1
|
García-González P, Puerta R, Cano A, Olivè C, Marquié M, Valero S, Rosende-Roca M, Alegret M, Sanz P, Brosseron F, Martino-Adami P, de Rojas I, Heneka M, Ramírez A, Navarro A, Sáez ME, Tárraga L, Cavazos JE, Boada M, Fernandez MV, Cabrera-Socorro A, Ruiz A. APOE Haplotype Phasing Using ONT Long-Read Sequencing Reveals Two Common ε3 and ε4 intragenic haplotypes in the Spanish Population. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.25.25324541. [PMID: 40196265 PMCID: PMC11974914 DOI: 10.1101/2025.03.25.25324541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Background The apolipoprotein E (APOE) gene is a key genetic determinant of Alzheimer's disease (AD) risk, with the ε4 allele significantly increasing susceptibility. While the pathogenic effects of the ε4 allele are well established, the functional impact of distinct haplotype configurations within the broader ε3 and ε4 backgrounds remains poorly understood. This study investigates the role of intragenic sub haplotypes in modulating APOE expression and their potential influence on AD progression. Methods We utilized Oxford Nanopore Technology (ONT) long-read sequencing to phase variants within a 4-kilobase comprising the APOE locus in a cohort of 1,265 individuals with known APOE genotypes. We evaluated the impact of the identified intragenic haplotypes on APOE protein levels in cerebrospinal fluid (CSF) using the Olink platform, adjusting for demographic and molecular covariates. Statistical modeling was employed to assess the independent effects of these haplotypes alongside traditional APOE genotypes. Additionally, their influence on dementia progression in mild cognitive impairment (MCI) subjects was analyzed using adjusted Cox proportional hazards models. Results Our analysis identified 48 Single Nucleotide Variants (SNVs) within a 4-kilobase region containing the APOE gene, including nine novel variants. Phasing of variants within the APOE locus revealed 59 unique haplotypes in the Spanish population, which were grouped into five major haplogroups-ε2, ε3A, ε3B, ε4A, and ε4B-including two common haplogroups for each of the ε3 and ε4 isoforms. The ε4A haplogroup was associated with a significant decrease in APOE ε4 protein levels in CSF (p = 0.004), suggesting a regulatory mechanism that may mitigate the toxic gain-of-function effect typically attributed to the ε4 allele. Conversely, the ε3B haplogroup was linked to increased APOE ε3 protein levels in ε3/ε4 carriers (p = 0.025), potentially serving a compensatory role.These effects were independent of overall APOE genotype and remained significant after adjusting for covariates. Both haplogroups (ε4A and ε3B) demonstrated protective effects in the progression from MCI to dementia, underscoring their potential relevance in Alzheimer's disease. Conclusions This study provides new insights into the intragenic allelic variability of the APOE gene, demonstrating that intragenic APOE haplogroups within the ε3 and ε4 backgrounds can modulate APOE isoform expression in ways that might modulate AD. Our findings highlight the importance of considering haplotype-specific effects when interpreting the functional impact of APOE and in designing targeted therapeutic strategies. Further research is needed to explore the broader regulatory network of the APOE locus and its interaction with neighboring loci in the 19q13 region.
Collapse
Affiliation(s)
- Pablo García-González
- ACE Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Barcelona, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- PhD program in Biotecnology, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028- Barcelona, Spain
| | - Raquel Puerta
- ACE Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Barcelona, Spain
- PhD program in Biotecnology, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028- Barcelona, Spain
| | - Amanda Cano
- ACE Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Barcelona, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Claudia Olivè
- ACE Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Barcelona, Spain
| | - Marta Marquié
- ACE Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Barcelona, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Sergi Valero
- ACE Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Barcelona, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Maitee Rosende-Roca
- ACE Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Barcelona, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Montserrat Alegret
- ACE Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Barcelona, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Pilar Sanz
- ACE Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Barcelona, Spain
| | - Frederik Brosseron
- Universitätsklinikum Bonn & Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Bonn, Germany
| | - Pamela Martino-Adami
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Itziar de Rojas
- ACE Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Barcelona, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Eschsur-Alzette/Belvaux, Luxembourg
| | - Michael Heneka
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Eschsur-Alzette/Belvaux, Luxembourg
| | - Alfredo Ramírez
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Medical Faculty, 53127 Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
- Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
- Department of Psychiatry and Glenn, Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, San An TX USA
| | - Arcadi Navarro
- Institució Catalana de Recerca i Estudis Avançats (ICREA) and Universitat Pompeu Fabra. Pg. Lluís Companys 23, 08010, Barcelona, Spain
- IBE, Institute of Evolutionary Biology (UPF-CSIC), Department of Medicine and Life Sciences, Universitat Pompeu Fabra. PRBB, C. Doctor Aiguader N88, 08003 Barcelona, Spain
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Av. Doctor Aiguader, N88, 08003 Barcelona, Spain
- BarcelonaBeta Brain Research Center, Pasqual Maragall Foundation, C. Wellington 30, 08005, Barcelona, Spain
| | - María Eugenia Sáez
- ACE Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Barcelona, Spain
- CAEBI, Centro Andaluz de Estudios Bioinformáticos, Sevilla, Spain
| | - Lluís Tárraga
- ACE Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Barcelona, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - José E. Cavazos
- South Texas Medical Science Training Program, University of Texas Health San Antonio, San Antonio, TX, USA
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, San Antonio, TX, USA
| | - Mercè Boada
- ACE Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Barcelona, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | | | | | - Agustín Ruiz
- ACE Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Barcelona, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, San Antonio, TX, USA
- Department of Microbiology, Immunology and Molecular Genetics. Long School of Medicine. University of Texas Health Science Center, San Antonio, TX, USA
| |
Collapse
|
2
|
Bello-Corral L, Seco-Calvo J, Molina Fresno A, González AI, Llorente A, Fernández-Lázaro D, Sánchez-Valdeón L. Prevalence of ApoE Alleles in a Spanish Population of Patients with a Clinical Diagnosis of Alzheimer's Disease: An Observational Case-Control Study. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1941. [PMID: 39768823 PMCID: PMC11679489 DOI: 10.3390/medicina60121941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 10/27/2024] [Accepted: 11/18/2024] [Indexed: 01/11/2025]
Abstract
Background and Objectives: Alzheimer's dementia is a progressive neurodegenerative disease that affects memory abilities due to genetic and environmental factors. A well-known gene that influences the risk of Alzheimer's disease is the apolipoprotein E (APOE) gene. The APOE gene is involved in the production of a protein that helps transport cholesterol and other types of fat in the bloodstream. Problems in this process are thought to contribute to the development of Alzheimer's disease. APOE comes in several forms, which are called alleles (ε2, ε3, ε4). Materials and Methods: Therefore, our study aims to identify those subjects with a higher genetic risk through the polymorphism of the APOE gene, using a population screening in patients with a clinical diagnosis of AD in a region of Spain, Castilla y León, as potential biomarkers and to identify individuals at increased genetic risk by polymorphism of the APOE gene. An observational case-control study was conducted in Castilla y León (Spain). Saliva samples were collected and the ApoE gene was analyzed by PCR and agarose gel electrophoresis, respecting ethical criteria. Results: In the Alzheimer's population in Castilla y León, a high prevalence of ApoE3 (74%) was found, followed by ApoE4 (22%); in addition, a higher presence of the ε4 allele was found in the Alzheimer's disease (AD) group than in the control group. It was also observed that the ε2/ε2 genotype was not found in any individual with AD but was found in healthy subjects and that the opposite was observed for the ε4/ε4 genotype. The odds ratio (OR) indicated a risk four times greater of having AD if having the ε4 allele. Conclusions: The demonstrated relation between the different isoforms and the likelihood of developing AD has led to its consideration as a biomarker and a potential pre-symptomatic therapy. The molecular mechanisms that confer a disruptive and protective role to ApoE4 and ApoE2, respectively, are still being studied.
Collapse
Affiliation(s)
- Laura Bello-Corral
- Health Research Nursing Group (GREIS), University of Leon, 24071 Leon, Spain; (L.B.-C.); (L.S.-V.)
- Department of Nursing and Physiotherapy, University of Leon, 24071 Leon, Spain
| | - Jesús Seco-Calvo
- Institute of Biomedicine, University of León, 24071 Leon, Spain
- Department of Physiology, University of the Basque Country, 48940 Leioa, Spain
| | | | - Ana Isabel González
- Genetics Area, Department of Molecular Biology, University of León, 24071 Leon, Spain; (A.I.G.); (A.L.)
| | - Ana Llorente
- Genetics Area, Department of Molecular Biology, University of León, 24071 Leon, Spain; (A.I.G.); (A.L.)
| | - Diego Fernández-Lázaro
- Department of Cellular Biology, Genetics, Histology and Pharmacology, Faculty of Health Sciences, University of Valladolid, Campus of Soria, 42004 Soria, Spain
- Neurobiology Research Group, Faculty of Medicine, University of Valladolid, 47002 Valladolid, Spain
| | - Leticia Sánchez-Valdeón
- Health Research Nursing Group (GREIS), University of Leon, 24071 Leon, Spain; (L.B.-C.); (L.S.-V.)
- Department of Nursing and Physiotherapy, University of Leon, 24071 Leon, Spain
| |
Collapse
|
3
|
Salian VS, Tang X, Thompson KJ, Curan GL, Lowe VJ, Li L, Kalari KR, Kandimalla KK. Molecular Mechanisms Underlying Amyloid Beta Peptide Mediated Upregulation of Vascular Cell Adhesion Molecule-1 in Alzheimer Disease. J Pharmacol Exp Ther 2024; 391:430-440. [PMID: 39455283 PMCID: PMC11585316 DOI: 10.1124/jpet.124.002280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 09/30/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Amyloid β(Aβ) deposition and neurofibrillary tangles are widely considered the primary pathological hallmarks of familial and sporadic forms of Alzheimer disease (AD). However, cerebrovascular inflammation, which is prevalent in 70% of AD patients, is emerging as another core feature of AD pathology. In our current work, we investigated the hypothesis that Aβ42 exposure drives an increase in vascular cell adhesion molecule-1 (VCAM-1) expression, a cerebrovascular inflammatory marker expressed on the blood-brain barrier (BBB) endothelium in humans and murine models. We have demonstrated that the inflammation signaling pathway is upregulated in AD patient brains, and VCAM-1 expression is increased in AD patients compared with healthy controls. Furthermore, dynamic SPEC/CT imaging in APP,PS1 transgenic mice (a mouse model that overexpresses Aβ42) demonstrated VCAM-1 upregulation at the BBB. Although there is a strong association between Aβ42 exposure and an increase in VCAM-1 expression, the underlying mechanisms remain partially understood. Molecular mechanisms driving VCAM-1 expression at the BBB were investigated in polarized human cerebral microvascular endothelial cell monolayers. Moreover, by employing reverse-phase protein array assays and immunocytochemistry we demonstrated that Aβ42 increases VCAM-1 expression via the Src/p38/MEK signaling pathway. Therefore, targeting the Src/p38/MEK pathway may help modulate VCAM-1 expression at the BBB and help mitigate cerebrovascular inflammation in Alzheimer disease. SIGNIFICANCE STATEMENT: Although considered a core pathological feature of Alzheimer disease, molecular pathways leading to cerebrovascular inflammation remain only partially understood. Moreover, clinical diagnostic methods for detecting cerebrovascular inflammation are underdeveloped. This study demonstrated the detection of VCAM-1 using radio-iodinated VCAM-1 antibody and single-photon emission computed tomography/computed tomography imaging. Additionally, exposure to Aβ42 increases VCAM-1 expression on the blood-brain barrier endothelium via the Src/p38/MEK pathway. These findings are expected to aid in the development of diagnostic and therapeutic approaches for addressing cerebrovascular inflammation in AD.
