1
|
Chung S, Yi Y, Ullah I, Chung K, Park S, Lim J, Kim C, Pyun SH, Kim M, Kim D, Lee M, Rhim T, Lee SK. Systemic Treatment with Fas-Blocking Peptide Attenuates Apoptosis in Brain Ischemia. Int J Mol Sci 2024; 25:661. [PMID: 38203830 PMCID: PMC10780202 DOI: 10.3390/ijms25010661] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 12/30/2023] [Accepted: 01/02/2024] [Indexed: 01/12/2024] Open
Abstract
Apoptosis plays a crucial role in neuronal injury, with substantial evidence implicating Fas-mediated cell death as a key factor in ischemic strokes. To address this, inhibition of Fas-signaling has emerged as a promising strategy in preventing neuronal cell death and alleviating brain ischemia. However, the challenge of overcoming the blood-brain barrier (BBB) hampers the effective delivery of therapeutic drugs to the central nervous system (CNS). In this study, we employed a 30 amino acid-long leptin peptide to facilitate BBB penetration. By conjugating the leptin peptide with a Fas-blocking peptide (FBP) using polyethylene glycol (PEG), we achieved specific accumulation in the Fas-expressing infarction region of the brain following systemic administration. Notably, administration in leptin receptor-deficient db/db mice demonstrated that leptin facilitated the delivery of FBP peptide. We found that the systemic administration of leptin-PEG-FBP effectively inhibited Fas-mediated apoptosis in the ischemic region, resulting in a significant reduction of neuronal cell death, decreased infarct volumes, and accelerated recovery. Importantly, neither leptin nor PEG-FBP influenced apoptotic signaling in brain ischemia. Here, we demonstrate that the systemic delivery of leptin-PEG-FBP presents a promising and viable strategy for treating cerebral ischemic stroke. Our approach not only highlights the therapeutic potential but also emphasizes the importance of overcoming BBB challenges to advance treatments for neurological disorders.
Collapse
Affiliation(s)
- Sungeun Chung
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea (Y.Y.); (M.L.)
| | - Yujong Yi
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea (Y.Y.); (M.L.)
| | - Irfan Ullah
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea (Y.Y.); (M.L.)
- Department of Internal Medicine, Yale University, New Haven, CT 06520, USA
| | - Kunho Chung
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea (Y.Y.); (M.L.)
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Seongjun Park
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea (Y.Y.); (M.L.)
| | - Jaeyeoung Lim
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea (Y.Y.); (M.L.)
| | - Chaeyeon Kim
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea (Y.Y.); (M.L.)
| | - Seon-Hong Pyun
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea (Y.Y.); (M.L.)
| | - Minkyung Kim
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea (Y.Y.); (M.L.)
| | - Dokyoung Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Minhyung Lee
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea (Y.Y.); (M.L.)
| | - Taiyoun Rhim
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea (Y.Y.); (M.L.)
| | - Sang-Kyung Lee
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea (Y.Y.); (M.L.)
| |
Collapse
|
2
|
Mackiewicz J, Lisek M, Boczek T. Targeting CaN/NFAT in Alzheimer's brain degeneration. Front Immunol 2023; 14:1281882. [PMID: 38077352 PMCID: PMC10701682 DOI: 10.3389/fimmu.2023.1281882] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 11/06/2023] [Indexed: 12/18/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by a progressive loss of cognitive functions. While the exact causes of this debilitating disorder remain elusive, numerous investigations have characterized its two core pathologies: the presence of β-amyloid plaques and tau tangles. Additionally, multiple studies of postmortem brain tissue, as well as results from AD preclinical models, have consistently demonstrated the presence of a sustained inflammatory response. As the persistent immune response is associated with neurodegeneration, it became clear that it may also exacerbate other AD pathologies, providing a link between the initial deposition of β-amyloid plaques and the later development of neurofibrillary tangles. Initially discovered in T cells, the nuclear factor of activated T-cells (NFAT) is one of the main transcription factors driving the expression of inflammatory genes and thus regulating immune responses. NFAT-dependent production of inflammatory mediators is controlled by Ca2+-dependent protein phosphatase calcineurin (CaN), which dephosphorylates NFAT and promotes its transcriptional activity. A substantial body of evidence has demonstrated that aberrant CaN/NFAT signaling is linked to several pathologies observed in AD, including neuronal apoptosis, synaptic deficits, and glia activation. In view of this, the role of NFAT isoforms in AD has been linked to disease progression at different stages, some of which are paralleled to diminished cognitive status. The use of classical inhibitors of CaN/NFAT signaling, such as tacrolimus or cyclosporine, or adeno-associated viruses to specifically inhibit astrocytic NFAT activation, has alleviated some symptoms of AD by diminishing β-amyloid neurotoxicity and neuroinflammation. In this article, we discuss the recent findings related to the contribution of CaN/NFAT signaling to the progression of AD and highlight the possible benefits of targeting this pathway in AD treatment.
Collapse
Affiliation(s)
| | | | - Tomasz Boczek
- Department of Molecular Neurochemistry, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
3
|
Pudjihartono N, Ho D, Golovina E, Fadason T, Kempa-Liehr AW, O'Sullivan JM. Juvenile idiopathic arthritis-associated genetic loci exhibit spatially constrained gene regulatory effects across multiple tissues and immune cell types. J Autoimmun 2023; 138:103046. [PMID: 37229810 DOI: 10.1016/j.jaut.2023.103046] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/04/2023] [Accepted: 04/15/2023] [Indexed: 05/27/2023]
Abstract
Juvenile idiopathic arthritis (JIA) is an autoimmune, inflammatory joint disease with complex genetic etiology. Previous GWAS have found many genetic loci associated with JIA. However, the biological mechanism behind JIA remains unknown mainly because most risk loci are located in non-coding genetic regions. Interestingly, increasing evidence has found that regulatory elements in the non-coding regions can regulate the expression of distant target genes through spatial (physical) interactions. Here, we used information on the 3D genome organization (Hi-C data) to identify target genes that physically interact with SNPs within JIA risk loci. Subsequent analysis of these SNP-gene pairs using data from tissue and immune cell type-specific expression quantitative trait loci (eQTL) databases allowed the identification of risk loci that regulate the expression of their target genes. In total, we identified 59 JIA-risk loci that regulate the expression of 210 target genes across diverse tissues and immune cell types. Functional annotation of spatial eQTLs within JIA risk loci identified significant overlap with gene regulatory elements (i.e., enhancers and transcription factor binding sites). We found target genes involved in immune-related pathways such as antigen processing and presentation (e.g., ERAP2, HLA class I and II), the release of pro-inflammatory cytokines (e.g., LTBR, TYK2), proliferation and differentiation of specific immune cell types (e.g., AURKA in Th17 cells), and genes involved in physiological mechanisms related to pathological joint inflammation (e.g., LRG1 in arteries). Notably, many of the tissues where JIA-risk loci act as spatial eQTLs are not classically considered central to JIA pathology. Overall, our findings highlight the potential tissue and immune cell type-specific regulatory changes contributing to JIA pathogenesis. Future integration of our data with clinical studies can contribute to the development of improved JIA therapy.
