John AS, Madadakere K, Lutz SE, Cai K, Tseng KY, Tai LM. Endothelial Cell APOE3 Regulates Neurovascular, Neuronal, and Behavioral Function. Arterioscler Thromb Vasc Biol 2023; 43:1952-1966. [PMID: 37650329 PMCID: PMC10521805 DOI: 10.1161/atvbaha.123.319816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 08/17/2023] [Indexed: 09/01/2023]
Abstract
BACKGROUND Specialized brain endothelial cells and human APOE3 are independently important for neurovascular function, yet whether APOE3 expression by endothelial cells contributes to brain function is currently unknown. In the present study, we determined whether the loss of endothelial cell APOE3 impacts brain vascular and neural function. METHODS We developed APOE3fl/fl/Cdh5(PAC)-CreERT2+/- (APOE3Cre+/-) and APOE3fl/fl/Cdh5(PAC)-CreERT2-/- (APOE3Cre-/-, control) mice and induced endothelial cell APOE3 knockdown with tamoxifen at ≈4 to 5 weeks of age. Neurovascular and neuronal function were evaluated by biochemistry, immunohistochemistry, behavioral testing, and electrophysiology at 9 months of age. RESULTS We found that the loss of endothelial APOE3 expression was sufficient to cause neurovascular dysfunction including higher permeability and lower vessel coverage in tandem with deficits in spatial memory and fear memory extinction and a disruption of cortical excitatory/inhibitory balance. CONCLUSIONS Our data collectively support the novel concept that endothelial APOE3 plays a critical role in the regulation of the neurovasculature, neural circuit function, and behavior.
Collapse
Affiliation(s)
- Felecia M. Marottoli
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Hui Zhang
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Eden Flores-Barrera
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Emilce Artur de la Villarmois
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | | | - Anabel M.M. Miguelez Fernández
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Hannah V. Blesson
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Rohan Chaudhary
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Anthony L. Nguyen
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Amanda E. Nwokeji
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Ruju Talati
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Ashwin S. John
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Kushi Madadakere
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Sarah E. Lutz
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Kejia Cai
- Radiology (F.C.D., K.C.), University of Illinois at Chicago
- Bioengineering (K.C.), University of Illinois at Chicago
| | - Kuei Y. Tseng
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Leon M. Tai
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| |
Collapse
|
20
|
Abyadeh M, Gupta V, Paulo JA, Sheriff S, Shadfar S, Fitzhenry M, Amirkhani A, Gupta V, Salekdeh GH, Haynes PA, Graham SL, Mirzaei M. Apolipoprotein ε in Brain and Retinal Neurodegenerative Diseases. Aging Dis 2023; 14:1311-1330. [PMID: 37199411 PMCID: PMC10389820 DOI: 10.14336/ad.2023.0312-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/12/2023] [Indexed: 05/19/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia that remains incurable and has become a major medical, social, and economic challenge worldwide. AD is characterized by pathological hallmarks of senile plaques (SP) and neurofibrillary tangles (NFTs) that damage the brain up to twenty years before a clinical diagnosis is made. Interestingly these pathological features have also been observed in retinal neurodegenerative diseases including age related macular degeneration (ARMD), glaucoma and diabetic retinopathy (DR). An association of AD with these diseases has been suggested in epidemiological studies and several common pathological events and risk factors have been identified between these diseases. The E4 allele of Apolipoprotein E (APOE) is a well-established genetic risk factor for late onset AD. The ApoE ε4 allele is also associated with retinal neurodegenerative diseases however in contrast to AD, it is considered protective in AMD, likewise ApoE E2 allele, which is a protective factor for AD, has been implicated as a risk factor for AMD and glaucoma. This review summarizes the evidence on the effects of ApoE in retinal neurodegenerative diseases and discusses the overlapping molecular pathways in AD. The involvement of ApoE in regulating amyloid beta (Aβ) and tau pathology, inflammation, vascular integrity, glucose metabolism and vascular endothelial growth factor (VEGF) signaling is also discussed.
Collapse
Affiliation(s)
| | - Vivek Gupta
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW 2109, Australia.
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA.
| | - Samran Sheriff
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW 2109, Australia.
| | - Sina Shadfar
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW 2109, Australia.
| | - Matthew Fitzhenry
- Australian Proteome Analysis Facility, Macquarie University, Macquarie Park, NSW 2113, Australia.
| | - Ardeshir Amirkhani
- Australian Proteome Analysis Facility, Macquarie University, Macquarie Park, NSW 2113, Australia.
| | - Veer Gupta
- School of Medicine, Deakin University, VIC, Australia.
| | - Ghasem H Salekdeh
- School of Natural Sciences, Macquarie University, Macquarie Park, NSW 2109, Australia.
| | - Paul A Haynes
- School of Natural Sciences, Macquarie University, Macquarie Park, NSW 2109, Australia.
| | - Stuart L Graham
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW 2109, Australia.
| | - Mehdi Mirzaei
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW 2109, Australia.
| |
Collapse
|
21
|
Paseban T, Alavi MS, Etemad L, Roohbakhsh A. The role of the ATP-Binding Cassette A1 (ABCA1) in neurological disorders: a mechanistic review. Expert Opin Ther Targets 2023; 27:531-552. [PMID: 37428709 DOI: 10.1080/14728222.2023.2235718] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 07/09/2023] [Indexed: 07/12/2023]
Abstract
INTRODUCTION Cholesterol homeostasis is critical for normal brain function. It is tightly controlled by various biological elements. ATP-binding cassette transporter A1 (ABCA1) is a membrane transporter that effluxes cholesterol from cells, particularly astrocytes, into the extracellular space. The recent studies pertaining to ABCA1's role in CNS disorders were included in this study. AREAS COVERED In this comprehensive literature review, preclinical and human studies showed that ABCA1 has a significant role in the following diseases or disorders: Alzheimer's disease, Parkinson's disease, Huntington's disease, multiple sclerosis, neuropathy, anxiety, depression, psychosis, epilepsy, stroke, and brain ischemia and trauma. EXPERT OPINION ABCA1 via modulating normal and aberrant brain functions such as apoptosis, phagocytosis, BBB leakage, neuroinflammation, amyloid β efflux, myelination, synaptogenesis, neurite outgrowth, and neurotransmission promotes beneficial effects in aforementioned diseases. ABCA1 is a key molecule in the CNS. By boosting its expression or function, some CNS disorders may be resolved. In preclinical studies, liver X receptor agonists have shown promise in treating CNS disorders via ABCA1 and apoE enhancement.
Collapse
Affiliation(s)
- Tahere Paseban
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohaddeseh Sadat Alavi
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Etemad
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Roohbakhsh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
22
|
Chen L, Zhao S, Wang Y, Niu X, Zhang B, Li X, Peng D. Genetic Insights into Obesity and Brain: Combine Mendelian Randomization Study and Gene Expression Analysis. Brain Sci 2023; 13:892. [PMID: 37371369 PMCID: PMC10295948 DOI: 10.3390/brainsci13060892] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 05/27/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
As a major public-health concern, obesity is imposing an increasing social burden around the world. The link between obesity and brain-health problems has been reported, but controversy remains. To investigate the relationship among obesity, brain-structure changes and diseases, a two-stage analysis was performed. At first, we used the Mendelian-randomization (MR) approach to identify the causal relationship between obesity and cerebral structure. Obesity-related data were retrieved from the Genetic Investigation of ANthropometric Traits (GIANT) consortium and the UK Biobank, whereas the cortical morphological data were from the Enhancing NeuroImaging Genetics through Meta-Analysis (ENIGMA) consortium. Further, we extracted region-specific expressed genes according to the Allen Human Brian Atlas (AHBA) and carried out a series of bioinformatics analyses to find the potential mechanism of obesity and diseases. In the univariable MR, a higher body mass index (BMI) or larger visceral adipose tissue (VAT) was associated with a smaller global cortical thickness (pBMI = 0.006, pVAT = 1.34 × 10-4). Regional associations were found between obesity and specific gyrus regions, mainly in the fusiform gyrus and inferior parietal gyrus. Multivariable MR results showed that a greater body fat percentage was linked to a smaller fusiform-gyrus thickness (p = 0.029) and precuneus surface area (p = 0.035). As for the gene analysis, region-related genes were enriched to several neurobiological processes, such as compound transport, neuropeptide-signaling pathway, and neuroactive ligand-receptor interaction. These genes contained a strong relationship with some neuropsychiatric diseases, such as Alzheimer's disease, epilepsy, and other disorders. Our results reveal a causal relationship between obesity and brain abnormalities and suggest a pathway from obesity to brain-structure abnormalities to neuropsychiatric diseases.