Collapse
Affiliation(s)
- Vrishali S Salian
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (V.S.S., K.K.K.); Department of Radiology, Mayo Clinic College of Medicine, Rochester, Minnesota (G.L.C., V.J.L.); Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota (X.T., K.J.T., K.R.K.); and Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (L.L.)
| | - Xiaojia Tang
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (V.S.S., K.K.K.); Department of Radiology, Mayo Clinic College of Medicine, Rochester, Minnesota (G.L.C., V.J.L.); Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota (X.T., K.J.T., K.R.K.); and Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (L.L.)
| | - Kevin J Thompson
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (V.S.S., K.K.K.); Department of Radiology, Mayo Clinic College of Medicine, Rochester, Minnesota (G.L.C., V.J.L.); Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota (X.T., K.J.T., K.R.K.); and Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (L.L.)
| | - Geoffry L Curan
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (V.S.S., K.K.K.); Department of Radiology, Mayo Clinic College of Medicine, Rochester, Minnesota (G.L.C., V.J.L.); Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota (X.T., K.J.T., K.R.K.); and Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (L.L.)
| | - Val J Lowe
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (V.S.S., K.K.K.); Department of Radiology, Mayo Clinic College of Medicine, Rochester, Minnesota (G.L.C., V.J.L.); Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota (X.T., K.J.T., K.R.K.); and Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (L.L.)
| | - Ling Li
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (V.S.S., K.K.K.); Department of Radiology, Mayo Clinic College of Medicine, Rochester, Minnesota (G.L.C., V.J.L.); Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota (X.T., K.J.T., K.R.K.); and Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (L.L.)
| | - Krishna R Kalari
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (V.S.S., K.K.K.); Department of Radiology, Mayo Clinic College of Medicine, Rochester, Minnesota (G.L.C., V.J.L.); Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota (X.T., K.J.T., K.R.K.); and Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (L.L.)
| | - Karunya K Kandimalla
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (V.S.S., K.K.K.); Department of Radiology, Mayo Clinic College of Medicine, Rochester, Minnesota (G.L.C., V.J.L.); Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota (X.T., K.J.T., K.R.K.); and Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (L.L.)
| |
Collapse
|
4
|
Shen MQ, Guo Q, Li W, Qian ZM. Apolipoprotein E deficiency leads to the polarization of splenic macrophages towards M1 phenotype by increasing iron content. Genes Immun 2024; 25:381-388. [PMID: 39103538 DOI: 10.1038/s41435-024-00290-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/19/2024] [Accepted: 07/24/2024] [Indexed: 08/07/2024]
Abstract
Apolipoprotein E (ApoE) plays a crucial role in iron homeostasis in the body, while macrophages are the principal cells responsible for handling iron in mammals. However, it is unknown whether ApoE can affect the functional subtypes and the iron handling capacity of splenic macrophages (SM). Here, we investigated the effects of ApoE deficiency (ApoE-/-) on the polarization and iron content of SM and its potential mechanisms. ApoE-/- was found to induce a significant increase in the expressions of M1 marker genes CD86, IL-1β, IL-6, IL-12, TNF-α and iNOS and a reduction in M2 marker genes CD206, Arg-1, IL-10 and Ym-1 in SM of mice aged 28 weeks, Meanwhile, ApoE-/- caused a significant increase in iron content and expression of ferritin, transferrin receptor 1 (TfR1), iron regulatory protein 1 (IRP1) and heme oxygenase-1 (HO-1) and a reduction in ferroportin1 (Fpn1) in spleen and/or SM of mice aged 28 weeks. It was concluded that ApoE-/- can increase iron content through increased iron uptake mediated by TfR/ IRPs and decreased iron release mediated by Fpn1, leading to polarization of the SM to M1 phenotype.
Collapse
Affiliation(s)
- Meng-Qi Shen
- Institute of Translational and Precision Medicine, Nantong University, Nantong, Jiangsu, China
- School of Health Medicine, Nantong Polytechnic College, Nantong, China
| | - Qian Guo
- School of Medicine, Shanghai University, Shanghai, China.
| | - Wei Li
- Institute of Translational and Precision Medicine, Nantong University, Nantong, Jiangsu, China
| | - Zhong-Ming Qian
- Institute of Translational and Precision Medicine, Nantong University, Nantong, Jiangsu, China.
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
5
|
Kausar G, Chauhan SB, Roy R, Kumar S, Engwerda C, Nylen S, Kumar R, Wilson ME, Sundar S. Apolipoprotein E Is Upregulated in Blood and Circulating Monocytes of Indian Patients With Visceral Leishmaniasis. Parasite Immunol 2024; 46:e13036. [PMID: 38720445 PMCID: PMC11141729 DOI: 10.1111/pim.13036] [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: 11/29/2023] [Revised: 04/17/2024] [Accepted: 04/22/2024] [Indexed: 05/18/2024]
Abstract
Apolipoprotein E (ApoE) has been associated with several diseases including Parkinson's disease, Alzheimer's and multiple sclerosis. ApoE also has documented immunomodulatory functions. We investigated gene expression in circulating monocytes and in bone marrows of patients with visceral leishmaniasis (VL) living in an endemic area in Bihar, India, and contrasted these with control healthy subjects or other diagnostic bone marrows from individuals in the same region. Samples from VL patients were obtained prior to initiating treatment. Our study revealed significant upregulated expression of the apoE transcript in patients with VL. Furthermore, the levels of ApoE protein were elevated in serum samples of subjects with VL compared with healthy endemic controls. These observations may provide clues regarding the complex interactions between lipid metabolism and immunoregulation of infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Gulafsha Kausar
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi-221005, UP, India
| | - Shashi Bhushan Chauhan
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi-221005, UP, India
- Department of Microbiology, Immunology & Tropical Medicine, School of Medicine and Health Sciences, The George Washington University, Washington D.C. 20052, USA
| | - Ritirupa Roy
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi-221005, UP, India
| | - Shashi Kumar
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi-221005, UP, India
| | - Christian Engwerda
- QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia
| | - Susanne Nylen
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Rajiv Kumar
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi-221005, UP, India
| | - Mary E. Wilson
- Departments of Internal Medicine and Microbiology & Immunology, University of Iowa, Iowa City, IA 52242, USA
- Veterans’ Affairs Medical Center, Iowa City, IA 52240, USA
| | - Shyam Sundar
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi-221005, UP, India
| |
Collapse
|
6
|
Ramachandran AK, Das S, Shenoy GG, Mudgal J, Joseph A. Relation between Apolipoprotein E in Alzheimer's Disease and SARS-CoV-2 and their Treatment Strategy: A Review. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:9-20. [PMID: 36573058 DOI: 10.2174/1871527322666221226145141] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 10/27/2022] [Accepted: 11/01/2022] [Indexed: 12/28/2022]
Abstract
COVID-19, which primarily affects the pulmonary system, turned out to be a global pandemic, whereas the effects on other systems are still unknown. SARS-CoV-2, binds to angiotensinconverting enzyme 2 (ACE2) receptors in the lungs, causing pneumonia-like symptoms. The same ACE receptors are also present in organs other than the lungs. Therefore, there is a need to study the impact of coronavirus on other human body organs. Recently, UK Biobank reports on the genetic risk factor of the virus attack. A double mutation in the apolipoprotein E (APOE4) allele has shown a significant role in COVID-19. The same APOE4 mutation has already been proven to hold a key role in developing early-onset Alzheimer's disease (EOAD). Despite this data, Alzheimer's disease is believed to be a comorbidity of COVID-19. Previous virus attacks on the same viral family, Coronaviridae, produced neurological effects like neurodegeneration, neuronal inflammation, and other central nervous system-related dysfunctions. Since the long-term implications of COVID-19 are unknown, more research into the impact of the virus on the central nervous system is needed. Both COVID-19 and AD share a common genetic factor, so that AD patients may have a greater risk of SARS-CoV-2. Here, in this review, we have briefly discussed the role of APOE4 in the pathogenesis of AD and SARS-CoV-2, along with their treatment strategy, current scenario, and possible future directions.
Collapse
Affiliation(s)
- Anu Kunnath Ramachandran
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Subham Das
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
- Manipal McGill Centre for Infectious Diseases, Prasanna School of Public Health, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Gurupur Gautham Shenoy
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Jayesh Mudgal
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Alex Joseph
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| |
Collapse
|
7
|
Ji X, Peng X, Tang H, Pan H, Wang W, Wu J, Chen J, Wei N. Alzheimer's disease phenotype based upon the carrier status of the apolipoprotein E ɛ4 allele. Brain Pathol 2024; 34:e13208. [PMID: 37646624 PMCID: PMC10711266 DOI: 10.1111/bpa.13208] [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: 12/06/2022] [Accepted: 08/05/2023] [Indexed: 09/01/2023] Open
Abstract
The apolipoprotein E ɛ4 allele (APOE4) is universally acknowledged as the most potent genetic risk factor for Alzheimer's disease (AD). APOE4 promotes the initiation and progression of AD. Although the underlying mechanisms are unclearly understood, differences in lipid-bound affinity among the three APOE isoforms may constitute the basis. The protein APOE4 isoform has a high affinity with triglycerides and cholesterol. A distinction in lipid metabolism extensively impacts neurons, microglia, and astrocytes. APOE4 carriers exhibit phenotypic differences from non-carriers in clinical examinations and respond differently to multiple treatments. Therefore, we hypothesized that phenotypic classification of AD patients according to the status of APOE4 carrier will help specify research and promote its use in diagnosing and treating AD. Recent reviews have mainly evaluated the differences between APOE4 allele carriers and non-carriers from gene to protein structures, clinical features, neuroimaging, pathology, the neural network, and the response to various treatments, and have provided the feasibility of phenotypic group classification based on APOE4 carrier status. This review will facilitate the application of APOE phenomics concept in clinical practice and promote further medical research on AD.
Collapse
Affiliation(s)
- Xiao‐Yu Ji
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeGuangdongChina
- Brain Function and Disease LaboratoryShantou University Medical CollegeGuangdongChina
| | - Xin‐Yuan Peng
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeGuangdongChina
| | - Hai‐Liang Tang
- Fudan University Huashan Hospital, Department of Neurosurgery, State Key Laboratory for Medical NeurobiologyInstitutes of Brain Science, Shanghai Medical College‐Fudan UniversityShanghaiChina
| | - Hui Pan
- Shantou Longhu People's HospitalShantouGuangdongChina
| | - Wei‐Tang Wang
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeGuangdongChina
| | - Jie Wu
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeGuangdongChina
- Brain Function and Disease LaboratoryShantou University Medical CollegeGuangdongChina
| | - Jian Chen
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeGuangdongChina
| | - Nai‐Li Wei
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeGuangdongChina
| |
Collapse
|
8
|
Laskowitz DT, Van Wyck DW. ApoE Mimetic Peptides as Therapy for Traumatic Brain Injury. Neurotherapeutics 2023; 20:1496-1507. [PMID: 37592168 PMCID: PMC10684461 DOI: 10.1007/s13311-023-01413-0] [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] [Accepted: 07/17/2023] [Indexed: 08/19/2023] Open
Abstract
The lack of targeted therapies for traumatic brain injury (TBI) remains a compelling clinical unmet need. Although knowledge of the pathophysiologic cascades involved in TBI has expanded rapidly, the development of novel pharmacological therapies has remained largely stagnant. Difficulties in creating animal models that recapitulate the different facets of clinical TBI pathology and flaws in the design of clinical trials have contributed to the ongoing failures in neuroprotective drug development. Furthermore, multiple pathophysiological mechanisms initiated early after TBI that progress in the subacute and chronic setting may limit the potential of traditional approaches that target a specific cellular pathway for acute therapeutic intervention. We describe a reverse translational approach that focuses on translating endogenous mechanisms known to influence outcomes after TBI to develop druggable targets. In particular, numerous clinical observations have demonstrated an association between apolipoprotein E (apoE) polymorphism and functional recovery after brain injury. ApoE has been shown to mitigate the response to acute brain injury by exerting immunomodulatory properties that reduce secondary tissue injury as well as protecting neurons from excitotoxicity. CN-105 represents an apoE mimetic peptide that can effectively penetrate the CNS compartment and retains the neuroprotective properties of the intact protein.
Collapse
Affiliation(s)
- Daniel T Laskowitz
- Department of Neurology, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Neurobiology, Duke University School of Medicine, Durham, NC, 27710, USA
- AegisCN LLC, 701 W Main Street, Durham, NC, 27701, USA
| | - David W Van Wyck
- Department of Neurology, Duke University School of Medicine, Durham, NC, 27710, USA.
| |
Collapse
|
9
|
Sex and APOE Genotype Alter the Basal and Induced Inflammatory States of Primary Microglia from APOE Targeted Replacement Mice. Int J Mol Sci 2022; 23:ijms23179829. [PMID: 36077227 PMCID: PMC9456163 DOI: 10.3390/ijms23179829] [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: 07/20/2022] [Revised: 08/17/2022] [Accepted: 08/19/2022] [Indexed: 11/16/2022] Open
Abstract
The sex and APOE4 genotype are significant risk factors for Alzheimer’s disease (AD); however, the mechanism(s) responsible for this interaction are still a matter of debate. Here, we assess the responses of mixed-sex and sex-specific APOE3 and APOE4 primary microglia (PMG) to lipopolysaccharide and interferon-gamma. In our investigation, inflammatory cytokine profiles were assessed by qPCR and multiplex ELISA assays. Mixed-sex APOE4 PMG exhibited higher basal mRNA expression and secreted levels of TNFa and IL1b. In sex-specific cultures, basal expression and secreted levels of IL1b, TNFa, IL6, and NOS2 were 2−3 fold higher in APOE4 female PMG compared to APOE4 males, with both higher than APOE3 cells. Following an inflammatory stimulus, the expression of pro-inflammatory cytokines and the secreted cytokine level were upregulated in the order E4 female > E4 male > E3 female > E3 male in sex-specific cultures. These data indicate that the APOE4 genotype and female sex together contribute to a greater inflammatory response in PMG isolated from targeted replacement humanized APOE mice. These data are consistent with clinical data and indicate that sex-specific PMG may provide a platform for exploring mechanisms of genotype and sex differences in AD related to neuroinflammation and neurodegeneration.