Collapse
Affiliation(s)
- N Pudjihartono
- The Liggins Institute, The University of Auckland, Auckland, New Zealand.
| | - D Ho
- The Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - E Golovina
- The Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - T Fadason
- The Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - A W Kempa-Liehr
- Department of Engineering Science, The University of Auckland, Auckland, New Zealand
| | - J M O'Sullivan
- The Liggins Institute, The University of Auckland, Auckland, New Zealand; The Maurice Wilkins Centre, The University of Auckland, Auckland, New Zealand; MRC Lifecourse Epidemiology Unit, University of Southampton, United Kingdom; Australian Parkinsons Mission, Garvan Institute of Medical Research, Sydney, New South Wales, 384 Victoria Street, Darlinghurst, NSW, 2010, Australia; A*STAR Singapore Institute for Clinical Sciences, Singapore, Singapore.
| |
Collapse
|
4
|
Saleki K, Banazadeh M, Miri NS, Azadmehr A. Triangle of cytokine storm, central nervous system involvement, and viral infection in COVID-19: the role of sFasL and neuropilin-1. Rev Neurosci 2021; 33:147-160. [PMID: 34225390 DOI: 10.1515/revneuro-2021-0047] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 05/27/2021] [Indexed: 02/07/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) is identified as the cause of coronavirus disease 2019 (COVID-19), and is often linked to extreme inflammatory responses by over activation of neutrophil extracellular traps (NETs), cytokine storm, and sepsis. These are robust causes for multi-organ damage. In particular, potential routes of SARS-CoV2 entry, such as angiotensin-converting enzyme 2 (ACE2), have been linked to central nervous system (CNS) involvement. CNS has been recognized as one of the most susceptible compartments to cytokine storm, which can be affected by neuropilin-1 (NRP-1). ACE2 is widely-recognized as a SARS-CoV2 entry pathway; However, NRP-1 has been recently introduced as a novel path of viral entry. Apoptosis of cells invaded by this virus involves Fas receptor-Fas ligand (FasL) signaling; moreover, Fas receptor may function as a controller of inflammation. Furthermore, NRP-1 may influence FasL and modulate cytokine profile. The neuroimmunological insult by SARS-CoV2 infection may be inhibited by therapeutic approaches targeting soluble Fas ligand (sFasL), cytokine storm elements, or related viral entry pathways. In the current review, we explain pivotal players behind the activation of cytokine storm that are associated with vast CNS injury. We also hypothesize that sFasL may affect neuroinflammatory processes and trigger the cytokine storm in COVID-19.
Collapse
Affiliation(s)
- Kiarash Saleki
- Student Research Committee, Babol University of Medical Sciences, Babol, 47176-47745, Iran
- USERN Office, Babol University of Medical Sciences, Babol, 47176-47745, Iran
- National Elite Foundation, Mazandaran Province Branch, Tehran, 48157-66435, Iran
| | - Mohammad Banazadeh
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, 76169-13555, Iran
| | - Niloufar Sadat Miri
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, 47176-47745, Iran
| | - Abbas Azadmehr
- National Elite Foundation, Mazandaran Province Branch, Tehran, 48157-66435, Iran
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, 47176-47745, Iran
- Medical Immunology Department, Babol University of Medical Sciences, Babol, 47176-47745, Iran
| |
Collapse
|
5
|
ADAS-viewer: web-based application for integrative analysis of multi-omics data in Alzheimer's disease. NPJ Syst Biol Appl 2021; 7:18. [PMID: 33741983 PMCID: PMC7979890 DOI: 10.1038/s41540-021-00177-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 02/16/2021] [Indexed: 11/08/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder and is represented by complicated biological mechanisms and complexity of brain tissue. Our understanding of the complicated molecular architecture that contributes to AD progression benefits from performing comprehensive and systemic investigations with multi-layered molecular and biological data from different brain regions. Since recently different independent studies generated various omics data in different brain regions of AD patients, multi-omics data integration can be a useful resource for better comprehensive understanding of AD. Here we present a web platform, ADAS-viewer, that provides researchers with the ability to comprehensively investigate and visualize multi-omics data from multiple brain regions of AD patients. ADAS-viewer offers means to identify functional changes in transcript and exon expression (i.e., alternative splicing) along with associated genetic or epigenetic regulatory effects. Specifically, it integrates genomic, transcriptomic, methylation, and miRNA data collected from seven different brain regions (cerebellum, temporal cortex, dorsolateral prefrontal cortex, frontal pole, inferior frontal gyrus, parahippocampal gyrus, and superior temporal gyrus) across three independent cohort datasets. ADAS-viewer is particularly useful as a web-based application for analyzing and visualizing multi-omics data across multiple brain regions at both transcript and exon level, allowing the identification of candidate biomarkers of Alzheimer's disease.