Collapse
Affiliation(s)
- Leian Chen
- Department of Neurology, China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100029, China
| | - Shaokun Zhao
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China
| | - Yuye Wang
- Department of Neurology, China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100029, China
| | - Xiaoqian Niu
- Department of Neurology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing 100029, China
| | - Bin Zhang
- Department of Neurology, China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100029, China
| | - Xin Li
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China
| | - Dantao Peng
- Department of Neurology, China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100029, China
- Department of Neurology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing 100029, China
| |
Collapse
|
23
|
Calderón-Garcidueñas L, Hernández-Luna J, Aiello-Mora M, Brito-Aguilar R, Evelson PA, Villarreal-Ríos R, Torres-Jardón R, Ayala A, Mukherjee PS. APOE Peripheral and Brain Impact: APOE4 Carriers Accelerate Their Alzheimer Continuum and Have a High Risk of Suicide in PM 2.5 Polluted Cities. Biomolecules 2023; 13:927. [PMID: 37371506 DOI: 10.3390/biom13060927] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/22/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
This Review emphasizes the impact of APOE4-the most significant genetic risk factor for Alzheimer's disease (AD)-on peripheral and neural effects starting in childhood. We discuss major mechanistic players associated with the APOE alleles' effects in humans to understand their impact from conception through all life stages and the importance of detrimental, synergistic environmental exposures. APOE4 influences AD pathogenesis, and exposure to fine particulate matter (PM2.5), manufactured nanoparticles (NPs), and ultrafine particles (UFPs) associated with combustion and friction processes appear to be major contributors to cerebrovascular dysfunction, neuroinflammation, and oxidative stress. In the context of outdoor and indoor PM pollution burden-as well as Fe, Ti, and Al alloys; Hg, Cu, Ca, Sn, and Si UFPs/NPs-in placenta and fetal brain tissues, urban APOE3 and APOE4 carriers are developing AD biological disease hallmarks (hyperphosphorylated-tau (P-tau) and amyloid beta 42 plaques (Aβ42)). Strikingly, for Metropolitan Mexico City (MMC) young residents ≤ 40 y, APOE4 carriers have 4.92 times higher suicide odds and 23.6 times higher odds of reaching Braak NFT V stage versus APOE4 non-carriers. The National Institute on Aging and Alzheimer's Association (NIA-AA) framework could serve to test the hypothesis that UFPs and NPs are key players for oxidative stress, neuroinflammation, protein aggregation and misfolding, faulty complex protein quality control, and early damage to cell membranes and organelles of neural and vascular cells. Noninvasive biomarkers indicative of the P-tau and Aβ42 abnormal protein deposits are needed across the disease continuum starting in childhood. Among the 21.8 million MMC residents, we have potentially 4 million APOE4 carriers at accelerated AD progression. These APOE4 individuals are prime candidates for early neuroprotective interventional trials. APOE4 is key in the development of AD evolving from childhood in highly polluted urban centers dominated by anthropogenic and industrial sources of pollution. APOE4 subjects are at higher early risk of AD development, and neuroprotection ought to be implemented. Effective reductions of PM2.5, UFP, and NP emissions from all sources are urgently needed. Alzheimer's Disease prevention ought to be at the core of the public health response and physicians-scientist minority research be supported.
Collapse
Affiliation(s)
- Lilian Calderón-Garcidueñas
- College of Health, The University of Montana, Missoula, MT 59812, USA
- Universidad del Valle de México, Mexico City 14370, Mexico
| | | | - Mario Aiello-Mora
- Otorrinolaryngology Department, Instituto Nacional de Cardiología, Mexico City 14080, Mexico
| | | | - Pablo A Evelson
- Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires C1113 AAD, Argentina
| | | | - Ricardo Torres-Jardón
- Instituto de Ciencias de la Atmósfera y Cambio Climático, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Alberto Ayala
- Sacramento Metropolitan Air Quality Management District, Sacramento, CA 95814, USA
- West Virginia University, Morgantown, WV 26506, USA
| | - Partha S Mukherjee
- Interdisciplinary Statistical Research Unit, Indian Statistical Institute, Kolkata 700108, India
| |
Collapse
|
24
|
Wu H, Xu J, Sun J, Duan J, Xiao J, Ren Q, Zhou P, Yan J, Li Y, Xiong X, Zeng E. APOE as potential biomarkers of moyamoya disease. Front Neurol 2023; 14:1156894. [PMID: 37228412 PMCID: PMC10203507 DOI: 10.3389/fneur.2023.1156894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 04/17/2023] [Indexed: 05/27/2023] Open
Abstract
Objective The mechanisms underpinning Moyamoya disease (MMD) remain unclear, and effective biomarkers remain unknown. The purpose of this study was to identify novel serum biomarkers of MMD. Methods Serum samples were collected from 23 patients with MMD and 30 healthy controls (HCs). Serum proteins were identified using tandem tandem-mass-tag (TMT) labeling combined with liquid chromatography-tandem mass spectrometry (LC-MS/MS). Differentially expressed proteins (DEPs) in the serum samples were identified using the SwissProt database. The DEPs were assessed using the Kyoto Encyclopedia of Genes and Genomes (KEGG) database, Gene Ontology (GO), and protein-protein interaction (PPI) networks, and hub genes were identified and visualized using Cytoscape software. Microarray datasets GSE157628, GSE189993, and GSE100488 from the Gene Expression Omnibus (GEO) database were collected. Differentially expressed genes (DEGs) and differentially expressed miRNAs (DE-miRNAs) were identified, and miRNA targets of DEGs were predicted using the miRWalk3.0 database. Serum apolipoprotein E (APOE) levels were compared in 33 MMD patients and 28 Moyamoya syndrome (MMS) patients to investigate the potential of APOE to be as an MMD biomarker. Results We identified 85 DEPs, of which 34 were up- and 51 down-regulated. Bioinformatics analysis showed that some DEPs were significantly enriched in cholesterol metabolism. A total of 1105 DEGs were identified in the GSE157628 dataset (842 up- and 263 down-regulated), whereas 1290 were identified in the GSE189993 dataset (200 up- and 1,090 down-regulated). The APOE only overlaps with the upregulated gene expression in Proteomic Profiling and in GEO databases. Functional enrichment analysis demonstrated that APOE was associated with cholesterol metabolism. Moreover, 149 miRNAs of APOE were predicted in the miRWalk3.0 database, and hsa-miR-718 was the only DE-miRNA overlap identified in MMD samples. Serum APOE levels were significantly higher in patients with MMD than in those without. The performance of APOE as an individual biomarker to diagnose MMD was remarkable. Conclusions We present the first description of the protein profile of patients with MMD. APOE was identified as a potential biomarker for MMD. Cholesterol metabolism was found to potentially be related to MMD, which may provide helpful diagnostic and therapeutic insights for MMD.
Collapse
Affiliation(s)
- Haibin Wu
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jiang Xu
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jiarong Sun
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jian Duan
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jinlin Xiao
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Quan Ren
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Pengfei Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jian Yan
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Youping Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaoxing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Erming Zeng
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
25
|
Zaib M, Malik MNH, Shabbir R, Mushtaq MN, Younis W, Jahan S, Ahmed I, Kharl HAA. Imine Derivatives of Benzoxazole Attenuate High-Fat Diet-Induced Hyperlipidemia by Modulation of Lipid-Regulating Genes. ACS OMEGA 2023; 8:15306-15317. [PMID: 37151544 PMCID: PMC10157695 DOI: 10.1021/acsomega.3c00443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 04/03/2023] [Indexed: 05/09/2023]
Abstract
Purpose: Hyperlipidemia being the prominent risk factor of cardiovascular diseases and side effects associated with the current lipid-lowering drugs have attracted the interest of scientists in the quest for new alternatives. In view of the diverse pharmacological potentials of benzoxazole (BZX) compounds, this study was designed to evaluate the antihyperlipidemic activity of imine derivatives of BZX in high-fat diet (HFD)-fed rats. Methods: Hyperlipidemia was induced in Sprague-Dawley rats by using HFD for 28 days. On the 28th day, blood samples were collected, and animals having serum triglycerides (TG) greater than 400 mg/dL and total cholesterol (TC) greater than 280 mg/dL were selected for further study. Hyperlipidemic rats were daily treated with either a vehicle or simvastatin (SIM; 20 mg/kg) or BZX compounds (10, 20, and 30 mg/kg), for 12 consecutive days. After the specified time duration, hyperlipidemic biomarkers were evaluated in the blood samples of sacrificed rats. Liver samples were collected for histopathological and mRNA analyses. Binding affinities of BZX derivatives with different targets were assessed by molecular docking. Results: The present study revealed that the BZX derivatives dose-dependently reduced the serum levels of TC, TG, low-density lipoprotein, and very low-density lipoprotein along with improvement in high-density lipoprotein levels. Similarly, all the compounds reduced HFD-induced alanine transaminase and aspartate aminotransferase levels except BZX-4. Histopathology of liver samples demonstrated mild to moderate fatty changes upon treatment with BZX-1, BZX-2, and BZX-4. The hepatic architecture of the BZX-3-treated samples was close to normal, and only mild inflammation was witnessed in these samples. Moreover, all the compounds significantly increased superoxide dismutase and glutathione levels, indicating their antioxidant potentials. Gene expression data showed that BZX-1 and BZX-3 reduced lipid levels by inhibiting HMGCR, APOB, PCSK9, SRB1, and VCAM1 and via improving PPAR-α and APOE mRNA levels. BZX-2 demonstrated its antihyperlipidemic effects mainly due to inhibition of APOB, while BZX-4-mediated effects appeared to be due to attenuation of APOB, PCSK9, and SRB1. BZX derivatives displayed strong binding affinities with HMGCR, APOB, and VCAM1, which suggested that some of the interactions might be required for inhibition of these target proteins. Conclusions: Based on the current findings, it can be concluded that BZX derivatives exert their antihyperlipidemic effects via modulation of multiple lipid-regulating genes.