Collapse
|
10
|
Neuronal ApoE4 stimulates C/EBPβ activation, promoting Alzheimer’s disease pathology in a mouse model. Prog Neurobiol 2022; 209:102212. [DOI: 10.1016/j.pneurobio.2021.102212] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 12/05/2021] [Accepted: 12/22/2021] [Indexed: 12/15/2022]
|
11
|
Buchanan H, Hull C, Cacho Barraza M, Delibegovic M, Platt B. Apolipoprotein E loss of function: Influence on murine brain markers of physiology and pathology. AGING BRAIN 2022; 2:100055. [PMID: 36908879 PMCID: PMC9997145 DOI: 10.1016/j.nbas.2022.100055] [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: 06/23/2022] [Revised: 10/11/2022] [Accepted: 10/13/2022] [Indexed: 12/15/2022] Open
Abstract
The canonical role of Apolipoprotein E (ApoE) is related to lipid and cholesterol metabolism, however, additional functions of this protein have not been fully described. Given the association of ApoE with diseases such as Alzheimer's Disease (AD), it is clear that further characterisation of its roles, especially within the brain, is needed. Therefore, using protein and gene expression analyses of neonatal and 6-month old brain tissues from an ApoE knockout mouse model, we examined ApoE's contribution to several CNS pathways, with an emphasis on those linked to AD. Early neonatal changes associated with ApoE-/- were observed, with decreased soluble phosphorylated tau (p-tau, -40 %), increased synaptophysin (+36 %) and microglial Iba1 protein levels (+25 %) vs controls. Progression of the phenotype was evident upon analysis of 6-month-old tissue, where decreased p-tau was also confirmed in the insoluble fraction, alongside reduced synaptic and increased amyloid precursor protein (APP) protein levels. An age comparison further underlined deviations from WT animals and thus the impact of ApoE loss on neuronal maturation. Taken together, our data implicate ApoE modulation of multiple CNS roles. Loss of function is associated with alterations from birth, and include synaptic deficits, neuroinflammation, and changes to key AD pathologies, amyloid-β and tau.
Collapse
Affiliation(s)
| | | | | | | | - Bettina Platt
- Corresponding author at: Chair in Translational Neuroscience, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, Scotland, UK.
| |
Collapse
|
12
|
Smailovic U, Johansson C, Koenig T, Kåreholt I, Graff C, Jelic V. Decreased Global EEG Synchronization in Amyloid Positive Mild Cognitive Impairment and Alzheimer's Disease Patients-Relationship to APOE ε4. Brain Sci 2021; 11:brainsci11101359. [PMID: 34679423 PMCID: PMC8533770 DOI: 10.3390/brainsci11101359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 11/16/2022] Open
Abstract
The apolipoprotein E (APOE) ε4 allele is a risk factor for Alzheimer's disease (AD) that has been linked to changes in brain structure and function as well as to different biological subtypes of the disease. The present study aimed to investigate the association of APOE ε4 genotypes with brain functional impairment, as assessed by quantitative EEG (qEEG) in patients on the AD continuum. The study population included 101 amyloid positive patients diagnosed with mild cognitive impairment (MCI) (n = 50) and AD (n = 51) that underwent resting-state EEG recording and CSF Aβ42 analysis. In total, 31 patients were APOE ε4 non-carriers, 42 were carriers of one, and 28 were carriers of two APOE ε4 alleles. Quantitative EEG analysis included computation of the global field power (GFP) and global field synchronization (GFS) in conventional frequency bands. Amyloid positive patients who were carriers of APOE ε4 allele(s) had significantly higher GFP beta and significantly lower GFS in theta and beta bands compared to APOE ε4 non-carriers. Increased global EEG power in beta band in APOE ε4 carriers may represent a brain functional compensatory mechanism that offsets global EEG slowing in AD patients. Our findings suggest that decreased EEG measures of global synchronization in theta and beta bands reflect brain functional deficits related to the APOE ε4 genotype in patients that are on a biomarker-verified AD continuum.
Collapse
Affiliation(s)
- Una Smailovic
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 14152 Huddinge, Sweden;
- Department of Clinical Neurophysiology, Karolinska University Hospital, 14186 Huddinge, Sweden
- Correspondence:
| | - Charlotte Johansson
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 14152 Huddinge, Sweden; (C.J.); (C.G.)
- Clinic for Cognitive Disorders, Karolinska University Hospital, 14186 Huddinge, Sweden
| | - Thomas Koenig
- Translational Research Center, University Hospital of Psychiatry, University of Bern, 3012 Bern, Switzerland;
| | - Ingemar Kåreholt
- Aging Research Centre, Karolinska Institutet and Stockholm University, 17165 Solna, Sweden;
- School of Health and Welfare, Aging Research Network—Jönköping (ARN-J), Institute for Gerontology, Jönköping University, 55111 Jönköping, Sweden
| | - Caroline Graff
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 14152 Huddinge, Sweden; (C.J.); (C.G.)
- Unit for Hereditary Dementia, Karolinska University Hospital-Solna, 17176 Solna, Sweden
| | - Vesna Jelic
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 14152 Huddinge, Sweden;
- Clinic for Cognitive Disorders, Karolinska University Hospital, 14186 Huddinge, Sweden
| |
Collapse
|
13
|
Unique molecular characteristics and microglial origin of Kv1.3 channel-positive brain myeloid cells in Alzheimer's disease. Proc Natl Acad Sci U S A 2021; 118:2013545118. [PMID: 33649184 DOI: 10.1073/pnas.2013545118] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Kv1.3 potassium channels, expressed by proinflammatory central nervous system mononuclear phagocytes (CNS-MPs), are promising therapeutic targets for modulating neuroinflammation in Alzheimer's disease (AD). The molecular characteristics of Kv1.3-high CNS-MPs and their cellular origin from microglia or CNS-infiltrating monocytes are unclear. While Kv1.3 blockade reduces amyloid beta (Aβ) burden in mouse models, the downstream immune effects on molecular profiles of CNS-MPs remain unknown. We show that functional Kv1.3 channels are selectively expressed by a subset of CD11b+CD45+ CNS-MPs acutely isolated from an Aβ mouse model (5xFAD) as well as fresh postmortem human AD brain. Transcriptomic profiling of purified CD11b+Kv1.3+ CNS-MPs, CD11b+CD45int Kv1.3neg microglia, and peripheral monocytes from 5xFAD mice revealed that Kv1.3-high CNS-MPs highly express canonical microglial markers (Tmem119, P2ry12) and are distinct from peripheral Ly6chigh/Ly6clow monocytes. Unlike homeostatic microglia, Kv1.3-high CNS-MPs express relatively lower levels of homeostatic genes, higher levels of CD11c, and increased levels of glutamatergic transcripts, potentially representing phagocytic uptake of neuronal elements. Using irradiation bone marrow CD45.1/CD45.2 chimerism in 5xFAD mice, we show that Kv1.3+ CNS-MPs originate from microglia and not blood-derived monocytes. We show that Kv1.3 channels regulate membrane potential and early signaling events in microglia. Finally, in vivo blockade of Kv1.3 channels in 5xFAD mice by ShK-223 reduced Aβ burden, increased CD11c+ CNS-MPs, and expression of phagocytic genes while suppressing proinflammatory genes (IL1b). Our results confirm the microglial origin and identify unique molecular features of Kv1.3-expressing CNS-MPs. In addition, we provide evidence for CNS immunomodulation by Kv1.3 blockers in AD mouse models resulting in a prophagocytic phenotype.
Collapse
|
14
|
Desimone A, Hong J, Brockie ST, Yu W, Laliberte AM, Fehlings MG. The influence of ApoE4 on the clinical outcomes and pathophysiology of degenerative cervical myelopathy. JCI Insight 2021; 6:e149227. [PMID: 34369386 PMCID: PMC8410082 DOI: 10.1172/jci.insight.149227] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 06/30/2021] [Indexed: 11/17/2022] Open
Abstract
Degenerative cervical myelopathy (DCM) is the most common cause of nontraumatic spinal cord injury in adults worldwide. Surgical decompression is generally effective in improving neurological outcomes and halting progression of myelopathic deterioration. However, a subset of patients experience suboptimal neurological outcomes. Given the emerging evidence that apolipoprotein E4 (ApoE4) allelic status influences neurodegenerative conditions, we examined whether the presence of the ApoE4 allele may account for the clinical heterogeneity of treatment outcomes in patients with DCM. Our results demonstrate that human ApoE4+ DCM patients have a significantly lower extent of improvement after decompression surgery. Functional analysis of our DCM mouse model in targeted-replacement mice expressing human ApoE4 revealed delayed gait recovery, forelimb grip strength, and hind limb mechanical sensitivity after decompression surgery, compared with their ApoE3 counterparts. This was accompanied by an exacerbated proinflammatory response resulting in higher concentrations of TNF-α, IL-6, CCL3, and CXCL9. At the site of injury, there was a significant decrease in gray matter area, an increase in the activation of microglia/macrophages, and increased astrogliosis after decompression surgery in the ApoE4 mice. Our study is the first to our knowledge to investigate the pathophysiological underpinnings of ApoE4 in DCM, which suggests a possible personalized medicine approach for the treatment of DCM in ApoE4 carriers.
Collapse
Affiliation(s)
- Alexa Desimone
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Sciences
| | - James Hong
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Sciences
| | - Sydney T Brockie
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Sciences
| | - Wenru Yu
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Alex M Laliberte
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Sciences
| | - Michael G Fehlings
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Sciences.,Division of Neurosurgery, Department of Surgery, and.,Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
15
|
Bianchi L, Sframeli M, Vantaggiato L, Vita GL, Ciranni A, Polito F, Oteri R, Gitto E, Di Giuseppe F, Angelucci S, Versaci A, Messina S, Vita G, Bini L, Aguennouz M. Nusinersen Modulates Proteomics Profiles of Cerebrospinal Fluid in Spinal Muscular Atrophy Type 1 Patients. Int J Mol Sci 2021; 22:ijms22094329. [PMID: 33919289 PMCID: PMC8122268 DOI: 10.3390/ijms22094329] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 04/19/2021] [Indexed: 02/06/2023] Open
Abstract
Spinal muscular atrophy (SMA) type 1 is a severe infantile autosomal-recessive neuromuscular disorder caused by a survival motor neuron 1 gene (SMN1) mutation and characterized by progressive muscle weakness. Without supportive care, SMA type 1 is rapidly fatal. The antisense oligonucleotide nusinersen has recently improved the natural course of this disease. Here, we investigated, with a functional proteomic approach, cerebrospinal fluid (CSF) protein profiles from SMA type 1 patients who underwent nusinersen administration to clarify the biochemical response to the treatment and to monitor disease progression based on therapy. Six months after starting treatment (12 mg/5 mL × four doses of loading regimen administered at days 0, 14, 28, and 63), we observed a generalized reversion trend of the CSF protein pattern from our patient cohort to that of control donors. Notably, a marked up-regulation of apolipoprotein A1 and apolipoprotein E and a consistent variation in transthyretin proteoform occurrence were detected. Since these multifunctional proteins are critically active in biomolecular processes aberrant in SMA, i.e., synaptogenesis and neurite growth, neuronal survival and plasticity, inflammation, and oxidative stress control, their nusinersen induced modulation may support SMN improved-expression effects. Hence, these lipoproteins and transthyretin could represent valuable biomarkers to assess patient responsiveness and disease progression.
Collapse
Affiliation(s)
- Laura Bianchi
- Functional Proteomics Laboratory, Department of Life Sciences, University of Siena, 53100 Siena, Italy; (L.B.); (L.V.); (L.B.)
| | - Maria Sframeli
- Nemo Sud Clinical Centre, 98125 Messina, Italy; (M.S.); (G.L.V.)
| | - Lorenza Vantaggiato
- Functional Proteomics Laboratory, Department of Life Sciences, University of Siena, 53100 Siena, Italy; (L.B.); (L.V.); (L.B.)
| | - Gian Luca Vita
- Nemo Sud Clinical Centre, 98125 Messina, Italy; (M.S.); (G.L.V.)
| | - Annamaria Ciranni
- Unit of Neurology and Neuromuscular Diseases, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (A.C.); (F.P.); (R.O.); (S.M.); (M.A.)
| | - Francesca Polito
- Unit of Neurology and Neuromuscular Diseases, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (A.C.); (F.P.); (R.O.); (S.M.); (M.A.)
| | - Rosaria Oteri
- Unit of Neurology and Neuromuscular Diseases, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (A.C.); (F.P.); (R.O.); (S.M.); (M.A.)
| | - Eloisa Gitto
- Neonatal and Paediatric Intensive Care Unit, Department of Human Pathology in Adult and Developmental Age, University of Messina, 98125 Messina, Italy;
| | - Fabrizio Di Giuseppe
- Dentistry and Biotechnology, and Proteomics Unit, Centre of Advanced Studies and Technoloy, Department Medical, Oral & Biotechnological Sciences, “G. d’Annunzio”, University of Chieti-Pescara, 66100 Chieti, Italy; (F.D.G.); (S.A.)
| | - Stefania Angelucci
- Dentistry and Biotechnology, and Proteomics Unit, Centre of Advanced Studies and Technoloy, Department Medical, Oral & Biotechnological Sciences, “G. d’Annunzio”, University of Chieti-Pescara, 66100 Chieti, Italy; (F.D.G.); (S.A.)
| | - Antonio Versaci
- Intensive Care Unit, AOU Policlinico “G. Martino”, 98125 Messina, Italy;
| | - Sonia Messina
- Unit of Neurology and Neuromuscular Diseases, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (A.C.); (F.P.); (R.O.); (S.M.); (M.A.)
| | - Giuseppe Vita
- Nemo Sud Clinical Centre, 98125 Messina, Italy; (M.S.); (G.L.V.)