Collapse
|
6
|
Li J, Xu C, Zhang J, Jin C, Shi X, Zhang C, Jia S, Xu J, Gui X, Xing L, Lu L, Xu L. Identification of miRNA-Target Gene Pairs in the Parietal and Frontal Lobes of the Brain in Patients with Alzheimer's Disease Using Bioinformatic Analyses. Neurochem Res 2021; 46:964-979. [PMID: 33586092 DOI: 10.1007/s11064-020-03215-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/05/2020] [Accepted: 12/23/2020] [Indexed: 12/17/2022]
Abstract
Alzheimer's disease (AD) is a growing health concern worldwide. MicroRNAs (miRNAs) have been extensively studied in many diseases, including AD. To identify differentially expressed miRNAs (DEmiRNAs) and genes specific to AD, we used bioinformatic analyses to investigate candidate miRNA-mRNA pairs involved in the pathogenesis of AD. We focused on differentially expressed genes (DEGs) that are targets of DEmiRNAs. The GEO2R tool and the HISAT2-DESeq2 software were used to identify DEmiRNAs and DEGs. Bioinformatic tools available online, such as TAM and the Database for Annotation, Visualization and Integrated Discovery (DAVID), were used to perform functional annotation and enrichment analysis. Targets of miRNAs were predicted using the miRTarBase. The Search Tool for the Retrieval of Interacting Genes (STRING) and Cytoscape, which are available online, were utilized to construct protein-protein interaction (PPI) networks and identify hub genes. Furthermore, transcription factors (TFs) encoded by the DEGs were predicted using the TransmiR database and TF-miRNA-mRNA networks were constructed. Finally, the expression profile of a hub gene in peripheral blood mononuclear cells was compared between healthy individuals and AD patients. We identified 26 correlated miRNA-mRNA pairs. In the parietal lobe, miRNA-mRNA pairs involved in protein folding were enriched, and in the frontal lobe, miRNA-mRNA pairs involved in synaptic transmission, abnormal protein degradation, and apoptosis were enriched. In addition, HSP90AB1 in peripheral blood mononuclear cells was found to be significantly downregulated in AD patients, and this was consistent with its expression profile in the parietal lobe of AD patients. Our results provide brain region-specific changes in miRNA-mRNA associations in AD patients, further our understanding of potential underlying molecular mechanisms of AD, and reveal promising diagnostic and therapeutic targets for AD.
Collapse
Affiliation(s)
- Jiao Li
- Teaching Laboratory Center of Medicine and Life Science, Tongji University School of Medicine, Shanghai, 200092, China
| | - Chunli Xu
- Department of Neurology, The Seventh People's Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China
| | - Junfang Zhang
- Teaching Laboratory Center of Medicine and Life Science, Tongji University School of Medicine, Shanghai, 200092, China
| | - Caixia Jin
- Department of Biochemistry and Molecular Biology, School of Medicine, Tongji University, Shanghai, China
| | - Xiujuan Shi
- Department of Biochemistry and Molecular Biology, School of Medicine, Tongji University, Shanghai, China
| | - Chen Zhang
- Department of Laboratory Research Center, Tongji University School of Medicine, Shanghai, China
| | - Song Jia
- Teaching Laboratory Center of Medicine and Life Science, Tongji University School of Medicine, Shanghai, 200092, China
| | - Jie Xu
- Teaching Laboratory Center of Medicine and Life Science, Tongji University School of Medicine, Shanghai, 200092, China
| | - Xin Gui
- School of Life Science and Technology, Tongji University, Shanghai, China
| | - Libo Xing
- School of Life Science and Technology, Tongji University, Shanghai, China
| | - Lixia Lu
- Department of Biochemistry and Molecular Biology, School of Medicine, Tongji University, Shanghai, China
| | - Lei Xu
- Department of Biochemistry and Molecular Biology, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
7
|
Turkez H, Cacciatore I, Marinelli L, Fornasari E, Aslan ME, Cadirci K, Kahraman CY, Caglar O, Tatar A, Di Biase G, Hacimuftuoglu A, Di Stefano A, Mardinoglu A. Glycyl-L-Prolyl-L-Glutamate Pseudotripeptides for Treatment of Alzheimer's Disease. Biomolecules 2021; 11:biom11010126. [PMID: 33478054 PMCID: PMC7835747 DOI: 10.3390/biom11010126] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/13/2021] [Accepted: 01/15/2021] [Indexed: 12/15/2022] Open
Abstract
So far, there is no effective disease-modifying therapies for Alzheimer’s Disease (AD) in clinical practice. In this context, glycine-L-proline-L-glutamate (GPE) and its analogs may open the way for developing a novel molecule for treating neurodegenerative disorders, including AD. In turn, this study was aimed to investigate the neuroprotective potentials exerted by three novel GPE peptidomimetics (GPE1, GPE2, and GPE3) using an in vitro AD model. Anti-Alzheimer potentials were determined using a wide array of techniques, such as measurements of mitochondrial viability (MTT) and lactate dehydrogenase (LDH) release assays, determination of acetylcholinesterase (AChE), α-secretase and β-secretase activities, comparisons of total antioxidant capacity (TAC) and total oxidative status (TOS) levels, flow cytometric and microscopic detection of apoptotic and necrotic neuronal death, and investigating gene expression responses via PCR arrays involving 64 critical genes related to 10 different pathways. Our analysis showed that GPE peptidomimetics modulate oxidative stress, ACh depletion, α-secretase inactivation, apoptotic, and necrotic cell death. In vitro results suggested that treatments with novel GPE analogs might be promising therapeutic agents for treatment and/or or prevention of AD.
Collapse
Affiliation(s)
- Hasan Turkez
- Department of Medical Biology, Faculty of Medicine, Atatürk University, 25240 Erzurum, Turkey
- Correspondence: (H.T.); (A.M.)
| | - Ivana Cacciatore
- Department of Pharmacy, Univerisity “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti Scalo, Chieti, Italy; (I.C.); (L.M.); (E.F.); (G.D.B.); (A.D.S.)
| | - Lisa Marinelli
- Department of Pharmacy, Univerisity “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti Scalo, Chieti, Italy; (I.C.); (L.M.); (E.F.); (G.D.B.); (A.D.S.)
| | - Erika Fornasari
- Department of Pharmacy, Univerisity “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti Scalo, Chieti, Italy; (I.C.); (L.M.); (E.F.); (G.D.B.); (A.D.S.)
| | - Mehmet Enes Aslan
- Department of Molecular Biology and Genetics, Faculty of Science, Erzurum Technical University, 25200 Erzurum, Turkey; (M.E.A.); (O.C.)