Collapse
Affiliation(s)
- Maryam Zaib
- Department
of Pharmacology, Faculty of Pharmacy, The
University of Lahore, Lahore 54000, Pakistan
| | - Muhammad Nasir Hayat Malik
- Department
of Pharmacology, Faculty of Pharmacy, The
University of Lahore, Lahore 54000, Pakistan
- ; . Tel: +92 334
846 640 7
| | - Ramla Shabbir
- Department
of Pharmacology, Faculty of Pharmacy, The
University of Lahore, Lahore 54000, Pakistan
| | - Muhammad Naveed Mushtaq
- Department
of Pharmacology, Faculty of Pharmacy, The
University of Lahore, Lahore 54000, Pakistan
| | - Waqas Younis
- Department
of Pharmacology, Faculty of Pharmacy, The
University of Lahore, Lahore 54000, Pakistan
- Department
of Pharmacology, Physiology and Neuroscience, New Jersey Medical School-Rutgers, Newark, New Jersey 07103, United States
| | - Shah Jahan
- Department
of Immunology, University of Health Sciences, Lahore 54000, Pakistan
| | - Ishtiaq Ahmed
- Department
of Pathobiology, University of Veterinary
and Animal Sciences (Jhang Campus), Jhang 35200, Pakistan
| | - Hafiz Aamir Ali Kharl
- Riphah
Institute of Pharmaceutical Sciences, Riphah
International University, Islamabad 44000, Pakistan
| |
Collapse
|
26
|
Fleeman RM, Snyder AM, Kuhn MK, Chan DC, Smith GC, Crowley NA, Arnold AC, Proctor EA. Predictive link between systemic metabolism and cytokine signatures in the brain of apolipoprotein E ε4 mice. Neurobiol Aging 2023; 123:154-169. [PMID: 36572594 PMCID: PMC9892258 DOI: 10.1016/j.neurobiolaging.2022.11.015] [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: 06/29/2022] [Revised: 11/23/2022] [Accepted: 11/25/2022] [Indexed: 12/12/2022]
Abstract
The ε4 variant of apolipoprotein E (APOE) is the strongest and most common genetic risk factor for Alzheimer's disease (AD). While the mechanism of conveyed risk is incompletely understood, promotion of inflammation, dysregulated metabolism, and protein misfolding and aggregation are contributors to accelerating disease. Here we determined the concurrent effects of systemic metabolic changes and brain inflammation in young (3-month-old) and aged (18-month-old) male and female mice carrying the APOE4 gene. Using functional metabolic assays alongside multivariate modeling of hippocampal cytokine levels, we found that brain cytokine signatures are predictive of systemic metabolic outcomes, independent of AD proteinopathies. Male and female mice each produce different cytokine signatures as they age and as their systemic metabolic phenotype declines, and these signatures are APOE genotype dependent. Ours is the first study to identify a quantitative and predictive link between systemic metabolism and specific pathological cytokine signatures in the brain. Our results highlight the effects of APOE4 beyond the brain and suggest the potential for bi-directional influence of risk factors in the brain and periphery.
Collapse
Affiliation(s)
- Rebecca M Fleeman
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA; Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
| | - Amanda M Snyder
- Department of Neurology, Penn State College of Medicine, Hershey, PA, USA
| | - Madison K Kuhn
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA; Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA; Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA; Center for Neural Engineering, Pennsylvania State University, University Park, PA, USA
| | - Dennis C Chan
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA; Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA; Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA; Center for Neural Engineering, Pennsylvania State University, University Park, PA, USA
| | - Grace C Smith
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA; Department of Biology, Pennsylvania State University, University Park, PA, USA
| | - Nicole A Crowley
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA; Center for Neural Engineering, Pennsylvania State University, University Park, PA, USA; Department of Biology, Pennsylvania State University, University Park, PA, USA
| | - Amy C Arnold
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA, USA
| | - Elizabeth A Proctor
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA; Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA; Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA; Center for Neural Engineering, Pennsylvania State University, University Park, PA, USA; Department of Engineering Science & Mechanics, Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
27
|
Yassine HN, Self W, Kerman BE, Santoni G, Navalpur Shanmugam N, Abdullah L, Golden LR, Fonteh AN, Harrington MG, Gräff J, Gibson GE, Kalaria R, Luchsinger JA, Feldman HH, Swerdlow RH, Johnson LA, Albensi BC, Zlokovic BV, Tanzi R, Cunnane S, Samieri C, Scarmeas N, Bowman GL. Nutritional metabolism and cerebral bioenergetics in Alzheimer's disease and related dementias. Alzheimers Dement 2023; 19:1041-1066. [PMID: 36479795 PMCID: PMC10576546 DOI: 10.1002/alz.12845] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/12/2022] [Accepted: 10/05/2022] [Indexed: 12/13/2022]
Abstract
Disturbances in the brain's capacity to meet its energy demand increase the risk of synaptic loss, neurodegeneration, and cognitive decline. Nutritional and metabolic interventions that target metabolic pathways combined with diagnostics to identify deficits in cerebral bioenergetics may therefore offer novel therapeutic potential for Alzheimer's disease (AD) prevention and management. Many diet-derived natural bioactive components can govern cellular energy metabolism but their effects on brain aging are not clear. This review examines how nutritional metabolism can regulate brain bioenergetics and mitigate AD risk. We focus on leading mechanisms of cerebral bioenergetic breakdown in the aging brain at the cellular level, as well as the putative causes and consequences of disturbed bioenergetics, particularly at the blood-brain barrier with implications for nutrient brain delivery and nutritional interventions. Novel therapeutic nutrition approaches including diet patterns are provided, integrating studies of the gut microbiome, neuroimaging, and other biomarkers to guide future personalized nutritional interventions.
Collapse
Affiliation(s)
- Hussein N Yassine
- Department of Medicine, Keck School of Medicine, University of Southern, California, Los Angeles, California, USA
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Wade Self
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Bilal E Kerman
- Department of Medicine, Keck School of Medicine, University of Southern, California, Los Angeles, California, USA
| | - Giulia Santoni
- Laboratory of Neuroepigenetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale Lausanne (EPFL), Lausanne, Switzerland
| | - NandaKumar Navalpur Shanmugam
- Department of Neurology, Genetics and Aging Research Unit, McCance Center for Brain Health, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Lesley R Golden
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Alfred N Fonteh
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- Huntington Medical Research Institutes, Pasadena, California, USA
| | - Michael G Harrington
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Johannes Gräff
- Laboratory of Neuroepigenetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale Lausanne (EPFL), Lausanne, Switzerland
| | - Gary E Gibson
- Brain and Mind Research Institute, Weill Cornell Medicine, Burke Neurological Institute, White Plains, New York, USA
| | - Raj Kalaria
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Jose A Luchsinger
- Department of Medicine and Epidemiology, Columbia University Irving Medical Center, New York City, New York, USA
| | - Howard H Feldman
- Department of Neurosciences, University of California, San Diego, California, USA
| | - Russell H Swerdlow
- Department of Neurology, University of Kansas School of Medicine, Kansas City, Kansas, USA
| | - Lance A Johnson
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Benedict C Albensi
- Nova Southeastern Univ. College of Pharmacy, Davie, Florida, USA
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Manitoba, Canada
| | - Berislav V Zlokovic
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Rudolph Tanzi
- Department of Neurology, Genetics and Aging Research Unit, McCance Center for Brain Health, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Stephen Cunnane
- Department of Medicine, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Cécilia Samieri
- Univ. Bordeaux, INSERM, BPH, U1219, F-33000, Bordeaux, France
| | - Nikolaos Scarmeas
- 1st Department of Neurology, Aiginition Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
- Department of Neurology, Columbia University, New York City, New York, USA
| | - Gene L Bowman
- Department of Neurology, Genetics and Aging Research Unit, McCance Center for Brain Health, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Helfgott Research Institute, National University of Natural Medicine, Portland, Oregon, USA
| |
Collapse
|
28
|
Chen F, Chen Y, Ke Q, Wang Y, Gong Z, Chen X, Cai Y, Li S, Sun Y, Peng X, Ji Y, Zhang T, Wu W, Cui L, Wang Y. ApoE4 associated with severe COVID-19 outcomes via downregulation of ACE2 and imbalanced RAS pathway. J Transl Med 2023; 21:103. [PMID: 36759834 PMCID: PMC9910247 DOI: 10.1186/s12967-023-03945-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 01/31/2023] [Indexed: 02/11/2023] Open
Abstract
BACKGROUND Recent numerous epidemiology and clinical association studies reported that ApoE polymorphism might be associated with the risk and severity of coronavirus disease 2019 (COVID-19), and yielded inconsistent results. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection relies on its spike protein binding to angiotensin-converting enzyme 2 (ACE2) receptor expressed on host cell membranes. METHODS A meta-analysis was conducted to clarify the association between ApoE polymorphism and the risk and severity of COVID-19. Multiple protein interaction assays were utilized to investigate the potential molecular link between ApoE and the SARS-CoV-2 primary receptor ACE2, ApoE and spike protein. Immunoblotting and immunofluorescence staining methods were used to access the regulatory effect of different ApoE isoform on ACE2 protein expression. RESULTS ApoE gene polymorphism (ε4 carrier genotypes VS non-ε4 carrier genotypes) is associated with the increased risk (P = 0.0003, OR = 1.44, 95% CI 1.18-1.76) and progression (P < 0.00001, OR = 1.85, 95% CI 1.50-2.28) of COVID-19. ApoE interacts with both ACE2 and the spike protein but did not show isoform-dependent binding effects. ApoE4 significantly downregulates ACE2 protein expression in vitro and in vivo and subsequently decreases the conversion of Ang II to Ang 1-7. CONCLUSIONS ApoE4 increases SARS-CoV-2 infectivity in a manner that may not depend on differential interactions with the spike protein or ACE2. Instead, ApoE4 downregulates ACE2 protein expression and subsequently the dysregulation of renin-angiotensin system (RAS) may provide explanation by which ApoE4 exacerbates COVID-19 disease.