- Unit of Neurology and Neuromuscular Diseases, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (A.C.); (F.P.); (R.O.); (S.M.); (M.A.)
- Correspondence:
| | - Luca Bini
- Functional Proteomics Laboratory, Department of Life Sciences, University of Siena, 53100 Siena, Italy; (L.B.); (L.V.); (L.B.)
| | - M’hammed Aguennouz
- Unit of Neurology and Neuromuscular Diseases, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (A.C.); (F.P.); (R.O.); (S.M.); (M.A.)
| |
Collapse
|
16
|
Wang ZH, Xia Y, Liu P, Liu X, Edgington-Mitchell L, Lei K, Yu SP, Wang XC, Ye K. ApoE4 activates C/EBPβ/δ-secretase with 27-hydroxycholesterol, driving the pathogenesis of Alzheimer's disease. Prog Neurobiol 2021; 202:102032. [PMID: 33716161 DOI: 10.1016/j.pneurobio.2021.102032] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 01/17/2021] [Accepted: 03/09/2021] [Indexed: 02/06/2023]
Abstract
ApoE4, an apolipoprotein implicated in cholesterol transport and amyloid-β (Aβ) metabolism, is a major genetic risk determinant for Alzheimer's Disease (AD) and drives its pathogenesis via Aβ-dependent and -independent pathways. C/EBPβ, a proinflammatory cytokines-activated transcription factor, is upregulated in AD and mediates cytokines and δ-secretase expression. However, how ApoE4 contributes to AD pathogenesis remains incompletely understood. Here we show that ApoE4 and 27-hydroxycholesterol (27-OHC) co-activate C/EBPβ/δ-secretase signaling in neurons, mediating AD pathogenesis, and this effect is dependent on neuronal secreted Aβ and inflammatory cytokines. Inhibition of cholesterol metabolism with lovastatin diminishes neuronal ApoE4's stimulatory effects. Furthermore, ApoE4 and 27-OHC also mediate lysosomal δ-secretase leakage, activation, secretion and endocytosis. Notably, 27-OHC strongly activates C/EBPβ/δ-secretase pathway in human ApoE4-TR mice and triggers AD pathologies and cognitive deficits, which is blocked by C/EBPβ depletion. Hence, our findings demonstrate that ApoE4 and 27-OHC additively trigger AD pathogenesis via activating C/EBPβ/δ-secretase pathway. Lowering cholesterol levels with statins should benefit the ApoE4 AD carriers.
Collapse
Affiliation(s)
- Zhi-Hao Wang
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, 30322, USA; Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yiyuan Xia
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, 30322, USA; Department of Pathophysiology, Key Laboratory of Ministry of Education of Neurological Diseases, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pai Liu
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, 30322, USA; Neuroscience Graduate Program, Laney Graduate School, Emory University, Atlanta, GA, 30322, USA
| | - Xia Liu
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Laura Edgington-Mitchell
- Department of Biochemistry and Molecular Biology, The University of Melbourne, Melbourne Victoria, 3010, Australia
| | - Kecheng Lei
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Shan Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Xiao-Chuan Wang
- Department of Pathophysiology, Key Laboratory of Ministry of Education of Neurological Diseases, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, JS, 226001, China.
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
17
|
Ma J, Qian C, Bao Y, Liu MY, Ma HM, Shen MQ, Li W, Wang JJ, Bao YX, Liu Y, Ke Y, Qian ZM. Apolipoprotein E deficiency induces a progressive increase in tissue iron contents with age in mice. Redox Biol 2021; 40:101865. [PMID: 33493903 PMCID: PMC7823209 DOI: 10.1016/j.redox.2021.101865] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/10/2021] [Accepted: 01/10/2021] [Indexed: 12/21/2022] Open
Abstract
Association of both iron/hepcidin and apolipoprotein E (ApoE) with development of Alzheimer disease (AD) and atherosclerosis led us to hypothesize that ApoE might be required for body iron homeostasis. Here, we demonstrated that ApoE knock-out (KO) induced a progressive accumulation of iron with age in the liver and spleen of mice. Subsequent investigations showed that the increased iron in the liver and spleen was due to phosphorylated extracellular regulated protein kinases (pERK) mediated up-regulation of transferrin receptor 1 (TfR1), and nuclear factor erythroid 2-related factor-2 (Nrf2)-dependent down-regulation of ferroportin 1. Furthermore, replenishment of ApoE could partially reverse the iron-related phenotype in ApoE KO mice. The findings imply that ApoE may be essential for body iron homeostasis and also suggest that clinical late-onset diseases with unexplained iron abnormality may partly be related to deficiency or reduced expression of ApoE. Apolipoprotein E deficiency induces a progressive increase in tissue iron contents with age in mice. ApoE−/− induced a progressive accumulation of iron with age in the liver and spleen of mice. The increased iron was due to upregulation of TfR1 and downregulation of Fpn1. Replenishment of ApoE could partially reverse the iron-related phenotype in ApoE KO mice. ApoE may be essential for body iron homeostasis.
Collapse
Affiliation(s)
- Juan Ma
- Institute of Translational and Precision Medicine, Nantong University, 19 Qi Xiu Road, Nantong, 226001, China; Laboratory of Neuropharmacology of Pharmacy School, and National Clinical Research Center for Aging and Medicine of Huashan Hospital, Fudan University, Shanghai, 201203, China
| | - Christopher Qian
- School of Biomedical Sciences and Gerald Choa Neuroscience Centre, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong, China
| | - Yong Bao
- Institute of Translational and Precision Medicine, Nantong University, 19 Qi Xiu Road, Nantong, 226001, China
| | - Meng-Yue Liu
- Institute of Translational and Precision Medicine, Nantong University, 19 Qi Xiu Road, Nantong, 226001, China
| | - Hui-Min Ma
- Institute of Translational and Precision Medicine, Nantong University, 19 Qi Xiu Road, Nantong, 226001, China
| | - Meng-Qi Shen
- Institute of Translational and Precision Medicine, Nantong University, 19 Qi Xiu Road, Nantong, 226001, China
| | - Wei Li
- Institute of Translational and Precision Medicine, Nantong University, 19 Qi Xiu Road, Nantong, 226001, China
| | - Jiao-Jiao Wang
- Laboratory of Neuropharmacology of Pharmacy School, and National Clinical Research Center for Aging and Medicine of Huashan Hospital, Fudan University, Shanghai, 201203, China; Research Center for Medicine and Biology, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Yu-Xin Bao
- Research Center for Medicine and Biology, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Yong Liu
- Department of Pain and Rehabilitation, The Second Affiliated Hospital, The Army Medical University, Chongqing, China
| | - Ya Ke
- School of Biomedical Sciences and Gerald Choa Neuroscience Centre, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong, China.
| | - Zhong-Ming Qian
- Institute of Translational and Precision Medicine, Nantong University, 19 Qi Xiu Road, Nantong, 226001, China; Laboratory of Neuropharmacology of Pharmacy School, and National Clinical Research Center for Aging and Medicine of Huashan Hospital, Fudan University, Shanghai, 201203, China.
| |
Collapse
|
18
|
Exercise as Potential Therapeutic Target to Modulate Alzheimer's Disease Pathology in APOE ε4 Carriers: A Systematic Review. Cardiol Ther 2021; 10:67-88. [PMID: 33403644 PMCID: PMC8126521 DOI: 10.1007/s40119-020-00209-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease for which no effective treatment exists at present. Previous research has found that exercise reduces the risk of AD. Since the apolipoprotein E (APOE) ε4 allele increases the risk of AD and is associated with faster disease progression than the other isoforms, we aimed to highlight the impact of exercise on AD pathology in APOE ε4 carriers. This review focuses on the effect of exercise on cognitive function, dementia risk, amyloid-β (Aβ) metabolism, lipid metabolism, neuroinflammation, neurotrophic factors and vascularization in APOE ε4 carriers. We searched the literature in the PubMed electronic database using the following search terms: physical activity, exercise, aerobic fitness, training, sport, APOE4, Alzheimer's disease, AD and dementia. By cross-referencing, additional publications were identified. Selected studies required older adults to take part in an exercise intervention or to make use of self-reported physical activity questionnaires. All included studies were written and published in English between 2000 and 2020. From these studies, we conclude that exercise is a non-pharmacological treatment option for high-risk APOE ε4 carriers to ameliorate the AD pathological processes including reducing Aβ load, protecting against hippocampal atrophy, improving cognitive function, stabilizing cholesterol levels and lowering pro-inflammatory signals. Variation in study design related to age, cognitive outcomes and the type of intervention explained the differences in study outcomes. However, exercise seems to be effective in delaying the onset of AD and may improve the quality of life of AD patients.
Collapse
|
19
|
Ayton S, Janelidze S, Roberts B, Palmqvist S, Kalinowski P, Diouf I, Belaidi AA, Stomrud E, Bush AI, Hansson O. Acute phase markers in CSF reveal inflammatory changes in Alzheimer's disease that intersect with pathology, APOE ε4, sex and age. Prog Neurobiol 2020; 198:101904. [PMID: 32882319 DOI: 10.1016/j.pneurobio.2020.101904] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 07/28/2020] [Accepted: 08/03/2020] [Indexed: 01/31/2023]
Abstract
It is unknown how neuroinflammation may feature in the etiology of Alzheimer's disease (AD). We profiled acute phase response (APR) proteins (α1-antitrypsin, α1-antichymotrypsin, ceruloplasmin, complement C3, ferritin, α-fibrinogen, β-fibrinogen, γ-fibrinogen, haptoglobin, hemopexin) in CSF of 1291 subjects along the clinical and biomarker spectrum of AD to investigate the association between inflammatory changes, disease outcomes, and demographic variables. Subjects were stratified by Aβ42/t-tau as well as the following clinical diagnoses: cognitively normal (CN); subjective cognitive decline (SCD); mild cognitive impairment (MCI); and AD dementia. In separate multiple regressions (adjusting for diagnosis, age, sex, APOE-ε4) of each APR protein and a composite of all APR proteins, CSF Aβ42/t-tau status was associated with elevated ferritin, but not any other APR protein in CN and SCD subjects. Rather, the APR was elevated along with symptomatic progression (CN < SCD < MCI < AD), and this was elevation was mediated by CSF p-tau181. APOE ε4 status did not affect levels of any APR proteins in CSF, while these were elevated in males and with increased age. The performance of the APR in predicting clinical diagnosis was influenced by APOE ε4 status, sex, and age. These data provide new insight into inflammatory changes in AD and how this intersects with pathology changes and patient demographics.
Collapse
Affiliation(s)
- Scott Ayton
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Blaine Roberts
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Pawel Kalinowski
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | - Ibrahima Diouf
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | - Abdel A Belaidi
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Ashley I Bush
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia.
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
20
|
Sullivan P. Influence of Western diet and APOE genotype on Alzheimer's disease risk. Neurobiol Dis 2020; 138:104790. [DOI: 10.1016/j.nbd.2020.104790] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 01/28/2020] [Accepted: 02/03/2020] [Indexed: 10/25/2022] Open
|
21
|
Du S, Wang H, Cai J, Ren R, Zhang W, Wei W, Shen X. Apolipoprotein E2 modulates cell cycle function to promote proliferation in pancreatic cancer cells via regulation of the c-Myc–p21Waf1signalling pathway. Biochem Cell Biol 2020; 98:191-202. [PMID: 32167787 DOI: 10.1139/bcb-2018-0230] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Apolipoprotein E2 (ApoE2) is reportedly critical for cell proliferation and survival, and has been identified as a potential tumour-associated marker in many kinds of cancer. However, studies of the function and mechanisms of ApoE2 in pancreatic cancer proliferation and development are rare. In this study, we performed an analysis to determine the modulatory effects of ApoE2–LRP8 (lipoprotein receptor-related protein 8) pathway on cell cycle and cell proliferation, and explored its mechanisms in pancreatic cancer. High expression levels of ApoE2–LRP8/c-Myc were detected in tumour tissues and cell lines by immunohistochemistry and Western blotting. It was also shown that ApoE2–LRP8 induced phosphorylation of ERK1/2 to activate c-Myc and contribute to cell-cycle-related protein expression. ApoE2 conditions induced c-Myc binding to target gene sequences in the p21Waf1promoter, resulting in decreased transcription. ERK/c-Myc contributes to the promotion of the expression levels of cyclin D1, cdc2, and cyclin B1, and reduces p21Waf1activity, thereby promoting cell cycle distribution. We demonstrated the function of ApoE2–LRP8 in the activation of the ERK–c-Myc–p21Waf1signalling cascade and the modulation of G1/S and G2/M transition, indicating ApoE2–LRP8’s important role in the cancer cell proliferation. ApoE2 could serve as a diagnostic marker and chemotherapeutic target in pancreatic cancer.