| | - Kenan Cadirci
- Department of Internal Medicine, Erzurum Regional Training and Research Hospital, Health Sciences University, 25200 Erzurum, Turkey;
| | - Cigdem Yuce Kahraman
- Department of Medical Genetics, Faculty of Medicine, Atatürk University, 25240 Erzurum, Turkey; (C.Y.K.); (A.T.)
| | - Ozge Caglar
- Department of Molecular Biology and Genetics, Faculty of Science, Erzurum Technical University, 25200 Erzurum, Turkey; (M.E.A.); (O.C.)
| | - Abdulgani Tatar
- Department of Medical Genetics, Faculty of Medicine, Atatürk University, 25240 Erzurum, Turkey; (C.Y.K.); (A.T.)
| | - Giuseppe Di Biase
- Department of Pharmacy, Univerisity “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti Scalo, Chieti, Italy; (I.C.); (L.M.); (E.F.); (G.D.B.); (A.D.S.)
| | - Ahmet Hacimuftuoglu
- Department of Medical Pharmacology, Faculty of Medicine, Atatürk University, 25240 Erzurum, Turkey;
| | - Antonio Di Stefano
- Department of Pharmacy, Univerisity “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti Scalo, Chieti, Italy; (I.C.); (L.M.); (E.F.); (G.D.B.); (A.D.S.)
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH—Royal Institute of Technology, 24075 Stockholm, Sweden
- Centre for Host-Microbiome Interactions, Dental Institute, King’s College London, London SE1 9RT, UK
- Correspondence: (H.T.); (A.M.)
| |
Collapse
|
8
|
Zheng S, Rao J, Song Y, Zhang J, Xiao X, Fang EF, Yang Y, Niu Z. PharmKG: a dedicated knowledge graph benchmark for bomedical data mining. Brief Bioinform 2020; 22:6042240. [PMID: 33341877 DOI: 10.1093/bib/bbaa344] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/12/2020] [Accepted: 10/28/2020] [Indexed: 12/11/2022] Open
Abstract
Biomedical knowledge graphs (KGs), which can help with the understanding of complex biological systems and pathologies, have begun to play a critical role in medical practice and research. However, challenges remain in their embedding and use due to their complex nature and the specific demands of their construction. Existing studies often suffer from problems such as sparse and noisy datasets, insufficient modeling methods and non-uniform evaluation metrics. In this work, we established a comprehensive KG system for the biomedical field in an attempt to bridge the gap. Here, we introduced PharmKG, a multi-relational, attributed biomedical KG, composed of more than 500 000 individual interconnections between genes, drugs and diseases, with 29 relation types over a vocabulary of ~8000 disambiguated entities. Each entity in PharmKG is attached with heterogeneous, domain-specific information obtained from multi-omics data, i.e. gene expression, chemical structure and disease word embedding, while preserving the semantic and biomedical features. For baselines, we offered nine state-of-the-art KG embedding (KGE) approaches and a new biological, intuitive, graph neural network-based KGE method that uses a combination of both global network structure and heterogeneous domain features. Based on the proposed benchmark, we conducted extensive experiments to assess these KGE models using multiple evaluation metrics. Finally, we discussed our observations across various downstream biological tasks and provide insights and guidelines for how to use a KG in biomedicine. We hope that the unprecedented quality and diversity of PharmKG will lead to advances in biomedical KG construction, embedding and application.
Collapse
Affiliation(s)
- Shuangjia Zheng
- School of Data and Computer Science at the Sun Yat-Sen University
| | - Jiahua Rao
- School of Data and Computer Science at the Sun Yat-Sen University
| | - Ying Song
- School of Systems Science and Engineering at the Sun Yat-Sen University
| | | | | | - Evandro Fei Fang
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, Lørenskog, Norway
| | - Yuedong Yang
- School of Data and Computer Science and the National Super Computer Center at Guangzhou, Sun Yat-sen University, China
| | | |
Collapse
|
9
|
Selective Targeting of Non-nuclear Estrogen Receptors with PaPE-1 as a New Treatment Strategy for Alzheimer's Disease. Neurotox Res 2020; 38:957-966. [PMID: 33025361 PMCID: PMC7591444 DOI: 10.1007/s12640-020-00289-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/15/2020] [Accepted: 09/16/2020] [Indexed: 12/15/2022]
Abstract
Alzheimer’s disease (AD) is a multifactorial and severe neurodegenerative disorder characterized by progressive memory decline, the presence of Aβ plaques and tau tangles, brain atrophy, and neuronal loss. Available therapies provide moderate symptomatic relief but do not alter disease progression. This study demonstrated that PaPE-1, which has been designed to selectively activate non-nuclear estrogen receptors (ERs), has anti-AD capacity, as evidenced in a cellular model of the disease. In this model, the treatment of mouse neocortical neurons with Aβ (5 and 10 μM) induced apoptosis (loss of mitochondrial membrane potential, activation of caspase-3, induction of apoptosis-related genes and proteins) accompanied by increases in levels of reactive oxygen species (ROS) and lactate dehydrogenase (LDH) as well as reduced cell viability. Following 24 h of exposure, PaPE-1 inhibited Aβ-evoked effects, as shown by reduced parameters of neurotoxicity, oxidative stress, and apoptosis. Because PaPE-1 downregulated Aβ-induced Fas/FAS expression but upregulated that of Aβ-induced FasL, the role of PaPE-1 in controlling the external apoptotic pathway is controversial. However, PaPE-1 normalized Aβ-induced loss of mitochondrial membrane potential and restored the BAX/BCL2 ratio, suggesting that the anti-AD capacity of PaPE-1 particularly relies on inhibition of the mitochondrial apoptotic pathway. These data provide new evidence for an anti-AD strategy that utilizes the selective targeting of non-nuclear ERs with PaPE-1.