Collapse
Affiliation(s)
- Feng Chen
- grid.410560.60000 0004 1760 3078Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China ,grid.419010.d0000 0004 1792 7072Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province Kunming Institute of Zoology Chinese Academy of Sciences, Kunming, Yunnan China
| | - Yanting Chen
- grid.410560.60000 0004 1760 3078Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China ,grid.33199.310000 0004 0368 7223Department of Neurology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Qiongwei Ke
- grid.410560.60000 0004 1760 3078Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yongxiang Wang
- grid.410560.60000 0004 1760 3078Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Zheng Gong
- grid.410560.60000 0004 1760 3078Institute of Laboratory Animal Center, Guangdong Medical University, Zhanjiang, China
| | - Xiongjin Chen
- grid.410560.60000 0004 1760 3078Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yujie Cai
- grid.410560.60000 0004 1760 3078Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Shengnan Li
- grid.410560.60000 0004 1760 3078Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yuanhong Sun
- grid.266871.c0000 0000 9765 6057Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX USA
| | - Xiaoping Peng
- grid.410560.60000 0004 1760 3078Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yao Ji
- grid.410560.60000 0004 1760 3078Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Tianzhen Zhang
- grid.410560.60000 0004 1760 3078Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Wenxian Wu
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China. .,Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, China. .,Shenzhen Research Institute, Shandong University, Shenzhen, China.
| | - Lili Cui
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.
| | - Yan Wang
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.
| |
Collapse
|
29
|
Valencia-Olvera AC, Maldonado Weng J, Christensen A, LaDu MJ, Pike CJ. Role of estrogen in women's Alzheimer's disease risk as modified by APOE. J Neuroendocrinol 2023; 35:e13209. [PMID: 36420620 PMCID: PMC10049970 DOI: 10.1111/jne.13209] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 09/29/2022] [Accepted: 10/13/2022] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is characterized by numerous sexual dimorphisms that impact the development, progression, and probably the strategies to prevent and treat the most common form of dementia. In this review, we consider this topic from a female perspective with a specific focus on how women's vulnerability to the disease is affected by the individual and interactive effects of estrogens and apolipoprotein E (APOE) genotype. Importantly, APOE appears to modulate systemic and neural outcomes of both menopause and estrogen-based hormone therapy. In the brain, dementia risk is greater in APOE4 carriers, and the impacts of hormone therapy on cognitive decline and dementia risk vary according to both outcome measure and APOE genotype. Beyond the CNS, estrogen and APOE genotype affect vulnerability to menopause-associated bone loss, dyslipidemia and cardiovascular disease risk. An emerging concept that may link these relationships is the possibility that the effects of APOE in women interact with estrogen status by mechanisms that may include modulation of estrogen responsiveness. This review highlights the need to consider the key AD risk factors of advancing age in a sex-specific manner to optimize development of therapeutic approaches for AD, a view aligned with the principle of personalized medicine.
Collapse
Affiliation(s)
- AC Valencia-Olvera
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - J Maldonado Weng
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - A Christensen
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089 USA
| | - MJ LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - CJ Pike
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089 USA
| |
Collapse
|
30
|
Gottschalk WK, Mahon S, Hodgson D, Barrera J, Hill D, Wei A, Kumar M, Dai K, Anderson L, Mihovilovic M, Lutz MW, Chiba-Falek O. The APOE-TOMM40 Humanized Mouse Model: Characterization of Age, Sex, and PolyT Variant Effects on Gene Expression. J Alzheimers Dis 2023; 94:1563-1576. [PMID: 37458041 PMCID: PMC10733864 DOI: 10.3233/jad-230451] [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] [Indexed: 07/18/2023]
Abstract
BACKGROUND The human chromosome 19q13.32 is a gene rich region and has been associated with multiple phenotypes, including late onset Alzheimer's disease (LOAD) and other age-related conditions. OBJECTIVE Here we developed the first humanized mouse model that contains the entire TOMM40 and APOE genes with all intronic and intergenic sequences including the upstream and downstream regions. Thus, the mouse model carries the human TOMM40 and APOE genes and their intact regulatory sequences. METHODS We generated the APOE-TOMM40 humanized mouse model in which the entire mouse region was replaced with the human (h)APOE-TOMM40 loci including their upstream and downstream flanking regulatory sequences using recombineering technologies. We then measured the expression of the human TOMM40 and APOE genes in the mice brain, liver, and spleen tissues using TaqMan based mRNA expression assays. RESULTS We investigated the effects of the '523' polyT genotype (S/S or VL/VL), sex, and age on the human TOMM40- and APOE-mRNAs expression levels using our new humanized mouse model. The analysis revealed tissue specific and shared effects of the '523' polyT genotype, sex, and age on the regulation of the human TOMM40 and APOE genes. Noteworthy, the regulatory effect of the '523' polyT genotype was observed for all studied organs. CONCLUSION The model offers new opportunities for basic science, translational, and preclinical drug discovery studies focused on the APOE genomic region in relation to LOAD and other conditions in adulthood.
Collapse
Affiliation(s)
- William K. Gottschalk
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC, USA
| | - Scott Mahon
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC, USA
| | - Dellila Hodgson
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, USA
| | - Julio Barrera
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, USA
| | - Delaney Hill
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, USA
| | - Angela Wei
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, USA
| | - Manish Kumar
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, USA
| | - Kathy Dai
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, USA
| | - Lauren Anderson
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC, USA
| | - Mirta Mihovilovic
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC, USA
| | - Michael W. Lutz
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC, USA
| | - Ornit Chiba-Falek
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
31
|
Kunizheva SS, Volobaev VP, Plotnikova MY, Kupriyanova DA, Kuznetsova IL, Tyazhelova TV, Rogaev EI. Current Trends and Approaches to the Search for Genetic Determinants of Aging and Longevity. RUSS J GENET+ 2022; 58:1427-1443. [PMID: 36590179 PMCID: PMC9794410 DOI: 10.1134/s1022795422120067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 12/29/2022]
Abstract
Aging is a natural process of extinction of the body and the main aspect that determines the life expectancy for individuals who have survived to the post-reproductive period. The process of aging is accompanied by certain physiological, immune, and metabolic changes in the body, as well as the development of age-related diseases. The contribution of genetic factors to human life expectancy is estimated at about 25-30%. Despite the success in identifying genes and metabolic pathways that may be involved in the life extension process in model organisms, the key question remains to what extent these data can be extrapolated to humans, for example, because of the complexity of its biological and sociocultural systems, as well as possible species differences in life expectancy and causes of mortality. New molecular genetic methods have significantly expanded the possibilities for searching for genetic factors of human life expectancy and identifying metabolic pathways of aging, the interaction of genes and transcription factors, the regulation of gene expression at the level of transcription, and epigenetic modifications. The review presents the latest research and current strategies for studying the genetic basis of human aging and longevity: the study of individual candidate genes in genetic population studies, variations identified by the GWAS method, immunogenetic differences in aging, and genomic studies to identify factors of "healthy aging." Understanding the mechanisms of the interaction between factors affecting the life expectancy and the possibility of their regulation can become the basis for developing comprehensive measures to achieve healthy longevity. Supplementary Information The online version contains supplementary material available at 10.1134/S1022795422120067.
Collapse
Affiliation(s)
- S. S. Kunizheva
- Center for Genetics and Life Sciences, Sirius University of Science and Technology, 354340 Sochi, Russia
- Moscow State University, 119234 Moscow, Russia
- Vavilov Institute of General Genetics, Russian Academy of Sciences, 119991 Moscow, Russia
| | - V. P. Volobaev
- Center for Genetics and Life Sciences, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - M. Yu. Plotnikova
- Center for Genetics and Life Sciences, Sirius University of Science and Technology, 354340 Sochi, Russia
- Moscow State University, 119234 Moscow, Russia
| | - D. A. Kupriyanova
- Center for Genetics and Life Sciences, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - I. L. Kuznetsova
- Center for Genetics and Life Sciences, Sirius University of Science and Technology, 354340 Sochi, Russia
- Vavilov Institute of General Genetics, Russian Academy of Sciences, 119991 Moscow, Russia
| | - T. V. Tyazhelova
- Vavilov Institute of General Genetics, Russian Academy of Sciences, 119991 Moscow, Russia
| | - E. I. Rogaev
- Center for Genetics and Life Sciences, Sirius University of Science and Technology, 354340 Sochi, Russia
- Moscow State University, 119234 Moscow, Russia
- Vavilov Institute of General Genetics, Russian Academy of Sciences, 119991 Moscow, Russia
- University of Massachusetts Chan Medical School, 01545 Shrewsbury, MA United States
| |
Collapse
|
32
|
Pitts KM, Neeson CE, Hall NE, Lin JB, Falah HK, Wang SL, Lo KT, Song CE, Margeta MA, Solá-Del Valle DA. Neurodegeneration Markers Galectin-3 and Apolipoprotein E Are Elevated in the Aqueous Humor of Eyes With Glaucoma. Transl Vis Sci Technol 2022; 11:1. [DOI: 10.1167/tvst.11.11.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- Kristen M. Pitts
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Cameron E. Neeson
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Nathan E. Hall
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Jonathan B. Lin
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Henisk K. Falah
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Silas L. Wang
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Kristine T. Lo
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Christian E. Song
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Milica A. Margeta
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - David A. Solá-Del Valle
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
33
|
Scarfò G, Piccarducci R, Daniele S, Franzoni F, Martini C. Exploring the Role of Lipid-Binding Proteins and Oxidative Stress in Neurodegenerative Disorders: A Focus on the Neuroprotective Effects of Nutraceutical Supplementation and Physical Exercise. Antioxidants (Basel) 2022; 11:2116. [PMID: 36358488 PMCID: PMC9686611 DOI: 10.3390/antiox11112116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/20/2022] [Accepted: 10/25/2022] [Indexed: 07/29/2023] Open
Abstract
The human brain is primarily composed of lipids, and their homeostasis is crucial to carry on normal neuronal functions. In order to provide an adequate amount of lipid transport in and out of the central nervous system, organisms need a set of proteins able to bind them. Therefore, alterations in the structure or function of lipid-binding proteins negatively affect brain homeostasis, as well as increase inflammation and oxidative stress with the consequent risk of neurodegeneration. In this regard, lifestyle changes seem to be protective against neurodegenerative processes. Nutraceutical supplementation with antioxidant molecules has proven to be useful in proving cognitive functions. Additionally, regular physical activity seems to protect neuronal vitality and increases antioxidant defenses. The aim of the present review was to investigate mechanisms that link lipid-binding protein dysfunction and oxidative stress to cognitive decline, also underlining the neuroprotective effects of diet and exercise.