Collapse
Affiliation(s)
- Shaoxia Du
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Hui Wang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Jun Cai
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Runling Ren
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Wenwen Zhang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Wei Wei
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huanhu West Road, Tianjin 300060, China
| | - Xiaohong Shen
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| |
Collapse
|
22
|
Monroy E, Diaz A, Tendilla-Beltrán H, de la Cruz F, Flores G. Bexarotene treatment increases dendritic length in the nucleus accumbens without change in the locomotor activity and memory behaviors, in old mice. J Chem Neuroanat 2019; 104:101734. [PMID: 31887346 DOI: 10.1016/j.jchemneu.2019.101734] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 11/28/2019] [Accepted: 12/23/2019] [Indexed: 02/06/2023]
Abstract
The aged brain has biochemical and morphological alterations in the dendrites of the pyramidal neurons of the limbic system, which consequently trigger motor and cognitive deficits. Bexarotene 4-[1-(3,5,5,8,8-pentamethyl-6,7-dihydronaphthalen-2-yl)ethenyl]benzoic acid is a selective agonist of X-retinoid receptors which acts by binding to the intracellular retinoic acid receptors (RAR). It decreases oxidative and inflammatory activity, in addition to the transport of lipids, mechanisms that together could have a neuroprotective effect. Our objective was to evaluate the effect of bexarotene on the motor and cognitive processes, as well as its influence on the dendritic morphology of neurons in the limbic system of elderly mice. Dendritic morphology was evaluated with the Golgi-Cox staining procedure followed by the Sholl analysis. Bexarotene was administered at different doses: 0.0; 0.5; 2.5 and 5.0 mg/kg for 60 days in 18-month-old mice. After the treatment, locomotor activity in a novel environment and spatial memory in the water labyrinth were evaluated. Mice treated with bexarotene did not show significant changes in their behavior. Moreover, bexarotene-treated mice only showed a significant increase in the density of the dendritic spines and the dendritic length in the nucleus accumbens (NAcc) neurons. In conclusion, the administration of bexarotene improves the plasticity of the NAcc of aged mice, and therefore could be a pharmacological alternative to prevent or delay neuroplasticity disruptions in brain aging.
Collapse
Affiliation(s)
- Elibeth Monroy
- Laboratorio de Neuropsiquiatría, Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla. Puebla, Mexico; Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (IPN). CDMX, Mexico
| | - Alfonso Diaz
- Departamento de Farmacia, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla. Puebla, Mexico
| | - Hiram Tendilla-Beltrán
- Laboratorio de Neuropsiquiatría, Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla. Puebla, Mexico; Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (IPN). CDMX, Mexico
| | - Fidel de la Cruz
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (IPN). CDMX, Mexico
| | - Gonzalo Flores
- Laboratorio de Neuropsiquiatría, Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla. Puebla, Mexico.
| |
Collapse
|
23
|
Apolipoprotein E Deficiency Aggravates Neuronal Injury by Enhancing Neuroinflammation via the JNK/c-Jun Pathway in the Early Phase of Experimental Subarachnoid Hemorrhage in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3832648. [PMID: 31949876 PMCID: PMC6944964 DOI: 10.1155/2019/3832648] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 07/06/2019] [Accepted: 09/24/2019] [Indexed: 12/20/2022]
Abstract
Neuronal injury is the primary cause of poor outcome after subarachnoid hemorrhage (SAH). The apolipoprotein E (APOE) gene has been suggested to be involved in the prognosis of SAH patients. However, the role of APOE in neuronal injury after SAH has not been well studied. In this study, SAH was induced in APOE-knockout (APOE−/−) and wild-type (WT) mice to investigate the impact of APOE deficiency on neuronal injury in the early phase of SAH. The experiments of this study were performed in murine SAH models in vivo and primary cultured microglia and neurons in vitro. The SAH model was induced by endovascular perforation in APOE−/− and APOE WT mice. The mortality rate, weight loss, and neurological deficits were recorded within 72 h after SAH. The neuronal injury was assessed by detecting the neuronal apoptosis and axonal injury. The activation of microglia was assessed by immunofluorescent staining of Iba-1, and clodronate liposomes were used for inhibiting microglial activation. The expression of JNK/c-Jun was evaluated by immunofluorescent staining or western blotting. The expression of TNF-α, IL-1β, and IL-6 was evaluated by ELISA. Primary cultured microglia were treated with hemoglobin (Hb) in vitro for simulating the pathological process of SAH. SP600125, a JNK inhibitor, was used for evaluating the role of JNK in neuroinflammation. Nitrite production was detected for microglial activation, and flow cytometry was performed to detect apoptosis in vitro. The results suggested that SAH induced early neuronal injury and neurological deficits in mice. APOE deficiency resulted in more severe neurological deficits after SAH in mice. The neurological deficits were associated with exacerbation of neuronal injury, including neuronal apoptosis and axonal injury. Moreover, APOE deficiency enhanced microglial activation and related inflammatory injury on neurons. Inhibition of microglia attenuated neuronal injury in mice, whereas inhibition of JNK inhibited microglia-mediated inflammatory response in vitro. Taken together, JNK/c-Jun was involved in the enhancement of microglia-mediated inflammatory injury in APOE−/− mice. APOE deficiency aggravates neuronal injury which may account for the poor neurological outcomes of APOE−/− mice. The possible protective role of APOE against EBI via the modulation of inflammatory response indicates its potential treatment for SAH.
Collapse
|
24
|
Bexarotene Attenuates Focal Cerebral Ischemia–Reperfusion Injury via the Suppression of JNK/Caspase-3 Signaling Pathway. Neurochem Res 2019; 44:2809-2820. [DOI: 10.1007/s11064-019-02902-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 09/12/2019] [Accepted: 10/25/2019] [Indexed: 12/11/2022]
|
25
|
Ben-Moshe H, Luz I, Liraz O, Boehm-Cagan A, Salomon-Zimri S, Michaelson D. ApoE4 Exacerbates Hippocampal Pathology Following Acute Brain Penetration Injury in Female Mice. J Mol Neurosci 2019; 70:32-44. [DOI: 10.1007/s12031-019-01397-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 08/14/2019] [Indexed: 10/26/2022]
|
26
|
Hemonnot AL, Hua J, Ulmann L, Hirbec H. Microglia in Alzheimer Disease: Well-Known Targets and New Opportunities. Front Aging Neurosci 2019; 11:233. [PMID: 31543810 PMCID: PMC6730262 DOI: 10.3389/fnagi.2019.00233] [Citation(s) in RCA: 210] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 08/14/2019] [Indexed: 12/21/2022] Open
Abstract
Microglia are the resident macrophages of the central nervous system. They play key roles in brain development, and physiology during life and aging. Equipped with a variety of molecular sensors and through the various functions they can fulfill, they are critically involved in maintaining the brain’s homeostasis. In Alzheimer disease (AD), microglia reaction was initially thought to be incidental and triggered by amyloid deposits and dystrophic neurites. However, recent genome-wide association studies have established that the majority of AD risk loci are found in or near genes that are highly and sometimes uniquely expressed in microglia. This leads to the concept of microglia being critically involved in the early steps of the disease and identified them as important potential therapeutic targets. Whether microglia reaction is beneficial, detrimental or both to AD progression is still unclear and the subject of intense debate. In this review, we are presenting a state-of-knowledge report intended to highlight the variety of microglial functions and pathways shown to be critically involved in AD progression. We first address both the acquisition of new functions and the alteration of their homeostatic roles by reactive microglia. Second, we propose a summary of new important parameters currently emerging in the field that need to be considered to identify relevant microglial targets. Finally, we discuss the many obstacles in designing efficient therapeutic strategies for AD and present innovative technologies that may foster our understanding of microglia roles in the pathology. Ultimately, this work aims to fly over various microglial functions to make a general and reliable report of the current knowledge regarding microglia’s involvement in AD and of the new research opportunities in the field.
Collapse
Affiliation(s)
- Anne-Laure Hemonnot
- Institute for Functional Genomics (IGF), University of Montpellier, Centre National de la Recherche Scientififique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
| | - Jennifer Hua
- Institute for Functional Genomics (IGF), University of Montpellier, Centre National de la Recherche Scientififique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
| | - Lauriane Ulmann
- Institute for Functional Genomics (IGF), University of Montpellier, Centre National de la Recherche Scientififique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
| | - Hélène Hirbec
- Institute for Functional Genomics (IGF), University of Montpellier, Centre National de la Recherche Scientififique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
| |
Collapse
|
27
|
Chernick D, Ortiz-Valle S, Jeong A, Qu W, Li L. Peripheral versus central nervous system APOE in Alzheimer's disease: Interplay across the blood-brain barrier. Neurosci Lett 2019; 708:134306. [PMID: 31181302 DOI: 10.1016/j.neulet.2019.134306] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 05/28/2019] [Accepted: 05/29/2019] [Indexed: 12/20/2022]
Abstract
The apolipoprotein E (APOE) ε4 allele has been demonstrated as the preeminent genetic risk factor for late onset Alzheimer's disease (AD), which comprises greater than 90% of all AD cases. The discovery of the connection between different APOE genotypes and AD risk in the early 1990s spurred three decades of intense and comprehensive research into the function of APOE in the normal and diseased brain. The importance of APOE in the periphery has been well established, due to its pivotal role in maintaining cholesterol homeostasis and cardiovascular health. The influence of vascular factors on brain function and AD risk has been extensively studied in recent years. As a major apolipoprotein regulating multiple molecular pathways beyond its canonical lipid-related functions in the periphery and the central nervous system, APOE represents a critical link between the two compartments, and may influence AD risk from both sides of the blood-brain barrier. This review discusses recent advances in understanding the different functions of APOE in the periphery and in the brain, and highlights several promising APOE-targeted therapeutic strategies for AD.
Collapse
Affiliation(s)
| | | | - Angela Jeong
- Department of Experimental and Clinical Pharmacology, Minneapolis, MN, United States
| | - Wenhui Qu
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Ling Li
- Departments of Pharmacology, Minneapolis, MN, United States; Department of Experimental and Clinical Pharmacology, Minneapolis, MN, United States; Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, United States.
| |
Collapse
|
28
|
Tzioras M, Davies C, Newman A, Jackson R, Spires‐Jones T. Invited Review: APOE at the interface of inflammation, neurodegeneration and pathological protein spread in Alzheimer's disease. Neuropathol Appl Neurobiol 2019; 45:327-346. [PMID: 30394574 PMCID: PMC6563457 DOI: 10.1111/nan.12529] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 10/27/2018] [Indexed: 12/13/2022]
Abstract
Despite more than a century of research, the aetiology of sporadic Alzheimer's disease (AD) remains unclear and finding disease modifying treatments for AD presents one of the biggest medical challenges of our time. AD pathology is characterized by deposits of aggregated amyloid beta (Aβ) in amyloid plaques and aggregated tau in neurofibrillary tangles. These aggregates begin in distinct brain regions and spread throughout the brain in stereotypical patterns. Neurodegeneration, comprising loss of synapses and neurons, occurs in brain regions with high tangle pathology, and an inflammatory response of glial cells appears in brain regions with pathological aggregates. Inheriting an apolipoprotein E ε4 (APOE4) allele strongly increases the risk of developing AD for reasons that are not yet entirely clear. Substantial amounts of evidence support a role for APOE in modulating the aggregation and clearance of Aβ, and data have been accumulating recently implicating APOE4 in exacerbating neurodegeneration, tau pathology and inflammation. We hypothesize that APOE4 influences all the pathological hallmarks of AD and may sit at the interface between neurodegeneration, inflammation and the spread of pathologies through the brain. Here, we conducted a systematic search of the literature and review evidence supporting a role for APOE4 in neurodegeneration and inflammation. While there is no direct evidence yet for APOE4 influencing the spread of pathology, we postulate that this may be found in future based on the literature reviewed here. In conclusion, this review highlights the importance of understanding the role of APOE in multiple important pathological mechanisms in AD.
Collapse
Affiliation(s)
- M. Tzioras
- UK Dementia Research Institute and Centre for Discovery Brain SciencesThe University of EdinburghEdinburghUK
| | - C. Davies
- UK Dementia Research Institute and Centre for Discovery Brain SciencesThe University of EdinburghEdinburghUK
| | - A. Newman
- UK Dementia Research Institute and Centre for Discovery Brain SciencesThe University of EdinburghEdinburghUK
| | - R. Jackson
- UK Dementia Research Institute and Centre for Discovery Brain SciencesThe University of EdinburghEdinburghUK
- Massachusetts General Hospital and Harvard Medical SchoolCharlestownMAUSA
| | - T. Spires‐Jones
- UK Dementia Research Institute and Centre for Discovery Brain SciencesThe University of EdinburghEdinburghUK
| |
Collapse
|
29
|
Newcombe EA, Camats-Perna J, Silva ML, Valmas N, Huat TJ, Medeiros R. Inflammation: the link between comorbidities, genetics, and Alzheimer's disease. J Neuroinflammation 2018; 15:276. [PMID: 30249283 PMCID: PMC6154824 DOI: 10.1186/s12974-018-1313-3] [Citation(s) in RCA: 373] [Impact Index Per Article: 53.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 09/11/2018] [Indexed: 12/21/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder, most cases of which lack a clear causative event. This has made the disease difficult to characterize and, thus, diagnose. Although some cases are genetically linked, there are many diseases and lifestyle factors that can lead to an increased risk of developing AD, including traumatic brain injury, diabetes, hypertension, obesity, and other metabolic syndromes, in addition to aging. Identifying common factors and trends between these conditions could enhance our understanding of AD and lead to the development of more effective treatments. Although the immune system is one of the body’s key defense mechanisms, chronic inflammation has been increasingly linked with several age-related diseases. Moreover, it is now well accepted that chronic inflammation has an important role in the onset and progression of AD. In this review, the different inflammatory signals associated with AD and its risk factors will be outlined to demonstrate how chronic inflammation may be influencing individual susceptibility to AD. Our goal is to bring attention to potential shared signals presented by the immune system during different conditions that could lead to the development of successful treatments.