Collapse
|
10
|
Zuo H, Liu X, Li Y, Wang D, Hao Y, Yu C, Xu X, Peng R, Song T. The mitochondria/caspase-dependent apoptotic pathway plays a role in the positive effects of a power frequency electromagnetic field on Alzheimer's disease neuronal model. J Chem Neuroanat 2020; 109:101857. [PMID: 32918997 DOI: 10.1016/j.jchemneu.2020.101857] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/20/2020] [Accepted: 09/06/2020] [Indexed: 11/16/2022]
Abstract
In this study, rat pheochromocytoma (PC12) cells were induced into an Alzheimer's Disease (AD) neuronal model using nerve growth factor (NGF; 50 ng/mL) and Amyloid β25-35 (20 μmol/L). Changes in the morphological structure, cell viability, apoptosis rate, and expression of apoptosis-related protein induced by exposure to a power frequency electromagnetic field (PF-MF; 50 Hz, 100 μT, 24 h) were detected respectively by light and electron microscopy, the MTT assay, immunohistochemistry, flow cytometry and enzyme-linked immunosorbent assays. The results showed that 3-12 h after PF-MF exposure, the pathological injury was improved partly; metabolic activity was promoted and cell apoptosis was inhibited in the AD neuronal model. In addition, PF-MF exposure significantly inhibited the expression of Caspase8, Caspase3, and CytC, but increased the Bcl-2/Bax ratio of the AD neuronal model. Meanwhile, PF-MF seemed to have no effect on the expression of Fas and TNFR1. This study indicated that the mitochondria/caspase-dependent apoptotic pathway plays an important role in the positive effects of PF-MF on an AD neuronal model. The results suggested that PF-MF exposure might have potential therapeutic value for AD, and the underling molecular mechanisms still need further studies.
Collapse
Affiliation(s)
- Hongyan Zuo
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing 100850, China.
| | - Xiao Liu
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing 100850, China; Department ofPathology, Hainan Hospital of PLA General Hospital, Sanya 572013, China
| | - Yang Li
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing 100850, China; Anhui Medical University, Hefei 230032, China
| | - Dewen Wang
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Yanhui Hao
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Chao Yu
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Xinping Xu
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Ruiyun Peng
- Department of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Tao Song
- Beijing Key Laboratory of Bioelectromagnetics, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
11
|
Shukla M, Vincent B. The multi-faceted impact of methamphetamine on Alzheimer's disease: From a triggering role to a possible therapeutic use. Ageing Res Rev 2020; 60:101062. [PMID: 32304732 DOI: 10.1016/j.arr.2020.101062] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 03/05/2020] [Accepted: 03/28/2020] [Indexed: 12/15/2022]
Abstract
Although it has been initially synthesized for therapeutic purposes and currently FDA-approved and prescribed for obesity, attention-deficit/hyperactivity disorder, narcolepsy and depression, methamphetamine became a recreational drug that is nowadays massively manufactured illegally. Because it is a powerful and extremely addictive psychotropic agent, its abuse has turned out to become a major health problem worldwide. Importantly, the numerous effects triggered by this drug induce neurotoxicity in the brain ultimately leading to serious neurological impairments, tissue damage and neuropsychological disturbances that are reminiscent to most of the symptoms observed in Alzheimer's disease and other pathological manifestations in aging brain. In this context, there is a growing number of compelling evidence linking methamphetamine abuse with a higher probability of developing premature Alzheimer's disease and consequent neurodegeneration. This review proposes to establish a broad assessment of the effects that this drug can generate at the cellular and molecular levels in connection with the development of the age-related Alzheimer's disease. Altogether, the objective is to warn against the long-term effects that methamphetamine abuse may convey on young consumers and the increased risk of developing this devastating brain disorder at later stages of their lives, but also to discuss a more recently emerging concept suggesting a possible use of methamphetamine for treating this pathology under proper and strictly controlled conditions.
Collapse
|
12
|
Turkez H, Cacciatore I, Arslan ME, Fornasari E, Marinelli L, Di Stefano A, Mardinoglu A. Histidyl-Proline Diketopiperazine Isomers as Multipotent Anti-Alzheimer Drug Candidates. Biomolecules 2020; 10:biom10050737. [PMID: 32397415 PMCID: PMC7277666 DOI: 10.3390/biom10050737] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 04/26/2020] [Accepted: 04/29/2020] [Indexed: 12/13/2022] Open
Abstract
Cyclic dipeptides administered by both parenteral and oral routes are suggested as promising candidates for the treatment of neurodegeneration-related pathologies. In this study, we tested Cyclo (His-Pro) isomers (cHP1-4) for their anti-Alzheimer potential using a differentiated human neuroblastoma cell line (SH-SY5Y) as an Alzheimer’s disease (AD) experimental model. The SH-SY5Y cell line was differentiated by the application of all-trans retinoic acid (RA) to obtain mature neuron-like cells. Amyloid-beta 1-42 (Aβ1-42) peptides, the main effector in AD, were administered to the differentiated cell cultures to constitute the in vitro disease model. Next, we performed cell viability analyses 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and lactate dehydrogenase (LDH) release assays) to investigate the neuroprotective concentrations of cyclodipeptides using the in vitro AD model. We evaluated acetylcholinesterase (AChE), α- and β-secretase activities (TACE and BACE1), antioxidant potency, and apoptotic/necrotic properties and performed global gene expression analysis to understand the main mechanism behind the neuroprotective features of cHP1-4. Moreover, we conducted sister chromatid exchange (SCE), micronucleus (MN), and 8-hydroxy-2′-deoxyguanosine (8-OHdG) analyses to evaluate the genotoxic damage potential after applications with cHP1-4 on cultured human lymphocytes. Our results revealed that cHP1-4 isomers provide a different degree of neuroprotection against Aβ1-42-induced cell death on the in vitro AD model. The applications with cHP1-4 isomers altered the activity of AChE but not the activity of TACE and BACE1. Our analysis indicated that the cHP1-4 increased the total antioxidant capacity without altering total oxidative status levels in the cellular AD model and that cHP1-4 modulated the alterations of gene expressions by Aβ1-42 exposure. We also observed that cHP1-4 exhibited noncytotoxic and non-genotoxic features in cultured human whole blood cells. In conclusion, cHP1-4 isomers, especially cHP4, have been explored as novel promising therapeutics against AD.