Collapse
Affiliation(s)
- Giorgia Scarfò
- Department of Clinical and Experimental Medicine, Division of General Medicine, University of Pisa, 56126 Pisa, Italy
- Center for Rehabilitative Medicine “Sport and Anatomy”, University of Pisa, 56126 Pisa, Italy
| | | | - Simona Daniele
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
| | - Ferdinando Franzoni
- Department of Clinical and Experimental Medicine, Division of General Medicine, University of Pisa, 56126 Pisa, Italy
- Center for Rehabilitative Medicine “Sport and Anatomy”, University of Pisa, 56126 Pisa, Italy
| | - Claudia Martini
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
| |
Collapse
|
34
|
Wrona M, Skrypnik D. New-Onset Diabetes Mellitus, Hypertension, Dyslipidaemia as Sequelae of COVID-19 Infection-Systematic Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph192013280. [PMID: 36293857 PMCID: PMC9602450 DOI: 10.3390/ijerph192013280] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 10/06/2022] [Accepted: 10/13/2022] [Indexed: 06/05/2023]
Abstract
As the population recovers from the coronavirus disease 2019 (COVID-19) pandemic, a subset of individuals is emerging as post-coronavirus disease (post-COVID) patients who experience multifactorial long-term symptoms several weeks after the initial recovery from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. The aim of this systematic review is to present the latest scientific reports that evaluate changes in glucose levels, blood pressure readings and lipid profiles after recovery from COVID-19 to verify the hypothesis that new-onset diabetes mellitus, arterial hypertension and dyslipidaemia are a possible sequela of a COVID-19 infection. The open access databases PubMed and Google Scholar were searched. Articles investigating patients with residual clinical signs and biochemical alteration indicating diabetes, hypertension and dyslipidaemia at least a month after recovering from COVID-19 were included. It has been shown that a select number of patients were diagnosed with new-onset diabetes, arterial hypertension and dyslipidaemia after COVID-19 infection. Alterations in glucose levels, blood pressure and lipid profiles months after initial infection shows the importance of considering diabetes mellitus, arterial hypertension and dyslipidaemia as part of the multifactorial diagnostic criteria post-COVID to better provide evidence-based clinical care.
Collapse
Affiliation(s)
- Marysia Wrona
- Poznan University of Medical Sciences, 61-701 Poznan, Poland
| | - Damian Skrypnik
- Department of Treatment of Obesity, Metabolic Disorders and Clinical Dietetics, Poznan University of Medical Sciences, 60-569 Poznan, Poland
| |
Collapse
|
35
|
Pan X, Chen S, Chen X, Ren Q, Yue L, Niu S, Li Z, Zhu R, Chen X, Jia Z, Zhen R, Ban J. Effect of high-fat diet and empagliflozin on cardiac proteins in mice. Nutr Metab (Lond) 2022; 19:69. [PMID: 36242090 PMCID: PMC9563173 DOI: 10.1186/s12986-022-00705-0] [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: 06/15/2022] [Accepted: 10/02/2022] [Indexed: 11/16/2022] Open
Abstract
Using proteomic techniques the impact of the sodium-glucose transport protein 2 inhibitor empagliflozin on cardiac protein expression in a mouse model was assessed under normal and high-fat diet (HFD) conditions. We examined the effect of obesity on serological markers and heart function in obese mice treated with or without empagliflozin and used proteomic techniques to investigate alterations in cardiac protein expression. Using bioinformatic techniques, data were screened for differentially expressed proteins (DEPs) implicated in the putative mechanism of empagliflozin's cardioprotective effects. In C57BL/6 mice, HFD increased body weight, blood lipid, and glucose levels and was associated with structural damage to the heart. Empagliflozin reduces body weight, improves glucose and lipid metabolism, alleviates obesity-induced cardiac ventricular wall thickening, and lowers cardiac tissue collagen. The expression of several proteins was altered in the heart, mainly related to lipid metabolism. Following empagliflozin treatment, the expression of several lipid metabolism-related proteins was considerably reduced. Further examination of DEPs revealed that following empagliflozin treatment, the expressions of Apoe, Apoc1, Saa2, Apoa2, and Pon1 altered dramatically, suggesting that these proteins may be the main proteins that empagliflozin uses to treat obesity-induced aberrant lipid metabolism. Empagliflozin may protect the heart by altering the expression of genes including Apoe, Apoc1, Saa2, Apoa2, and Pon1, which are all involved in lipid metabolism disturbance in obesity.
Collapse
Affiliation(s)
- Xiaoyu Pan
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China.,Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
| | - Shuchun Chen
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China. .,Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China.
| | - Xing Chen
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China.,Department of Nephrology, Hebei General Hospital, Shijiazhuang, China
| | - Qingjuan Ren
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China.,Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
| | - Lin Yue
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China.,Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
| | - Shu Niu
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China.,Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
| | - Zelin Li
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China.,Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
| | - Ruiyi Zhu
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China.,Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
| | - Xiaoyi Chen
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
| | - Zhuoya Jia
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China.,Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
| | - Ruoxi Zhen
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China.,Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
| | - Jiangli Ban
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
| |
Collapse
|
36
|
Liver-ing in your head rent free: peripheral ApoE4 drives CNS pathology. Mol Neurodegener 2022; 17:65. [PMID: 36195891 PMCID: PMC9531445 DOI: 10.1186/s13024-022-00569-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 09/22/2022] [Indexed: 11/10/2022] Open
|
37
|
27-Hydroxycholesterol-Induced Dysregulation of Cholesterol Metabolism Impairs Learning and Memory Ability in ApoE ε4 Transgenic Mice. Int J Mol Sci 2022; 23:ijms231911639. [PMID: 36232940 PMCID: PMC9569856 DOI: 10.3390/ijms231911639] [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: 07/12/2022] [Revised: 09/18/2022] [Accepted: 09/20/2022] [Indexed: 11/09/2022] Open
Abstract
Dysregulated brain cholesterol metabolism is one of the characteristics of Alzheimer’s disease (AD). 27-Hydroxycholesterol (27-OHC) is a cholesterol metabolite that plays an essential role in regulating cholesterol metabolism and it is suggested that it contributes to AD-related cognitive deficits. However, the link between 27-OHC and cholesterol homeostasis, and how this relationship relates to AD pathogenesis, remain elusive. Here, 12-month-old ApoE ε4 transgenic mice were injected with saline, 27-OHC, 27-OHC synthetase inhibitor (anastrozole, ANS), and 27-OHC+ANS for 21 consecutive days. C57BL/6J mice injected with saline were used as wild-type controls. The indicators of cholesterol metabolism, synaptic structure, amyloid β 1-42 (Aβ1-42), and learning and memory abilities were measured. Compared with the wild-type mice, ApoE ε4 mice had poor memory and dysregulated cholesterol metabolism. Additionally, damaged brain tissue and synaptic structure, cognitive decline, and higher Aβ1-42 levels were observed in the 27-OHC group. Moreover, cholesterol transport proteins such as ATP-binding cassette transporter A1 (ABCA1), apolipoprotein E (ApoE), low-density lipoprotein receptor (LDLR), and low-density lipoprotein receptor-related protein1 (LRP1) were up-regulated in the cortex after the 27-OHC treatment. The levels of cholesterol metabolism-related indicators in the hippocampus were not consistent with those in the cortex. Additionally, higher serum apolipoprotein A1 (ApoA1) levels and lower serum ApoE levels were observed in the 27-OHC group. Notably, ANS partially reversed the effects of 27-OHC. In conclusion, the altered cholesterol metabolism induced by 27-OHC was involved in Aβ1-42 deposition and abnormalities in both the brain tissue and synaptic structure, ultimately leading to memory loss in the ApoE ε4 transgenic mice.
Collapse
|
38
|
Liu M, Chen MY, An L, Ma SQ, Mei J, Huang WH, Zhang W. Effects of apolipoprotein E on regulating insulin sensitivity via regulating insulin receptor signalosome in caveolae. Life Sci 2022; 308:120929. [PMID: 36063979 DOI: 10.1016/j.lfs.2022.120929] [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: 06/09/2022] [Revised: 08/28/2022] [Accepted: 08/30/2022] [Indexed: 10/31/2022]
Abstract
AIMS Although impaired insulin signaling at a post-receptor level was a well-established key driver of insulin resistance, the role of surface abnormal insulin receptor (INSR) location in insulin resistance pathogenesis tended to be ignored and its molecular mechanisms remained obscure. Herein, this study aimed to investigate hepatic apolipoprotein E (APOE) impaired cellular insulin action via reducing cell surface INSR, especially in caveolae. KEY FINDINGS Downregulation of APOE enhanced the caveolae translocation of INSR and glucose transporter 2 (GLUT2), and improved hepatic cells' sensitivity to insulin. Consistently, mice with selective suppression of liver tissue APOE showed lower fasting insulin and fasting glucose levels, better homeostatic model assessment (HOMA) index (HOMA-IS, HOMA-IR, HOMA-β) and quantitative insulin sensitivity check index (QUICKI). Furthermore, the co-localization of INSR and CAV1 in the liver of these mice were more substantial than controls. SIGNIFICANCE APOE might adversely set the basal gain of INSR signaling implied that APOE could be a new endogenous INSR regulator.