Collapse
Affiliation(s)
- Estella A Newcombe
- Neurula Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Building 79, Brisbane, 4072, QLD, Australia.
| | - Judith Camats-Perna
- Neurula Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Building 79, Brisbane, 4072, QLD, Australia
| | - Mallone L Silva
- Neurula Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Building 79, Brisbane, 4072, QLD, Australia
| | - Nicholas Valmas
- Queensland Brain Institute, The University of Queensland, Brisbane, 4072, QLD, Australia
| | - Tee Jong Huat
- Neurula Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Building 79, Brisbane, 4072, QLD, Australia.,Centre for Stem Cell Ageing and Regenerative Engineering, The University of Queensland, Brisbane, 4072, QLD, Australia
| | - Rodrigo Medeiros
- Neurula Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Building 79, Brisbane, 4072, QLD, Australia.
| |
Collapse
|
30
|
Clark IA, Vissel B. Therapeutic implications of how TNF links apolipoprotein E, phosphorylated tau, α-synuclein, amyloid-β and insulin resistance in neurodegenerative diseases. Br J Pharmacol 2018; 175:3859-3875. [PMID: 30097997 PMCID: PMC6151331 DOI: 10.1111/bph.14471] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 06/26/2018] [Accepted: 07/23/2018] [Indexed: 12/24/2022] Open
Abstract
While cytokines such as TNF have long been recognized as essential to normal cerebral physiology, the implications of their chronic excessive production within the brain are now also increasingly appreciated. Syndromes as diverse as malaria and lead poisoning, as well as non‐infectious neurodegenerative diseases, illustrate this. These cytokines also orchestrate changes in tau, α‐synuclein, amyloid‐β levels and degree of insulin resistance in most neurodegenerative states. New data on the effects of salbutamol, an indirect anti‐TNF agent, on α‐synuclein and Parkinson's disease, APOE4 and tau add considerably to the rationale of the anti‐TNF approach to understanding, and treating, these diseases. Therapeutic advances being tested, and arguably useful for a number of the neurodegenerative diseases, include a reduction of excess cerebral TNF, whether directly, with a specific anti‐TNF biological agent such as etanercept via Batson's plexus, or indirectly via surgically implanting stem cells. Inhaled salbutamol also warrants investigating further across the neurodegenerative disease spectrum. It is now timely to integrate this range of new information across the neurodegenerative disease spectrum, rather than keep seeing it through the lens of individual disease states.
Collapse
Affiliation(s)
- I A Clark
- Research School of Biology, Australian National University, Canberra, Australia
| | - B Vissel
- Centre for Neuroscience and Regenerative Medicine, Faculty of Science, University of Technology, Sydney, NSW, Australia.,St. Vincent's Centre for Applied Medical Research, Sydney, NSW, Australia
| |
Collapse
|
31
|
Castranio EL, Wolfe CM, Nam KN, Letronne F, Fitz NF, Lefterov I, Koldamova R. ABCA1 haplodeficiency affects the brain transcriptome following traumatic brain injury in mice expressing human APOE isoforms. Acta Neuropathol Commun 2018; 6:69. [PMID: 30049279 PMCID: PMC6062955 DOI: 10.1186/s40478-018-0569-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 07/12/2018] [Indexed: 02/07/2023] Open
Abstract
Expression of human Apolipoprotein E (APOE) modulates the inflammatory response in an isoform specific manner, with APOE4 isoform eliciting a stronger pro-inflammatory response, suggesting a possible mechanism for worse outcome following traumatic brain injury (TBI). APOE lipidation and stability is modulated by ATP-binding cassette transporter A1 (ABCA1), a transmembrane protein that transports lipids and cholesterol onto APOE. We examined the impact of Abca1 deficiency and APOE isoform expression on the response to TBI using 3-months-old, human APOE3+/+ (E3/Abca1+/+) and APOE4+/+ (E4/Abca1+/+) targeted replacement mice, and APOE3+/+ and APOE4+/+ mice with only one functional copy of the Abca1 gene (E3/Abca1+/-; E4/Abca1+/-). TBI-treated mice received a craniotomy followed by a controlled cortical impact (CCI) brain injury in the left hemisphere; sham-treated mice received the same surgical procedure without the impact. We performed RNA-seq using samples from cortices and hippocampi followed by genome-wide differential gene expression analysis. We found that TBI significantly impacted unique transcripts within each group, however, the proportion of unique transcripts was highest in E4/Abca1+/- mice. Additionally, we found that Abca1 haplodeficiency increased the expression of microglia sensome genes among only APOE4 injured mice, a response not seen in injured APOE3 mice, nor in either group of sham-treated mice. To identify gene networks, or modules, correlated to TBI, APOE isoform and Abca1 haplodeficiency, we used weighted gene co-expression network analysis (WGCNA). The module that positively correlated to TBI groups was associated with immune response and featured hub genes that were microglia-specific, including Trem2, Tyrobp, Cd68 and Hexb. The modules positively correlated with APOE4 isoform and negatively to Abca1 haplodeficient mice represented "protein translation" and "oxidation-reduction process", respectively. Our results reveal E4/Abca1+/- TBI mice have a distinct response to injury, and unique gene networks are associated with APOE isoform, Abca1 insufficiency and injury.
Collapse
Affiliation(s)
- Emilie L Castranio
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Cody M Wolfe
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Kyong Nyon Nam
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Florent Letronne
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Nicholas F Fitz
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Iliya Lefterov
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
| | - Radosveta Koldamova
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
32
|
APOE and Alzheimer's Disease: Evidence Mounts that Targeting APOE4 may Combat Alzheimer's Pathogenesis. Mol Neurobiol 2018; 56:2450-2465. [PMID: 30032423 DOI: 10.1007/s12035-018-1237-z] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 07/11/2018] [Indexed: 10/28/2022]
Abstract
Alzheimer's disease (AD) is an immutable neurodegenerative disease featured by the two hallmark brain pathologies that are the extracellular amyloid ß (Aß) and intraneuronal tau protein. People carrying the APOE4 allele are at high risk of AD concerning the ones carrying the ε3 allele, while the ε2 allele abates risk. ApoE isoforms exert a central role in controlling the transport of brain lipid, neuronal signaling, mitochondrial function, glucose metabolism, and neuroinflammation. Regardless of widespread indispensable studies, the appropriate function of APOE in AD etiology stays ambiguous. Existing proof recommends that the disparate outcomes of ApoE isoforms on Aβ accretion and clearance have a distinct function in AD pathogenesis. ApoE-lipoproteins combine diverse cell-surface receptors to transport lipids and moreover to lipophilic Aβ peptide, that is believed to begin deadly events that generate neurodegeneration in the AD. ApoE has great influence in tau pathogenesis, tau-mediated neurodegeneration, and neuroinflammation, as well as α-synucleinopathy, lipid metabolism, and synaptic plasticity despite the presence of Aβ pathology. ApoE4 shows the deleterious effect for AD while the lack of ApoE4 is defensive. Therapeutic strategies primarily depend on APOE suggest to lessen the noxious effects of ApoE4 and reestablish the protective aptitudes of ApoE. This appraisal represents the critical interactions of APOE and AD pathology, existing facts on ApoE levels in the central nervous system (CNS), and the credible active stratagems for AD therapy by aiming ApoE. This review also highlighted utmost ApoE targeting therapeutic tactics that are crucial for controlling Alzheimer's pathogenesis.
Collapse
|
33
|
Montoliu-Gaya L, Mulder SD, Herrebout MA, Baayen JC, Villegas S, Veerhuis R. Aβ-oligomer uptake and the resulting inflammatory response in adult human astrocytes are precluded by an anti-Aβ single chain variable fragment in combination with an apoE mimetic peptide. Mol Cell Neurosci 2018; 89:49-59. [DOI: 10.1016/j.mcn.2018.03.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 03/30/2018] [Accepted: 03/31/2018] [Indexed: 10/17/2022] Open
|
34
|
Yang X, Chen S, Shao Z, Li Y, Wu H, Li X, Mao L, Zhou Z, Bai L, Mei X, Liu C. Apolipoprotein E Deficiency Exacerbates Spinal Cord Injury in Mice: Inflammatory Response and Oxidative Stress Mediated by NF-κB Signaling Pathway. Front Cell Neurosci 2018; 12:142. [PMID: 29875635 PMCID: PMC5974465 DOI: 10.3389/fncel.2018.00142] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 05/09/2018] [Indexed: 12/30/2022] Open
Abstract
Spinal cord injury (SCI) is a severe neurological trauma that involves complex pathological processes. Inflammatory response and oxidative stress are prevalent during the second injury and can influence the functional recovery of SCI. Specially, Apolipoprotein E (APOE) induces neuronal repair and nerve regeneration, and the deficiency of Apoe impairs spinal cord-blood-barrier and reduces functional recovery after SCI. However, the mechanism by which Apoe mediates signaling pathways of inflammatory response and oxidative stress in SCI remains largely elusive. This study was designed to investigate the signaling pathways that regulate Apoe deficiency-dependent inflammatory response and oxidative stress in the acute stage of SCI. In the present study, Apoe-/- mice retarded functional recovery and had a larger lesion size when compared to wild-type mice after SCI. Moreover, deficiency of Apoe induced an exaggerated inflammatory response by increasing expression of interleukin-6 (IL-6) and interleukin-1β (IL-1β), and increased oxidative stress by reducing expression of Nrf2 and HO-1. Furthermore, lack of Apoe promoted neuronal apoptosis and decreased neuronal numbers in the anterior horn of the spinal cord after SCI. Mechanistically, we found that the absence of Apoe increased inflammation and oxidative stress through activation of NF-κB after SCI. In contrast, an inhibitor of nuclear factor-κB (NF-κB; Pyrrolidine dithiocarbamate) alleviates these changes. Collectively, these results indicate that a critical role for activation of NF-κB in regulating Apoe-deficiency dependent inflammation and oxidative stress is detrimental to recovery after SCI.
Collapse
Affiliation(s)
- Xuan Yang
- School of Nursing, Jinzhou Medical University, Jinzhou, China
| | - Shurui Chen
- Department of Orthopedics, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Zhenya Shao
- Department of Orthopedics, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Yuanlong Li
- Department of Orthopedics, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - He Wu
- Department of Endocrinology, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Xian Li
- Department of Orthopedics, Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Liang Mao
- Department of Oncology, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Zipeng Zhou
- Department of Orthopedics, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Liangjie Bai
- Department of Orthopedics, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xifan Mei
- Department of Orthopedics, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Chang Liu
- Department of Endocrinology, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
35
|
Pang J, Wu Y, Peng J, Yang P, Kuai L, Qin X, Cao F, Sun X, Chen L, Vitek MP, Jiang Y. Potential implications of Apolipoprotein E in early brain injury after experimental subarachnoid hemorrhage: Involvement in the modulation of blood-brain barrier integrity. Oncotarget 2018; 7:56030-56044. [PMID: 27463015 PMCID: PMC5302894 DOI: 10.18632/oncotarget.10821] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 07/10/2016] [Indexed: 01/02/2023] Open
Abstract
Apolipoprotein E (Apoe) genetic polymorphisms have been implicated in the long term outcome of subarachnoid haemorrhage (SAH), but little is known about the effect of Apoe on the early brain injury (EBI) after SAH. This study investigated the potential role of APOE in EBI post-SAH. Multiple techniques were used to determine the early BBB disruption in EBI post-SAH in a murine model using wild-type (WT) and Apoe−/− (KO) mice. Progressive BBB disruption (Evans blue extravasation and T2 hyperintensity in magnetic resonance imaging) was observed before the peak of endogenous APOE expression elevation at 48h after SAH. Moreover, Apoe−/− mice exhibited more severe BBB disruption charcteristics after SAH than WT mice, including higher levels of Evans blue and IgG extravasation, T2 hyperintensity in magnetic resonance imaging, tight junction proteins degradation and endothelial cells death. Mechanistically, we found that APOE restores the BBB integrity in the acute stage after SAH via the cyclophilin A (CypA)-NF-κB-proinflammatory cytokines-MMP-9 signalling pathway. Consequently, although early BBB disruption causes neurological dysfunctions after SAH, we capture a different aspect of the effects of APOE on EBI after SAH that previous studies had overlooked and open up the idea of BBB disruption as a target of APOE-based therapy for EBI amelioration research in the future.