Collapse
Affiliation(s)
- Hasan Turkez
- Department of Medical Biology, Faculty of Medicine, Atatürk University, 25240 Erzurum, Turkey
- Correspondence: (H.T.); (A.M.)
| | - Ivana Cacciatore
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, via dei Vestini 31, 66100 Chieti Scalo (CH), Italy; (I.C.); (E.F.); (L.M.); (A.D.S.)
| | - Mehmet Enes Arslan
- Department of Molecular Biology and Genetics, Faculty of Science, Erzurum Technical University, 25050 Erzurum, Turkey;
| | - Erika Fornasari
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, via dei Vestini 31, 66100 Chieti Scalo (CH), Italy; (I.C.); (E.F.); (L.M.); (A.D.S.)
| | - Lisa Marinelli
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, via dei Vestini 31, 66100 Chieti Scalo (CH), Italy; (I.C.); (E.F.); (L.M.); (A.D.S.)
| | - Antonio Di Stefano
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, via dei Vestini 31, 66100 Chieti Scalo (CH), Italy; (I.C.); (E.F.); (L.M.); (A.D.S.)
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH-Royal Institute of Technology, SE-17121 Stockholm, Sweden
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London SE1 9RT, UK
- Correspondence: (H.T.); (A.M.)
| |
Collapse
|
13
|
Wide Profiling of Circulating MicroRNAs in Spinocerebellar Ataxia Type 7. Mol Neurobiol 2019; 56:6106-6120. [PMID: 30721448 DOI: 10.1007/s12035-019-1480-y] [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: 10/05/2018] [Accepted: 01/10/2019] [Indexed: 12/28/2022]
Abstract
Spinocerebellar ataxia type 7 (SCA7), a neurodegenerative disease characterized by cerebellar ataxia and retinal degeneration, is caused by a CAG repeat expansion in the ATXN7 gene coding region. Disease onset and progression are highly variable between patients, thus identification of specific/sensitive biomarkers that can improve the monitoring of disease progression is an immediate need. Because altered expression of circulating microRNAs (miRNAs) has been shown in various neurological diseases, they could be useful biomarkers for SCA7. In this study, we showed, to our knowledge for the first time, the expression profile of circulating miRNAs in SCA7. Using the TaqMan profiling low density array (TLDA), we found 71 differentially expressed miRNAs in the plasma of SCA7 patients, compared with healthy controls. The reliability of TLDA data was validated independently by quantitative real-time polymerase chain reaction in an independent cohort of patients and controls. We identified four validated miRNAs that possesses the diagnostic value to discriminate between healthy controls and patients (hsa-let-7a-5p, hsa-let7e-5p, hsa-miR-18a-5p, and hsa-miR-30b-5p). The target genes of these four miRNAs were significantly enriched in cellular processes that are relevant to central nervous system function, including Fas-mediated cell-death, heparansulfate biosynthesis, and soluble-N-ethylmaleimide-sensitive factor activating protein receptor pathways. Finally, we identify a signature of four miRNAs associated with disease severity that discriminate between early onset and adult onset, highlighting their potential utility to surveillance disease progression. In summary, circulating miRNAs might provide accessible biomarkers for disease stage and progression and help to identify novel cellular processes involved in SCA7.
Collapse
|
14
|
Nazarian A, Yashin AI, Kulminski AM. Genome-wide analysis of genetic predisposition to Alzheimer's disease and related sex disparities. ALZHEIMERS RESEARCH & THERAPY 2019; 11:5. [PMID: 30636644 PMCID: PMC6330399 DOI: 10.1186/s13195-018-0458-8] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 12/06/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common cause of dementia in the elderly and the sixth leading cause of death in the United States. AD is mainly considered a complex disorder with polygenic inheritance. Despite discovering many susceptibility loci, a major proportion of AD genetic variance remains to be explained. METHODS We investigated the genetic architecture of AD in four publicly available independent datasets through genome-wide association, transcriptome-wide association, and gene-based and pathway-based analyses. To explore differences in the genetic basis of AD between males and females, analyses were performed on three samples in each dataset: males and females combined, only males, or only females. RESULTS Our genome-wide association analyses corroborated the associations of several previously detected AD loci and revealed novel significant associations of 35 single-nucleotide polymorphisms (SNPs) outside the chromosome 19q13 region at the suggestive significance level of p < 5E-06. These SNPs were mapped to 21 genes in 19 chromosomal regions. Of these, 17 genes were not associated with AD at genome-wide or suggestive levels of associations by previous genome-wide association studies. Also, the chromosomal regions corresponding to 8 genes did not contain any previously detected AD-associated SNPs with p < 5E-06. Our transcriptome-wide association and gene-based analyses revealed that 26 genes located in 20 chromosomal regions outside chromosome 19q13 had evidence of potential associations with AD at a false discovery rate of 0.05. Of these, 13 genes/regions did not contain any previously AD-associated SNPs at genome-wide or suggestive levels of associations. Most of the newly detected AD-associated SNPs and genes were sex specific, indicating sex disparities in the genetic basis of AD. Also, 7 of 26 pathways that showed evidence of associations with AD in our pathway-bases analyses were significant only in females. CONCLUSIONS Our findings, particularly the newly discovered sex-specific genetic contributors, provide novel insight into the genetic architecture of AD and can advance our understanding of its pathogenesis.
Collapse
Affiliation(s)
- Alireza Nazarian
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Erwin Mill Building, 2024 W. Main St., Durham, NC, 27705, USA.
| | - Anatoliy I Yashin
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Erwin Mill Building, 2024 W. Main St., Durham, NC, 27705, USA
| | - Alexander M Kulminski
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Erwin Mill Building, 2024 W. Main St., Durham, NC, 27705, USA.