Collapse
Affiliation(s)
- Miao Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, 110 Xiangya Road, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha 410008, Hunan, PR China
| | - Man-Yun Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, 110 Xiangya Road, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha 410008, Hunan, PR China
| | - Liang An
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, 110 Xiangya Road, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha 410008, Hunan, PR China
| | - Si-Qing Ma
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, 110 Xiangya Road, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha 410008, Hunan, PR China
| | - Jie Mei
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, 110 Xiangya Road, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha 410008, Hunan, PR China
| | - Wei-Hua Huang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, 110 Xiangya Road, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha 410008, Hunan, PR China; NHC Key Laboratory of Birth Defect for Research and Prevention (Hunan Provincial Maternal and Child Health Care Hospital), Hunan 410008, PR China.
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, 110 Xiangya Road, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha 410008, Hunan, PR China.
| |
Collapse
|
39
|
Patterson AM, Vemula S, Plett PA, Sampson CH, Chua HL, Fisher A, Wu T, Sellamuthu R, Feng H, Katz BP, DesRosiers CM, Pelus LM, Cox GN, MacVittie TJ, Orschell CM. Age and Sex Divergence in Hematopoietic Radiosensitivity in Aged Mouse Models of the Hematopoietic Acute Radiation Syndrome. Radiat Res 2022; 198:221-242. [PMID: 35834823 PMCID: PMC9512046 DOI: 10.1667/rade-22-00071.1] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 06/11/2022] [Indexed: 11/03/2022]
Abstract
The hematopoietic system is highly sensitive to stress from both aging and radiation exposure, and the hematopoietic acute radiation syndrome (H-ARS) should be modeled in the geriatric context separately from young for development of age-appropriate medical countermeasures (MCMs). Here we developed aging murine H-ARS models, defining radiation dose response relationships (DRRs) in 12-month-old middle-aged and 24-month-old geriatric male and female C57BL/6J mice, and characterized diverse factors affecting geriatric MCM testing. Groups of approximately 20 mice were exposed to ∼10 different doses of radiation to establish radiation DRRs for estimation of the LD50/30. Radioresistance increased with age and diverged dramatically between sexes. The LD50/30 in young adult mice averaged 853 cGy and was similar between sexes, but increased in middle age to 1,005 cGy in males and 920 cGy in females, with further sex divergence in geriatric mice to 1,008 cGy in males but 842 cGy in females. Correspondingly, neutrophils, platelets, and functional hematopoietic progenitor cells were all increased with age and rebounded faster after irradiation. These effects were higher in aged males, and neutrophil dysfunction was observed in aged females. Upstream of blood production, hematopoietic stem cell (HSC) markers associated with age and myeloid bias (CD61 and CD150) were higher in geriatric males vs. females, and sex-divergent gene signatures were found in HSCs relating to cholesterol metabolism, interferon signaling, and GIMAP family members. Fluid intake per gram body weight decreased with age in males, and decreased after irradiation in all mice. Geriatric mice of substrain C57BL/6JN sourced from the National Institute on Aging were significantly more radiosensitive than C57BL/6J mice from Jackson Labs aged at our institution, indicating mouse source and substrain should be considered in geriatric radiation studies. This work highlights the importance of sex, vendor, and other considerations in studies relating to hematopoiesis and aging, identifies novel sex-specific functional and molecular changes in aging hematopoietic cells at steady state and after irradiation, and presents well-characterized aging mouse models poised for MCM efficacy testing for treatment of acute radiation effects in the elderly.
Collapse
Affiliation(s)
- Andrea M. Patterson
- Department of Medicine Indiana University School of Medicine Indianapolis, Indiana
| | - Sasidhar Vemula
- Department of Medicine Indiana University School of Medicine Indianapolis, Indiana
| | - P. Artur Plett
- Department of Medicine Indiana University School of Medicine Indianapolis, Indiana
| | - Carol H. Sampson
- Department of Medicine Indiana University School of Medicine Indianapolis, Indiana
| | - Hui Lin Chua
- Department of Medicine Indiana University School of Medicine Indianapolis, Indiana
| | - Alexa Fisher
- Department of Medicine Indiana University School of Medicine Indianapolis, Indiana
| | - Tong Wu
- Department of Medicine Indiana University School of Medicine Indianapolis, Indiana
| | - Rajendran Sellamuthu
- Department of Medicine Indiana University School of Medicine Indianapolis, Indiana
| | - Hailin Feng
- Department of Medicine Indiana University School of Medicine Indianapolis, Indiana
| | - Barry P. Katz
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, Indiana
| | - Colleen M. DesRosiers
- Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Louis M. Pelus
- Department of Medicine Indiana University School of Medicine Indianapolis, Indiana
- Department of Microbiology & Immunology, Indiana University School of Medicine, Indianapolis, Indiana
| | | | | | - Christie M. Orschell
- Department of Medicine Indiana University School of Medicine Indianapolis, Indiana
| |
Collapse
|
40
|
Apolipoprotein E in Cardiometabolic and Neurological Health and Diseases. Int J Mol Sci 2022; 23:ijms23179892. [PMID: 36077289 PMCID: PMC9456500 DOI: 10.3390/ijms23179892] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/25/2022] [Accepted: 08/28/2022] [Indexed: 11/17/2022] Open
Abstract
A preponderance of evidence obtained from genetically modified mice and human population studies reveals the association of apolipoprotein E (apoE) deficiency and polymorphisms with pathogenesis of numerous chronic diseases, including atherosclerosis, obesity/diabetes, and Alzheimer’s disease. The human APOE gene is polymorphic with three major alleles, ε2, ε3 and ε4, encoding apoE2, apoE3, and apoE4, respectively. The APOE gene is expressed in many cell types, including hepatocytes, adipocytes, immune cells of the myeloid lineage, vascular smooth muscle cells, and in the brain. ApoE is present in subclasses of plasma lipoproteins, and it mediates the clearance of atherogenic lipoproteins from plasma circulation via its interaction with LDL receptor family proteins and heparan sulfate proteoglycans. Extracellular apoE also interacts with cell surface receptors and confers signaling events for cell regulation, while apoE expressed endogenously in various cell types regulates cell functions via autocrine and paracrine mechanisms. This review article focuses on lipoprotein transport-dependent and -independent mechanisms by which apoE deficiency or polymorphisms contribute to cardiovascular disease, metabolic disease, and neurological disorders.
Collapse
|
41
|
Prasad K, Gour P, Raghuvanshi S, Kumar V. The SARS-CoV-2 targeted human RNA binding proteins network biology to investigate COVID-19 associated manifestations. Int J Biol Macromol 2022; 217:853-863. [PMID: 35907451 PMCID: PMC9328843 DOI: 10.1016/j.ijbiomac.2022.07.200] [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: 02/06/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 11/21/2022]
Abstract
The global coronavirus disease 2019 (COVID-19) pandemic caused by the SARS-CoV-2 virus has had unprecedented social and economic ramifications. Identifying targets for drug repurposing could be an effective means to present new and fast treatments. Furthermore, the risk of morbidity and mortality from COVID-19 goes up when there are coexisting medical conditions, however, the underlying mechanisms remain unclear. In the current study, we have adopted a network-based systems biology approach to investigate the RNA binding proteins (RBPs)-based molecular interplay between COVID-19, various human cancers, and neurological disorders. The network based on RBPs commonly involved in the three disease conditions consisted of nine RBPs connecting 10 different cancer types, 22 brain disorders, and COVID-19 infection, ultimately hinting at the comorbidities and complexity of COVID-19. Further, we underscored five miRNAs with reported antiviral properties that target all of the nine shared RBPs and are thus therapeutically valuable. As a strategy to improve the clinical conditions in comorbidities associated with COVID-19, we propose perturbing the shared RBPs by drug repurposing. The network-based analysis presented hereby contributes to a better knowledge of the molecular underpinnings of the comorbidities associated with COVID-19.
Collapse
Affiliation(s)
- Kartikay Prasad
- Amity Institute of Neuropsychology & Neurosciences, Amity University, Noida, UP 201303, India
| | - Pratibha Gour
- Dept. of Plant Molecular Biology, University of Delhi, South Campus, New Delhi 110021, India
| | - Saurabh Raghuvanshi
- Dept. of Plant Molecular Biology, University of Delhi, South Campus, New Delhi 110021, India.
| | - Vijay Kumar
- Amity Institute of Neuropsychology & Neurosciences, Amity University, Noida, UP 201303, India.
| |
Collapse
|
42
|
The Role of ApoE Serum Levels and ApoE Gene Polymorphisms in Patients with Laryngeal Squamous Cell Carcinoma. Biomolecules 2022; 12:biom12081013. [PMID: 35892323 PMCID: PMC9331506 DOI: 10.3390/biom12081013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/13/2022] [Accepted: 07/19/2022] [Indexed: 11/17/2022] Open
Abstract
Recent studies have revealed that the inflammatory ApoE effect may play a significant role in various cancer development. However, this effect has still not been analyzed in patients with laryngeal squamous cell carcinoma (LSCC). In the present study, we evaluated two single nucleotide polymorphisms (SNPs) of ApoE (rs7412 and rs429358) and determined their associations with LSCC development and the LSCC patients’ five-year survival rate. Additionally, we analyzed serum ApoE levels using an enzyme-linked immunosorbent assay. A total of 602 subjects (291 histologically verified LSCC patients and 311 healthy controls) were involved in this study. The genotyping was carried out using the real-time PCR. We revealed that ApoE ε3/ε3 was associated with a 1.7-fold higher probability of developing LSCC (p = 0.001), with 1.7-fold increased odds of developing LSCC without metastasis to the lymph nodes (p = 0.002) and with a 2.0-fold increased odds of developing well-differentiated LSCC (p = 0.008), as well as 1.6-fold increased odds of developing poorly differentiated LSCC development (p = 0.012). The ApoE ε2/ε4 and ε3/ε4 genotypes were associated with a 2.9-fold and 1.5-fold decrease in the likelihood of developing LSCC (p = 0.042; p = 0.037, respectively). ApoE ε3/ε4 was found associated with a 2.4-fold decreased likelihood of developing well-differentiated LSCC (p = 0.013). Conclusion: ApoE ε2/ε4 and ε3/ε4 were found to play a protective role in LSCC development, while ApoE ε3/ε3 may have a risk position in LSCC development.