Collapse
Affiliation(s)
- Jinwei Pang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yue Wu
- Departement of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jianhua Peng
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Ping Yang
- Department of Vasculocardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Li Kuai
- Department of Ophthalmology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xinghu Qin
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Fang Cao
- Department of Neurovascular Disease, The Affiliated Hospital of Zunyi Medical College, Zunyi, China
| | - Xiaochuan Sun
- Departement of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ligang Chen
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Michael P Vitek
- Department of Medicine (Neurology), Duke University Medical Center, Durham, North Carolina, United States
| | - Yong Jiang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
36
|
Jiang Y, He T, Deng W, Sun P. Association between apolipoprotein E gene polymorphism and mild cognitive impairment: a meta-analysis. Clin Interv Aging 2017; 12:1941-1949. [PMID: 29180857 PMCID: PMC5691922 DOI: 10.2147/cia.s143632] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
A number of published case–control studies reported that the apolipoprotein E (ApoE) gene polymorphism was associated with the mild cognitive impairment (MCI). However, previous reports still remain conflicting. To estimate the association between ApoE polymorphism and MCI susceptibility, we searched the electronic databases including PubMed, Wanfang, CNKI (China National Knowledge Infrastructure), VIP, and EMBASE to retrieve all available studies. A total of 18 studies with 2,004 cases and 3,705 controls were included in this meta-analysis. The pooled analysis based on selected studies showed that statistically significant risk association was found between ApoE gene polymorphism and MCI in overall population (ε4 vs ε3: odds ratio [OR] =2.38, 95% confidence interval [CI]: 2.11–2.68; ε4/ε4 vs ε3/ε3: OR =4.45, 95% CI: 3.06–6.48; ε2/ε4 vs ε3/ε3: OR =2.57, 95% CI: 1.77–3.73; ε3/ε4 vs ε3/ε3: OR =2.31, 95% CI: 1.99–2.69). However, no significant association was detected in two genetic models: ε2 versus ε3 (OR =0.90, 95% CI: 0.77–1.05) and ε2/ε2 versus ε3/ε3 (OR =0.91, 95% CI: 0.50–1.65). Furthermore, ApoE ε2/ε3 genotype provided a slight protection for MCI in overall population (ε2/ε3 vs ε3/ε3: OR =0.80, 95% CI: 0.66–0.97). In the stratified analysis based on ethnicity, similar results were also observed in Chinese population (significant risk: ε4 vs ε3: OR =2.52, 95% CI: 2.19–2.90; ε4/ε4 vs ε3/ε3: OR =5.45, 95% CI: 3.41–8.70; ε2/ε4 vs ε3/ε3: OR =2.59, 95% CI: 1.74–3.86; ε3/ε4 vs ε3/ε3: OR =2.34, 95% CI: 1.97–2.79; slight protection: ε2/ε3 vs ε3/ε3: OR =0.79, 95% CI: 0.64–0.98; no association: ε2 vs ε3: OR =0.92, 95% CI: 0.78–1.09; and ε2/ε2 vs ε3/ε3: OR =1.04, 95% CI: 0.55–1.99). In summary, this meta-analysis of 5,709 subjects suggested that ApoE ε4 allele was associated with an increased risk of MCI. In addition, ApoE ε2/ε3 genotype provided a slight protection for MCI.
Collapse
Affiliation(s)
- Yunxia Jiang
- Nursing College of Qingdao University, Qingdao University
| | - Tao He
- Department of Neurosurgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wenshuai Deng
- Department of Neurosurgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Peng Sun
- Department of Neurosurgery, Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
37
|
Tu TM, Kolls BJ, Soderblom EJ, Cantillana V, Ferrell PD, Moseley MA, Wang H, Dawson HN, Laskowitz DT. Apolipoprotein E mimetic peptide, CN-105, improves outcomes in ischemic stroke. Ann Clin Transl Neurol 2017; 4:246-265. [PMID: 28382306 PMCID: PMC5376751 DOI: 10.1002/acn3.399] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 01/09/2017] [Accepted: 02/06/2017] [Indexed: 01/05/2023] Open
Abstract
Objective At present, the absence of a pharmacological neuroprotectant represents an important unmet clinical need in the treatment of ischemic and traumatic brain injury. Recent evidence suggests that administration of apolipoprotein E mimetic therapies represent a viable therapeutic strategy in this setting. We investigate the neuroprotective and anti‐inflammatory properties of the apolipoprotein E mimetic pentapeptide, CN‐105, in a microglial cell line and murine model of ischemic stroke. Methods Ten to 13‐week‐old male C57/BL6 mice underwent transient middle cerebral artery occlusion and were randomly selected to receive CN‐105 (0.1 mg/kg) in 100 μL volume or vehicle via tail vein injection at various time points. Survival, motor‐sensory functional outcomes using rotarod test and 4‐limb wire hanging test, infarct volume assessment using 2,3,5‐Triphenyltetrazolium chloride staining method, and microglial activation in the contralateral hippocampus using F4/80 immunostaining were assessed at various time points. In vitro assessment of tumor necrosis factor‐alpha secretion in a microglial cell line was performed, and phosphoproteomic analysis conducted to explore early mechanistic pathways of CN‐105 in ischemic stroke. Results Mice receiving CN‐105 demonstrated improved survival, improved functional outcomes, reduced infarct volume, and reduced microglial activation. CN‐105 also suppressed inflammatory cytokines secretion in microglial cells in vitro. Phosphoproteomic signals suggest that CN‐105 reduces proinflammatory pathways and lower oxidative stress. Interpretation CN‐105 improves functional and histological outcomes in a murine model of ischemic stroke via modulation of neuroinflammatory pathways. Recent clinical trial of this compound has demonstrated favorable pharmacokinetic and safety profile, suggesting that CN‐105 represents an attractive candidate for clinical translation.
Collapse
Affiliation(s)
- Tian Ming Tu
- Department of Neurology Duke University School of Medicine Durham North Carolina; Department of Neurology National Neuroscience Institute Tan Tock Seng Campus Singapore
| | - Brad J Kolls
- Department of Neurology Duke University School of Medicine Durham North Carolina
| | - Erik J Soderblom
- Duke Proteomics Core Facility Center for Genomic and Computational Biology Duke University Durham North Carolina
| | - Viviana Cantillana
- Department of Neurology Duke University School of Medicine Durham North Carolina
| | - Paul Durham Ferrell
- Department of Pathology Duke University School of Medicine Durham North Carolina
| | - M Arthur Moseley
- Duke Proteomics Core Facility Center for Genomic and Computational Biology Duke University Durham North Carolina
| | - Haichen Wang
- Department of Neurology Duke University School of Medicine Durham North Carolina
| | - Hana N Dawson
- Department of Neurology Duke University School of Medicine Durham North Carolina
| | - Daniel T Laskowitz
- Department of Neurology Duke University School of Medicine Durham North Carolina
| |
Collapse
|
38
|
Feng CZ, Yin JB, Yang JJ, Cao L. Regulatory factor X1 depresses ApoE-dependent Aβ uptake by miRNA-124 in microglial response to oxidative stress. Neuroscience 2017; 344:217-228. [DOI: 10.1016/j.neuroscience.2016.12.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 12/04/2016] [Accepted: 12/09/2016] [Indexed: 01/12/2023]
|
39
|
Finan GM, Realubit R, Chung S, Lütjohann D, Wang N, Cirrito JR, Karan C, Kim TW. Bioactive Compound Screen for Pharmacological Enhancers of Apolipoprotein E in Primary Human Astrocytes. Cell Chem Biol 2016; 23:1526-1538. [DOI: 10.1016/j.chembiol.2016.10.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 09/13/2016] [Accepted: 10/25/2016] [Indexed: 02/07/2023]
|
40
|
Zhang X, Hu J, Zhong L, Wang N, Yang L, Liu CC, Li H, Wang X, Zhou Y, Zhang Y, Xu H, Bu G, Zhuang J. Quercetin stabilizes apolipoprotein E and reduces brain Aβ levels in amyloid model mice. Neuropharmacology 2016; 108:179-92. [DOI: 10.1016/j.neuropharm.2016.04.032] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 04/18/2016] [Accepted: 04/21/2016] [Indexed: 10/21/2022]
|
41
|
Yates RB, Stafford-Smith M. The Genetic Determinants of Renal Impairment Following Cardiac Surgery. Semin Cardiothorac Vasc Anesth 2016; 10:314-26. [PMID: 17200089 DOI: 10.1177/1089253206294350] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Cardiac surgery is frequently performed, and acute renal dysfunction is a common adverse event following this procedure. Cardiac surgery-related renal injury independently predicts longer hospital stays and greater rates of morbidity and mortality. Although much work has been completed toward better understanding of this phenomenon, the state of knowledge concerning surgery-related renal injury remains limited. Currently, there is no effective paradigm to identify patients who are at risk for this condition; the specific mechanisms of renal injury during surgery are incompletely understood; and few therapies exist to prevent or treat this phenomenon. To better understand this common clinical problem, recent research has focused on the importance of genetic variability within the physiological and patho-physiological systems that underlie renal dysfunction following cardiac surgery. Emphasizing the importance of using genetics to elucidate molecular mechanisms of this disease, this article reviews the current literature on genetic polymorphisms and post cardiac surgery-related renal dysfunction.
Collapse
Affiliation(s)
- Robert B Yates
- Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | | |
Collapse
|
42
|
Dose J, Huebbe P, Nebel A, Rimbach G. APOE genotype and stress response - a mini review. Lipids Health Dis 2016; 15:121. [PMID: 27457486 PMCID: PMC4960866 DOI: 10.1186/s12944-016-0288-2] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 07/12/2016] [Indexed: 12/31/2022] Open
Abstract
The APOE gene is one of currently only two genes that have consistently been associated with longevity. Apolipoprotein E (APOE) is a plasma protein which plays an important role in lipid and lipoprotein metabolism. In humans, there are three major APOE isoforms, designated APOE2, APOE3, and APOE4. Of these three isoforms, APOE3 is most common while APOE4 was shown to be associated with age-related diseases, including cardiovascular and Alzheimer’s disease, and therefore an increased mortality risk with advanced age. Evidence accumulates, showing that oxidative stress and, correspondingly, mitochondrial function is affected in an APOE isoform-dependent manner. Accordingly, several stress response pathways implicated in the aging process, including the endoplasmic reticulum stress response and immune function, appear to be influenced by the APOE genotype. The investigation and development of treatment strategies targeting APOE4 have not resolved any therapeutic yet that could be entirely recommended. This mini-review provides an overview on the state of research concerning the impact of the APOE genotype on stress response-related processes, emphasizing the strong interconnection between mitochondrial function, endoplasmic reticulum stress and the immune response. Furthermore, this review addresses potential treatment strategies and associated pitfalls as well as lifestyle interventions that could benefit people with an at risk APOE4 genotype.
Collapse
Affiliation(s)
- Janina Dose
- Institute of Human Nutrition and Food Science, Kiel University, Hermann-Rodewald-Str. 6, D-24118, Kiel, Germany. .,Institute of Clinical Molecular Biology, Kiel University, Schittenhelmstr. 12, D-24105, Kiel, Germany.
| | - Patricia Huebbe
- Institute of Human Nutrition and Food Science, Kiel University, Hermann-Rodewald-Str. 6, D-24118, Kiel, Germany
| | - Almut Nebel
- Institute of Clinical Molecular Biology, Kiel University, Schittenhelmstr. 12, D-24105, Kiel, Germany
| | - Gerald Rimbach
- Institute of Human Nutrition and Food Science, Kiel University, Hermann-Rodewald-Str. 6, D-24118, Kiel, Germany
| |
Collapse
|
43
|
Zhong J, Cheng C, Liu H, Huang Z, Wu Y, Teng Z, He J, Zhang H, Wu J, Cao F, Jiang L, Sun X. Bexarotene protects against traumatic brain injury in mice partially through apolipoprotein E. Neuroscience 2016; 343:434-448. [PMID: 27235741 DOI: 10.1016/j.neuroscience.2016.05.033] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 05/14/2016] [Accepted: 05/17/2016] [Indexed: 01/29/2023]
Abstract
Bexarotene has been proved to have neuroprotective effects in many animal models of neurological diseases. However, its neuroprotection in traumatic brain injury (TBI) is still unknown. This study aims to explore the neuroprotective effects of bexarotene on TBI and its possible mechanism. Controlled cortical impact (CCI) model was used to simulate TBI in C57BL/6 mice as well as APOE gene knockout (APOE-KO) mice. After CCI, mice were daily dosed with bexarotene or vehicle solution intraperitoneally. The motor function, learning and memory, inflammatory factors, microglia amount, apoptosis condition around injury site and main side-effects were all measured. The results showed that, after CCI, bexarotene treatment markedly improved the motor function and spatial memory in C57BL/6 compare to APOE-KO mice which showed no improvement. The inflammatory cytokines, microglia amount, cell apoptosis rate, and protein of cleaved caspase-3 around the injury site were markedly upregulated after TBI in both C57BL/6 and APOE-KO mice, and all these upregulation were significantly mitigated by bexarotene treatment in C57BL/6 mice, but not in APOE-KO mice. No side-effects were detected after consecutive administration. Taken together, bexarotene inhibits the inflammatory response as well as cell apoptosis and improves the neurological function of mice after TBI partially through apolipoprotein E. This may make it a promising candidate for the therapeutic treatment after TBI.