| |
Collapse
|
15
|
Alexandraki KI, Apostolopoulos NV, Adamopoulos C, Stamouli E, Dalagiorgou G, Papaioannou TG, Analitis A, Karamanou M, Makrilakis K, Politis A, Piperi C. Differential Expression of Apoptotic and Low-Grade Inflammatory Markers in Alzheimer Disease Compared to Diabetes Mellitus Type 1 and 2. J Appl Lab Med 2018; 3:1003-1013. [PMID: 31639691 DOI: 10.1373/jalm.2018.027623] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 09/18/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Neuroinflammation, impaired brain insulin signaling, and neuronal apoptosis may be interrelated in the pathophysiology of people with Alzheimer disease (AD) and diabetes, either type 1 or 2 diabetes (T1D or T2D, respectively). METHODS We studied 116 patients: 41 with AD, 20 with T1D, 21 with T2D, and 34 healthy controls. The number (n) of cytokine-secreting peripheral blood mononuclear cells (PBMCs) before and after mitogenic stimulation was determined for interleukin 1β (IL1β), interleukin 6 (IL6), tumor necrosis factor (TNF) by the enzyme-linked-immuno-spot assay. Serum concentrations of C-reactive protein (CRP) and Fas ligand (FASLG) were determined by ELISA. RESULTS The studied subgroups did not differ in sex but differed in age. Higher CRP concentrations were detected in the AD group than in the T1D group (P = 0.02) and lower in controls (P < 0.001). The nPBMCs was higher in AD patients after stimulation than in basal conditions: after stimulation in nTNF (P < 0.001 vs T2D; P < 0.001 vs T1D; P = 0.001 vs control), nIL6 (P = 0.039 vs T2D; P < 0.001 vs T1D; P = 0.007 vs control), and nIL1β (P = 0.03 vs control). The nPBMCs increased after stimulation with ΡΜA in all the subgroups (P < 0.001). FASLG in the AD group displayed statistically higher concentrations than in all other subgroups (P < 0.001 vs T2D; P < 0.001 vs T1D; P = 0.012 vs control). The nPBMCs was positively correlated with plasma concentrations of FASLG in the AD subgroup. CONCLUSIONS Patients with AD display a low-grade systemic inflammation compared to people with diabetes. The FAS-FASLG pathway has a potential role because FASLG concentrations are positively correlated with the inflammatory response in AD. However, this positive correlation cannot be seen in people with diabetes, at least not with the apoptotic markers used in the present study.
Collapse
Affiliation(s)
- Krystallenia I Alexandraki
- Department of Biological Chemistry, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; .,Endocrine Unit, First Department of Propaedeutic Medicine, Laiko University Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Christos Adamopoulos
- Department of Biological Chemistry, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelia Stamouli
- Department of Biological Chemistry, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Georgia Dalagiorgou
- Department of Biological Chemistry, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Theodoros G Papaioannou
- First Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Antonios Analitis
- Department of Hygiene, Epidemiology and Medical Statistics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Konstantinos Makrilakis
- Diabetologic Center, First Department of Propedeutic Medicine, Laiko University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Antonios Politis
- First Department of Psychiatry, Eginitio Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christina Piperi
- Department of Biological Chemistry, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
16
|
Chi H, Chang HY, Sang TK. Neuronal Cell Death Mechanisms in Major Neurodegenerative Diseases. Int J Mol Sci 2018; 19:E3082. [PMID: 30304824 PMCID: PMC6213751 DOI: 10.3390/ijms19103082] [Citation(s) in RCA: 245] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/03/2018] [Accepted: 10/05/2018] [Indexed: 12/14/2022] Open
Abstract
Neuronal cell death in the central nervous system has always been a challenging process to decipher. In normal physiological conditions, neuronal cell death is restricted in the adult brain, even in aged individuals. However, in the pathological conditions of various neurodegenerative diseases, cell death and shrinkage in a specific region of the brain represent a fundamental pathological feature across different neurodegenerative diseases. In this review, we will briefly go through the general pathways of cell death and describe evidence for cell death in the context of individual common neurodegenerative diseases, discussing our current understanding of cell death by connecting with renowned pathogenic proteins, including Tau, amyloid-beta, alpha-synuclein, huntingtin and TDP-43.
Collapse
Affiliation(s)
- Hao Chi
- Institute of Biotechnology, National Tsing Hua University, Hsinchu City 30013, Taiwan.
| | - Hui-Yun Chang
- Institute of Systems Neuroscience, National Tsing Hua University, Hsinchu City 30013, Taiwan.
| | - Tzu-Kang Sang
- Institute of Biotechnology, National Tsing Hua University, Hsinchu City 30013, Taiwan.
| |
Collapse
|
17
|
Mihelčić M, Šimić G, Babić Leko M, Lavrač N, Džeroski S, Šmuc T, for the Alzheimer’s Disease Neuroimaging Initiative. Using redescription mining to relate clinical and biological characteristics of cognitively impaired and Alzheimer's disease patients. PLoS One 2017; 12:e0187364. [PMID: 29088293 PMCID: PMC5663625 DOI: 10.1371/journal.pone.0187364] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 10/18/2017] [Indexed: 11/18/2022] Open
Abstract
Based on a set of subjects and a collection of attributes obtained from the Alzheimer's Disease Neuroimaging Initiative database, we used redescription mining to find interpretable rules revealing associations between those determinants that provide insights about the Alzheimer's disease (AD). We extended the CLUS-RM redescription mining algorithm to a constraint-based redescription mining (CBRM) setting, which enables several modes of targeted exploration of specific, user-constrained associations. Redescription mining enabled finding specific constructs of clinical and biological attributes that describe many groups of subjects of different size, homogeneity and levels of cognitive impairment. We confirmed some previously known findings. However, in some instances, as with the attributes: testosterone, ciliary neurotrophic factor, brain natriuretic peptide, Fas ligand, the imaging attribute Spatial Pattern of Abnormalities for Recognition of Early AD, as well as the levels of leptin and angiopoietin-2 in plasma, we corroborated previously debatable findings or provided additional information about these variables and their association with AD pathogenesis. Moreover, applying redescription mining on ADNI data resulted with the discovery of one largely unknown attribute: the Pregnancy-Associated Protein-A (PAPP-A), which we found highly associated with cognitive impairment in AD. Statistically significant correlations (p ≤ 0.01) were found between PAPP-A and clinical tests: Alzheimer's Disease Assessment Scale, Clinical Dementia Rating Sum of Boxes, Mini Mental State Examination, etc. The high importance of this finding lies in the fact that PAPP-A is a metalloproteinase, known to cleave insulin-like growth factor binding proteins. Since it also shares similar substrates with A Disintegrin and the Metalloproteinase family of enzymes that act as α-secretase to physiologically cleave amyloid precursor protein (APP) in the non-amyloidogenic pathway, it could be directly involved in the metabolism of APP very early during the disease course. Therefore, further studies should investigate the role of PAPP-A in the development of AD more thoroughly.