Collapse
|
43
|
Wang L, Shao C, Han C, Li P, Wang F, Wang Y, Li J. Correlation of ApoE gene polymorphism with acute myocardial infarction and aspirin resistance after percutaneous coronary intervention. Am J Transl Res 2022; 14:3303-3310. [PMID: 35702102 PMCID: PMC9185036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 03/31/2022] [Indexed: 06/15/2023]
Abstract
OBJECTIVE To determine the correlation of Apolipoprotein E (ApoE) gene polymorphism with acute myocardial infarction (AMI) and aspirin (APC) resistance after percutaneous coronary intervention (PCI). METHODS In this randomized controlled trial (The Second People's Hospital of Lianyungang Ethics Committee No.L1719), a total of 120 AMI patients admitted to the Second People's Hospital of Lianyungang from January 2019 to June 2020 were enrolled into the research group (Res group) and 120 healthy individuals during the same time period into the control group (Con group). ApoE gene polymorphism was detected by gene microarray and analyzed statistically. The occurrence of APC resistance after PCI was recorded, and the relationship between ApoE gene polymorphism and APC resistance was analyzed. RESULTS The Res group showed a significantly lower level of ε3/ε3 gene and significantly higher levels of ε3/ε4 and ε4/ε4 genes than the Con group (all P<0.05), but no notable difference was found in the distribution of ApoE ε2 between the two groups (P>0.05). ApoE ε3 carriers were the main carriers in both groups. However, the Res group showed a lower frequency of ApoE ε3 and a higher frequency of ApoE ε4 compared to the Con group (both P<0.05), and patients with more severe AMI had a significantly higher frequency of ApoE ε4 genotype (P<0.05). According to logistic regression analysis, carrying ApoE ε4 allele (ε3/ε4, ε4/ε4) was a risk factor for AMI (P<0.05). Additionally, patients with APC resistance had a significantly higher frequency of ApoE ε4 allele than those without it (P<0.05). A higher frequency of ApoE ε4 allele was also a risk factor of APC resistance in AMI patients after PCI, and its adjusted risk ratio (OR) was 2.26 times (P<0.05). Moreover, no significant difference was observed among patients with different ApoE genotypes in the incidence of adverse events (P>0.05). CONCLUSION ApoE gene polymorphism is correlated with AMI and APC resistance after PCI, and ApoE ε4 genotype is probably the risk allele for AMI.
Collapse
Affiliation(s)
- Luoqing Wang
- Department of Cardiovascular Medicine, Lianyungang Second People’s Hospital Affiliated to Bengbu Medical CollegeLianyungang 222006, Jiangsu, China
| | - Chen Shao
- Department of Cardiovascular Medicine, Lianyungang Second People’s Hospital Affiliated to Bengbu Medical CollegeLianyungang 222006, Jiangsu, China
| | - Cuimin Han
- Department of Cardiovascular Medicine, Lianyungang Second People’s Hospital Affiliated to Bengbu Medical CollegeLianyungang 222006, Jiangsu, China
| | - Peng Li
- Department of Cardiovascular Medicine, Lianyungang Second People’s Hospital Affiliated to Bengbu Medical CollegeLianyungang 222006, Jiangsu, China
| | - Feixiang Wang
- Department of Cardiovascular Medicine, Lianyungang Second People’s Hospital Affiliated to Bengbu Medical CollegeLianyungang 222006, Jiangsu, China
| | - Yilian Wang
- Department of Cardiovascular Medicine, Lianyungang Second People’s Hospital Affiliated to Bengbu Medical CollegeLianyungang 222006, Jiangsu, China
| | - Junping Li
- Department of Electrocardiogram, Lianyungang Second People’s Hospital Affiliated to Bengbu Medical CollegeLianyungang 222006, Jiangsu, China
| |
Collapse
|
44
|
Ahmed S, Pande AH, Sharma SS. Therapeutic potential of ApoE-mimetic peptides in CNS disorders: Current perspective. Exp Neurol 2022; 353:114051. [DOI: 10.1016/j.expneurol.2022.114051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/23/2022] [Accepted: 03/14/2022] [Indexed: 02/07/2023]
|
45
|
Angiogenin Levels and Their Association with Cardiometabolic Indices Following Vitamin D Status Correction in Saudi Adults. BIOLOGY 2022; 11:biology11020286. [PMID: 35205153 PMCID: PMC8868634 DOI: 10.3390/biology11020286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/10/2022] [Accepted: 02/10/2022] [Indexed: 11/23/2022]
Abstract
Simple Summary Angiogenin (ANG) is a small 123 amino acid protein which in normal growth is associated with formation of new blood vessels in a process called angiogenesis; however, the abnormal levels of this protein in blood has been associated with diseases such as cancer, neurological disorders, and cardiovascular diseases. Vitamin D deficiency and elevated levels of blood lipids have also been associated with many diseases including cardiovascular diseases. In this study, the investigators tried to evaluate the relationship between the circulating levels of ANG, vitamin D, and lipids. The model used was vitamin D supplementation of deficient Saudi adults in order to find the effect of vitamin D correction on circulating levels of ANG and blood lipids. With vitamin D supplementation, modest but non-significant elevation in ANG was observed, as well as significant increase in apolipoproteins CIII and E and significant decrease in apo B. In addition, the correlation between circulating levels of ANG and apolipoproteins especially apo E observed in this study are interesting and should be investigated more as both are linked with neurologic disorders like Alzheimer’s and Parkinson’s diseases. Abstract Angiogenin (ANG), a multifunctional protein known to induce blood vessel formation, is a potential biomarker for cardiovascular diseases; however, whether it is affected by vitamin D supplementation is not known. This interventional study in vitamin D-deficient Saudi adults was designed to investigate it. A total of 100 vitamin D-deficient Saudi adults aged 30–50 years were randomly selected to undergo 6-month vitamin D supplementation. Circulating levels of fasting glucose, lipids, vitamin D, apolipoproteins (AI, AII, B, CI, CII, CIII, E, and H), and ANG were measured using commercially available assays at baseline and after six months. Overall, vitamin D levels increased significantly post intervention. With this, levels of apo-CIII and apo-E significantly increased (p-values of 0.001 and 0.009, respectively) with a significant parallel decrease in apo-B (p = 0.003). ANG levels were significantly positively associated with most apolipoproteins and inversely correlated with HDL-cholesterol. Post intervention, the changes in ANG levels were positively correlated with apo-E (r = 0.32; p < 0.01 in all subjects and r = 0.40; p < 0.05 in males). Vitamin D supplementation may modestly affect ANG levels. The association observed between ANG and apo-E is worthy of further investigation since both biomarkers have been linked to neurodegenerative disorders.
Collapse
|
46
|
Zajac DJ, Green SJ, Johnson LA, Estus S. APOE genetics influence murine gut microbiome. Sci Rep 2022; 12:1906. [PMID: 35115575 PMCID: PMC8814305 DOI: 10.1038/s41598-022-05763-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 01/17/2022] [Indexed: 01/03/2023] Open
Abstract
Apolipoprotein E (APOE) alleles impact pathogenesis and risk for multiple human diseases, making them primary targets for disease treatment and prevention. Previously, we and others reported an association between APOE alleles and the gut microbiome. Here, we evaluated effects of APOE heterozygosity and tested whether these overall results extended to mice maintained under ideal conditions for microbiome analyses. To model human APOE alleles, this study used APOE targeted replacement (TR) mice on a C57Bl/6 background. To minimize genetic drift, homozygous APOE3 mice were crossed to homozygous APOE2 or homozygous APOE4 mice prior to the study, and the resulting heterozygous progeny crossed further to generate the study mice. To maximize environmental homogeneity, mice with mixed genotypes were housed together and used bedding from the cages was mixed and added back as a portion of new bedding. Fecal samples were obtained from mice at 3-, 5- and 7-months of age, and microbiota analyzed by 16S ribosomal RNA gene amplicon sequencing. Linear discriminant analysis of effect size (LefSe) identified taxa associated with APOE status, depicted as cladograms to show phylogenetic relatedness. The influence of APOE status was tested on alpha-diversity (Shannon H index) and beta-diversity (principal coordinate analyses and PERMANOVA). Individual taxa associated with APOE status were identified by classical univariate analysis. Whether findings in the APOE mice were replicated in humans was evaluated by using published microbiome genome wide association data. Cladograms revealed robust differences with APOE in male mice and limited differences in female mice. The richness and evenness (alpha-diversity) and microbial community composition (beta-diversity) of the fecal microbiome was robustly associated with APOE status in male but not female mice. Classical univariate analysis revealed individual taxa that were significantly increased or decreased with APOE, illustrating a stepwise APOE2-APOE3-APOE4 pattern of association with heterozygous animals trending as intermediate in the stepwise pattern. The relative abundance of bacteria from the class Clostridia, order Clostridiales, family Ruminococacceae and related genera increased with APOE2 status. The relative abundance of Erysipelotrichia increased with APOE4 status, a finding that extended to humans. In this study, wherein mice were maintained in an ideal fashion for microbiome studies, gut microbiome profiles were strongly and significantly associated with APOE status in male APOE-TR mice. Erysipelotrichia are increased with APOE4 in both mice and humans. APOE allelic effects appeared generally intermediate in heterozygous animals. Further evaluation of these findings in humans, as well as studies evaluating the impact of the APOE-associated microbiota on disease-relevant phenotypes, will be necessary to determine if alterations in the gut microbiome represent a novel mechanism whereby APOE alleles impact disease.