Collapse
Affiliation(s)
- Jianjun Zhong
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Chongjie Cheng
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Han Liu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Zhijian Huang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yue Wu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Zhipeng Teng
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Junchi He
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Hongrong Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jinchuan Wu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Fang Cao
- Department of Cerebrovascular, The First Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 653000, China
| | - Li Jiang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xiaochuan Sun
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
44
|
Cao F, Jiang Y, Wu Y, Zhong J, Liu J, Qin X, Chen L, Vitek MP, Li F, Xu L, Sun X. Apolipoprotein E-Mimetic COG1410 Reduces Acute Vasogenic Edema following Traumatic Brain Injury. J Neurotrauma 2016; 33:175-82. [PMID: 26192010 PMCID: PMC4722604 DOI: 10.1089/neu.2015.3887] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The degree of post-traumatic brain edema and dysfunction of the blood-brain barrier (BBB) influences the neurofunctional outcome after a traumatic brain injury (TBI). Previous studies have demonstrated that the administration of apolipoprotein E-mimetic peptide COG1410 reduces the brain water content after subarachnoid hemorrhage, intra-cerebral hemorrhage, and focal brain ischemia. However, the effects of COG1410 on vasogenic edema following TBI are not known. The current study evaluated the effects of 1 mg/kg daily COG1410 versus saline administered intravenously after a controlled cortical impact (CCI) injury on BBB dysfunction and vasogenic edema at an acute stage in mice. The results demonstrated that treatment with COG1410 suppressed the activity of matrix metalloproteinase-9, reduced the disruption of the BBB and Evans Blue dye extravasation, reduced the TBI lesion volume and vasogenic edema, and decreased the functional deficits compared with mice treated with vehicle, at an acute stage after CCI. These findings suggest that COG1410 is a promising preclinical therapeutic agent for the treatment of traumatic brain injury.
Collapse
Affiliation(s)
- Fang Cao
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yong Jiang
- Department of Neurosurgery, the Affiliated Hospital of Luzhou Medical College, Luzhou, China
| | - Yue Wu
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jianjun Zhong
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jieshi Liu
- Department of Neurosurgery, the Affiliated Hospital of Luzhou Medical College, Luzhou, China
| | - Xinghu Qin
- Department of Neurosurgery, the Affiliated Hospital of Luzhou Medical College, Luzhou, China
| | - Ligang Chen
- Department of Neurosurgery, the Affiliated Hospital of Luzhou Medical College, Luzhou, China
| | - Michael P. Vitek
- Department of Medicine (Neurology), Duke University Medical Center, Durham, North Carolina
| | - Fengqiao Li
- Cognosci Inc., Research Triangle Park, North Carolina
| | - Lu Xu
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China
| | - Xiaochuan Sun
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
45
|
Chen YL, Wang LM, Chen Y, Gao JY, Marshall C, Cai ZY, Hu G, Xiao M. Changes in astrocyte functional markers and β-amyloid metabolism-related proteins in the early stages of hypercholesterolemia. Neuroscience 2015; 316:178-91. [PMID: 26724580 DOI: 10.1016/j.neuroscience.2015.12.039] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 12/11/2015] [Accepted: 12/19/2015] [Indexed: 12/21/2022]
Abstract
Cholesterol is an essential substance for maintaining normal structure and function of the brain. But unfortunately, a long-term high-cholesterol diet can lead to a variety of pathological changes of the brain such as β-amyloid (Aβ) accumulation, Tau hyperphosphorylation, reactive gliosis, neuroinflammation, neuronal death and synaptic degeneration. These pathological changes have complex internal relations with one other, causing memory impairment and participating in the pathogenesis of Alzheimer's disease (AD). However, early hypercholesterolemia-induced events that lead to brain deterioration are not clear. To address this, 6-month-old female mice were fed a 3% cholesterol diet for 8weeks, followed by behavioral, biochemical and neuropathological analyses. The high-cholesterol-fed mice did not show neuronal and synaptic impairment or cognitive deficits compared with mice given a normal diet, but astrocytes were mildly activated with increased expression of functional markers including apolipoprotein E and aquaporin 4 in the hippocampus. Hippocampal interleukin-1β expression slightly increased, but interleukin-6 (IL-6) and tumor necrosis factor-α did not change significantly compared with those in the control group. Levels of Aβ, and its precursor protein, were unaffected, but levels of presenilin 1 and insulin-degrading enzyme (IDE), that initiate Aβ generation and degradation, respectively, increased in the hippocampus of the model mice. In addition, Tau phosphorylation levels were not different between the control and model groups. These results suggest that changes in astrocyte functional markers and Aβ metabolism proteins, which contribute to maintaining brain cholesterol and Aβ homeostasis, are early events in the process of hypercholesterolemia-related neuropathological changes.
Collapse
Affiliation(s)
- Y L Chen
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 140 Hanzhong Road, Nanjing, Jiangsu 210029, China
| | - L M Wang
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 140 Hanzhong Road, Nanjing, Jiangsu 210029, China
| | - Y Chen
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 140 Hanzhong Road, Nanjing, Jiangsu 210029, China
| | - J Y Gao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 140 Hanzhong Road, Nanjing, Jiangsu 210029, China
| | - C Marshall
- Department of Rehabilitation Sciences, University of Kentucky Center of Excellence in Rural Health, Hazard, KY, USA
| | - Z Y Cai
- Department of Neurology, Shiyan Renmin Hospital, Hubei University of Medicine, No 39 Chaoyang Middle Road, Shiyan, Hubei Province 442000, People's Republic of China
| | - G Hu
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 140 Hanzhong Road, Nanjing, Jiangsu 210029, China
| | - M Xiao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 140 Hanzhong Road, Nanjing, Jiangsu 210029, China.
| |
Collapse
|
46
|
Moser VA, Pike CJ. Obesity and sex interact in the regulation of Alzheimer's disease. Neurosci Biobehav Rev 2015; 67:102-18. [PMID: 26708713 DOI: 10.1016/j.neubiorev.2015.08.021] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 08/01/2015] [Accepted: 08/03/2015] [Indexed: 01/09/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder, for which a number of genetic, environmental, and lifestyle risk factors have been identified. A significant modifiable risk factor is obesity in mid-life. Interestingly, both obesity and AD exhibit sex differences and are regulated by sex steroid hormones. Accumulating evidence suggests interactions between obesity and sex in regulation of AD risk, although the pathways underlying this relationship are unclear. Inflammation and the E4 allele of apolipoprotein E have been identified as independent risk factors for AD and both interact with obesity and sex steroid hormones. We review the individual and cooperative effects of obesity and sex on development of AD and examine the potential contributions of apolipoprotein E, inflammation, and their interactions to this relationship.
Collapse
Affiliation(s)
- V Alexandra Moser
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA 90089, USA.
| | - Christian J Pike
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA 90089, USA; Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
47
|
HWANG HYUNJUNG, JUNG SEUNGHEE, LEE HYUNGCHUL, HAN NAKYUNG, BAE INHWA, LEE MINYOUNG, HAN YOUNGHOON, KANG YOUNGSUN, LEE SUJAE, PARK HEONJOO, KO YOUNGGYU, LEE JAESEON. Identification of novel therapeutic targets in the secretome of ionizing radiation-induced senescent tumor cells. Oncol Rep 2015; 35:841-50. [DOI: 10.3892/or.2015.4473] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 10/26/2015] [Indexed: 11/06/2022] Open
|
48
|
Malik M, Parikh I, Vasquez JB, Smith C, Tai L, Bu G, LaDu MJ, Fardo DW, Rebeck GW, Estus S. Genetics ignite focus on microglial inflammation in Alzheimer's disease. Mol Neurodegener 2015; 10:52. [PMID: 26438529 PMCID: PMC4595327 DOI: 10.1186/s13024-015-0048-1] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 09/23/2015] [Indexed: 12/12/2022] Open
Abstract
In the past five years, a series of large-scale genetic studies have revealed novel risk factors for Alzheimer’s disease (AD). Analyses of these risk factors have focused attention upon the role of immune processes in AD, specifically microglial function. In this review, we discuss interpretation of genetic studies. We then focus upon six genes implicated by AD genetics that impact microglial function: TREM2, CD33, CR1, ABCA7, SHIP1, and APOE. We review the literature regarding the biological functions of these six proteins and their putative role in AD pathogenesis. We then present a model for how these factors may interact to modulate microglial function in AD.
Collapse
Affiliation(s)
- Manasi Malik
- Department of Physiology and Sanders-Brown Center on Aging, University of Kentucky, 800 S. Limestone St, Lexington, KY, 40536, USA.
| | - Ishita Parikh
- Department of Physiology and Sanders-Brown Center on Aging, University of Kentucky, 800 S. Limestone St, Lexington, KY, 40536, USA.
| | - Jared B Vasquez
- Department of Physiology and Sanders-Brown Center on Aging, University of Kentucky, 800 S. Limestone St, Lexington, KY, 40536, USA.
| | - Conor Smith
- Department of Anatomy and Cell Biology, University of Illinois, Chicago, IL, USA.
| | - Leon Tai
- Department of Anatomy and Cell Biology, University of Illinois, Chicago, IL, USA.
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
| | - Mary Jo LaDu
- Department of Anatomy and Cell Biology, University of Illinois, Chicago, IL, USA.
| | - David W Fardo
- Department of Biostatistics and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA.
| | - G William Rebeck
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA.
| | - Steven Estus
- Department of Physiology and Sanders-Brown Center on Aging, University of Kentucky, 800 S. Limestone St, Lexington, KY, 40536, USA.
| |
Collapse
|
49
|
Neuronal Regulation of Neuroprotective Microglial Apolipoprotein E Secretion in Rat In Vitro Models of Brain Pathophysiology. J Neuropathol Exp Neurol 2015; 74:818-34. [PMID: 26185969 DOI: 10.1097/nen.0000000000000222] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Apolipoprotein E (ApoE) is mainly secreted by glial cells and is involved in many brain functions, including neuronal plasticity, β-amyloid clearance, and neuroprotection. Microglia--the main immune cells of the brain--are one source of ApoE, but little is known about the physiologic regulation of microglial ApoE secretion by neurons and whether this release changes under inflammatory or neurodegenerative conditions. Using rat primary neural cell cultures, we show that microglia release ApoE through a Golgi-mediated secretion pathway and that ApoE progressively accumulates in neuroprotective microglia-conditioned medium. This constitutive ApoE release is negatively affected by microglial activation both with lipopolysaccharide and with ATP. Microglial ApoE release is stimulated by neuron-conditioned media and under coculture conditions. Neuron-stimulated microglial ApoE release is mediated by serine and glutamate through N-methyl-D-aspartate receptors and is differently regulated by activation states (i.e. lipopolysaccharide vs ATP) and by 6-hydroxydopamine. Microglial ApoE silencing abrogated protection of cerebellar granule neurons against 6-hydroxydopamine toxicity in cocultures, indicating that microglial ApoE release is neuroprotective. Our findings shed light on the reciprocal cross-talk between neurons and microglia that is crucial for normal brain functions. They also open the way for the identification of possible pharmacologic targets that can modulate neuroprotective microglial ApoE release under pathologic conditions.
Collapse
|
50
|
Miao J, Wang F, Zheng W, Zhuang X. Association of the Apolipoprotein E polymorphism with migraine: a meta-analysis. BMC Neurol 2015; 15:138. [PMID: 26264634 PMCID: PMC4534059 DOI: 10.1186/s12883-015-0385-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 07/17/2015] [Indexed: 11/17/2022] Open
Abstract
Background Apolipoprotein E (ApoE) gene has been reported to be associated with migraine and tension-type headache (TTH), but the results are conflicting. This study aimed to evaluate the association of ApoE with migraine by a meta-analysis. Methods MEDLINE, ISI Web of Knowledge, The Cochrane Central Register of Controlled Trials, and EMBASE databases were searched to identify eligible studies published in English from 2000 to 2014. Data were extracted using standardized forms. The association was assessed by relative risk (RR) with 95 % confidence intervals (CIs) using a fixed or random effects model. Results Four studies, comprising 649 migraineurs, 229 TTH subjects and 975 controls, met all the criteria and were included in the meta-analysis. No significant difference was found comparing genotypic and allelic frequencies in the case of migraineurs versus controls and TTH subjects versus controls. Only when migraineurs and TTH subjects were considered as a whole group, ApoE4 was found to increase the relative risk of headache by 1.48 (95 % CI 1.16, 1.90; P = 0.002), compared to controls. Conclusions ApoE ε4 allele is not associated with migraine susceptibility, but is positively related to headache (including migraine and TTH).
Collapse
Affiliation(s)
- Jiayin Miao
- Department of Neurology, Affiliated Zhongshan Hospital of Xiamen University, 201 Hubinnan Road, Xiamen, 361004, China.
| | - Feng Wang
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen, 361005, China. .,College of Computer Engineering, Jimei University, Xiamen, 361021, China.
| | - Weihong Zheng
- Department of Neurology, Affiliated Zhongshan Hospital of Xiamen University, 201 Hubinnan Road, Xiamen, 361004, China.
| | - Xiaorong Zhuang
- Department of Neurology, Affiliated Zhongshan Hospital of Xiamen University, 201 Hubinnan Road, Xiamen, 361004, China.
| |
Collapse
|