Collapse
Affiliation(s)
- Diana J Zajac
- Department of Physiology and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Stefan J Green
- Genome Research Core, Research Resources Center, University of Illinois at Chicago, Chicago, IL, USA
- Genomics and Microbiome Core Facility, Rush University Medical Center, Chicago, IL, USA
| | - Lance A Johnson
- Department of Physiology and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Steven Estus
- Department of Physiology and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA.
- Department of Physiology, University of Kentucky, 789 S. Limestone, Rm. 537, Lexington, KY, 40536, USA.
| |
Collapse
|
47
|
Tatulian SA. Challenges and hopes for Alzheimer's disease. Drug Discov Today 2022; 27:1027-1043. [PMID: 35121174 DOI: 10.1016/j.drudis.2022.01.016] [Citation(s) in RCA: 113] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 09/01/2021] [Accepted: 01/27/2022] [Indexed: 12/11/2022]
Abstract
Recent drug development efforts targeting Alzheimer's disease (AD) have failed to produce effective disease-modifying agents for many reasons, including the substantial presymptomatic neuronal damage that is caused by the accumulation of the amyloid β (Aβ) peptide and tau protein abnormalities, deleterious adverse effects of drug candidates, and inadequate design of clinical trials. New molecular targets, biomarkers, and diagnostic techniques, as well as alternative nonpharmacological approaches, are sorely needed to detect and treat early pathological events. This article analyzes the successes and debacles of pharmaceutical endeavors to date, and highlights new technologies that may lead to the more effective diagnosis and treatment of the pathologies that underlie AD. The use of focused ultrasound, deep brain stimulation, stem cell therapy, and gene therapy, in parallel with pharmaceuticals and judicious lifestyle adjustments, holds promise for the deceleration, prevention, or cure of AD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Suren A Tatulian
- Department of Physics, College of Sciences, and Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA.
| |
Collapse
|
48
|
Sun S, Meng Q, Bai Y, Cao C, Li J, Cheng B, Shi B, Shan A. Lycopene improves maternal reproductive performance by modulating milk composition and placental antioxidative and immune status. Food Funct 2021; 12:12448-12467. [PMID: 34792070 DOI: 10.1039/d1fo01595h] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Placental health and milk quality are important for maternal reproductive performance during pregnancy and lactation. Lycopene plays an important role in antioxidation, anti-inflammation and regulating lipid metabolism. The goal of the present study was to investigate the effects of dietary lycopene supplementation in the pig model on reproductive performance, placental health and milk composition during maternal gestation and lactation. In the present study, the litter size of live piglets was increased and the litter size of dead piglets was decreased by lycopene supplementation of the diet of sows. The litter weight at birth and weaning were increased in the lycopene group. Through placental proteomics, we enriched differentially expressed proteins (DEPs), gene ontology (GO) terms, and Kyoto encyclopedia of proteins and genomes (KEGG) pathways involved in immunity, anti-inflammation, antioxidants, and lipid metabolism and transport. Furthermore, in terms of placental health, we analyzed the levels of related enzymes, metabolites and mRNA expression in the placenta. Lycopene was shown to reduce mRNA expression and the levels of placental inflammatory factors, increase the content of immunoglobulin, improve the antioxidant capacity, and improve lipid metabolism and lipid transport in the placenta. In terms of sow milk composition, lycopene increased the levels of immunoglobulins in colostrum and lactose in colostrum and milk. Overall, the results of the present study demonstrate that dietary lycopene supplementation of sows during gestation and lactation improves the reproductive performance to a certain extent; this may be due to lycopene improving the placental health and milk composition of sows.
Collapse
Affiliation(s)
- Shishuai Sun
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, P. R. China.
| | - Qingwei Meng
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, P. R. China.
| | - Yongsong Bai
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, P. R. China.
| | - Chunyu Cao
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, P. R. China.
| | - Jibo Li
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, P. R. China.
| | - Baojing Cheng
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, P. R. China.
| | - Baoming Shi
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, P. R. China.
| | - Anshan Shan
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, P. R. China.
| |
Collapse
|
49
|
Abstract
This review summarizes the available data about genetic factors which can link ischemic stroke and sleep. Sleep patterns (subjective and objective measures) are characterized by heritability and comprise up to 38-46%. According to Mendelian randomization analysis, genetic liability for short sleep duration and frequent insomnia symptoms is associated with ischemic stroke (predominantly of large artery subtype). The potential genetic links include variants of circadian genes, genes encoding components of neurotransmitter systems, common cardiovascular risk factors, as well as specific genetic factors related to certain sleep disorders.
Collapse
Affiliation(s)
- Lyudmila Korostovtseva
- Sleep Laboratory, Research Department for Hypertension, Department for Cardiology, Almazov National Medical Research Centre, 2 Akkuratov Str., Saint Petersburg, 197341, Russia.
| |
Collapse
|
50
|
Liu X, Lin Q, Fan K, Tang M, Zhang W, Yang B, Ou Q. The effects of genetic polymorphisms of APOE on circulating lipid levels in middle-aged and elderly chinese Fujian Han population: toward age- and sex-personalized management. Lipids Health Dis 2021; 20:158. [PMID: 34749748 PMCID: PMC8573895 DOI: 10.1186/s12944-021-01587-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 10/25/2021] [Indexed: 11/23/2022] Open
Abstract
Background Increased evidence has reported the association of genetic polymorphisms of Apolipoprotein E (APOE) with serum lipids. However, few studies have explored the combined effects of APOE, gender, and age. Methods A total of 1,419 middle-aged and elderly subjects were randomly selected and studied. The APOE genotypes and the serum lipids were detected. The effects of APOE, gender, and age on serum lipids were preliminarily observed in general. The subjects were then divided into the middle-aged group (40–64 years old) and the elderly group (≥ 65 years old), for both males and females, to explore the combined effects of the APOE, gender, and age on serum lipids. Finally, a multivariate logistic regression model was used to evaluate the associations between the APOE allele carriers and the at-risk levels of dyslipidemia. Results The serum TC, LDL-C, and ApoB in the ε2 carriers were lower than the ε3 carriers (all P < 0.05), and there was no significant difference in the ε4 carriers compared to the ε3 carriers in general (all P > 0.05). The serum LDL-C and ApoB of the ε2 carriers were lower than the noncarriers in the middle-aged and elderly males (all P < 0.05). The serum TC in the ε2 carriers was lower than the noncarriers only in middle-aged males (P < 0.05). As to the levels of serum HDL-C and ApoA1, the ε2 carriers were higher than the noncarriers in middle-aged females (all P < 0.05), and the ε4 carriers were lower than noncarriers in middle-aged males (P < 0.05). Especially, the serum TG in the ε4 carriers was significantly higher than the noncarriers in elderly females. The logistic regression analysis indicated that the ε2 carriers were less likely to have at-risk levels of high LDL-C in middle-aged and elderly males (all P < 0.05) versus low HDL-C in middle-aged females (P < 0.05). In contrast, the ε4 carriers were more likely to have at-risk levels of high TG in elderly females (P < 0.05). Conclusions The effects of the genetic polymorphisms of APOE on the serum lipids were both gender- and age-dependent in the middle-aged and elderly Chinese Fujian Han population.
Collapse
Affiliation(s)
- Xiaofeng Liu
- Department of Laboratory Medicine, the First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, 350005, Fuzhou, China. .,Fujian Key Laboratory of Laboratory Medicine, No.20 Chazhong Road, 20 Chazhong Road, 350005, Fuzhou, China. .,Gene Diagnostic Laboratory, the First Affiliated Hospital of Fujian Medical University, 350005, Fuzhou, China.
| | - Qingwen Lin
- Department of Laboratory Medicine, the First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, 350005, Fuzhou, China
| | - Kengna Fan
- Department of Laboratory Medicine, the First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, 350005, Fuzhou, China
| | - Minjie Tang
- Department of Laboratory Medicine, the First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, 350005, Fuzhou, China
| | - Weiqing Zhang
- Department of Laboratory Medicine, the First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, 350005, Fuzhou, China
| | - Bin Yang
- Department of Laboratory Medicine, the First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, 350005, Fuzhou, China.,Fujian Key Laboratory of Laboratory Medicine, No.20 Chazhong Road, 20 Chazhong Road, 350005, Fuzhou, China.,Gene Diagnostic Laboratory, the First Affiliated Hospital of Fujian Medical University, 350005, Fuzhou, China
| | - Qishui Ou
- Department of Laboratory Medicine, the First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, 350005, Fuzhou, China. .,Fujian Key Laboratory of Laboratory Medicine, No.20 Chazhong Road, 20 Chazhong Road, 350005, Fuzhou, China. .,Gene Diagnostic Laboratory, the First Affiliated Hospital of Fujian Medical University, 350005, Fuzhou, China.
| |
Collapse
|