Collapse
Affiliation(s)
- Matej Mihelčić
- Division of Electronics, Ruđer Bošković Institute, Zagreb, Croatia
- Jožef Stefan International Postgraduate School, Ljubljana, Slovenia
| | - Goran Šimić
- Department for Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia
| | - Mirjana Babić Leko
- Department for Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia
| | - Nada Lavrač
- Jožef Stefan International Postgraduate School, Ljubljana, Slovenia
- Department of Knowledge Technologies, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Sašo Džeroski
- Jožef Stefan International Postgraduate School, Ljubljana, Slovenia
- Department of Knowledge Technologies, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Tomislav Šmuc
- Division of Electronics, Ruđer Bošković Institute, Zagreb, Croatia
| | | |
Collapse
|
18
|
Samy DM, Ismail CA, Nassra RA, Zeitoun TM, Nomair AM. Downstream modulation of extrinsic apoptotic pathway in streptozotocin-induced Alzheimer's dementia in rats: Erythropoietin versus curcumin. Eur J Pharmacol 2015; 770:52-60. [PMID: 26638997 DOI: 10.1016/j.ejphar.2015.11.046] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 10/28/2015] [Accepted: 11/25/2015] [Indexed: 01/18/2023]
Abstract
Erythropoietin and curcumin showed promising neuroprotective effects in various models of Alzheimer's dementia. This study was designed to compare the beneficial effects of erythropoietin and/or curcumin in intracerebro-ventricular (ICV) streptozotocin-induced Alzheimer's like disease in rats. Rats received ICV injection of either saline (control, n=8 rats), or streptozotocin. Three weeks following surgery, streptozotocin-injected rats were assigned into 4 groups (8 rats each); vehicle, curcumin (80mg/kg/day, orally), erythropoietin (500 IU/kg every other day, intraperitoneally) and combined (curcumin and erythropoietin)-treated groups. After 3 months of treatment, rats were subjected to neurobehavioral testing, and then killed for biochemical and histological assessment of hippocampus. Fas ligand protein and caspase-8 activity as mediators of extrinsic apoptotic pathway, oxidative stress markers (malondialdehyde and reduced glutathione) and β-amyloid (1-40 and 1-42) peptides were measured. The results showed that administration of erythropoietin suppressed extrinsic apoptosis better than curcumin, while curcumin was more effective in combating oxidative stress in ICV-streptozotocin injected rats. Both erythropoietin and curcumin treatments (individually or combined) equally reduced the hippocampal β-amyloid accumulation and improved cognitive impairment in Morris water maze and passive avoidance tasks. The combined treatment was the most effective in ameliorating apoptosis and oxidative stress rather than behavioral responses or β-amyloid burden. In conclusion, ICV-streptozotocin-induced Alzheimer's dementia activates hippocampal Fas ligand-mediated apoptosis, which could be reduced by erythropoietin and/or curcumin treatment. Curcumin supplementation alone could ameliorate cognitive deficits and reverse biochemical alterations in ICV-streptozotocin Alzheimer's rat model without the hazardous polycythemic effect of long-term erythropoietin injection.
Collapse
Affiliation(s)
- Doaa M Samy
- Department of Medical Physiology, Faculty of Medicine, Al-Moassat Hospital, University of Alexandria, Egypt.
| | - Cherine A Ismail
- Department of Clinical Pharmacology, Faculty of Medicine, Al-Moassat Hospital, University of Alexandria, Egypt.
| | - Rasha A Nassra
- Department of Medical Biochemistry, Faculty of Medicine, Al-Moassat Hospital, University of Alexandria, Egypt.
| | - Teshreen M Zeitoun
- Department of Histology and Cell Biology, Faculty of Medicine, Al-Moassat Hospital, University of Alexandria, Egypt.
| | - Azhar M Nomair
- Department of Chemical Pathology, Medical Research Institute, University of Alexandria, Egypt.
| |
Collapse
|
19
|
Mufson EJ, Malek-Ahmadi M, Perez SE, Chen K. Braak staging, plaque pathology, and APOE status in elderly persons without cognitive impairment. Neurobiol Aging 2015; 37:147-153. [PMID: 26686670 DOI: 10.1016/j.neurobiolaging.2015.10.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Revised: 10/13/2015] [Accepted: 10/14/2015] [Indexed: 12/21/2022]
Abstract
Clinico-pathological studies reveal that some elderly people with no cognitive impairment have high burdens of neurofibrillary tangles (NFTs), a pathology associated with Alzheimer's disease. We examined a total of 123 elderly participants without dementia and free of other neurological disorders or pathologies who at autopsy were classified as Braak NFT stages of I-V. We found that women were significantly more likely to have a high Braak score. Significant associations were found between high Braak scores and entorhinal cortex amyloid load, combined hippocampal and entorhinal cortex amyloid loads with perceptual speed in the low Braak group after adjusting for age, gender and apolipoprotein E ε4 status. Elderly with preserved cognitive function show a wide range of Braak scores and plaque pathology similar to that seen in prodromal and frank Alzheimer's disease at death. These data suggest that some older people with extensive NFT and plaque pathology demonstrate brain resilience or reserve leading to preserved cognitive function.
Collapse
Affiliation(s)
- Elliott J Mufson
- Alzheimer's Disease Research Laboratory, Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, USA.
| | | | - Sylvia E Perez
- Alzheimer's Disease Research Laboratory, Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Kewei Chen
- Banner Alzheimer's Institute, Phoenix, AZ, USA
| |
Collapse
|
20
|
Ramanan VK, Nho K, Shen L, Risacher SL, Kim S, McDonald BC, Farlow MR, Foroud TM, Gao S, Soininen H, Kłoszewska I, Mecocci P, Tsolaki M, Vellas B, Lovestone S, Aisen PS, Petersen RC, Jack CR, Shaw LM, Trojanowski JQ, Weiner MW, Green RC, Toga AW, De Jager PL, Yu L, Bennett DA, Saykin AJ, the Alzheimer's Disease Neuroimaging Initiative (ADNI). FASTKD2 is associated with memory and hippocampal structure in older adults. Mol Psychiatry 2015; 20:1197-204. [PMID: 25385369 PMCID: PMC4427556 DOI: 10.1038/mp.2014.142] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 09/05/2014] [Accepted: 09/10/2014] [Indexed: 12/15/2022]
Abstract
Memory impairment is the cardinal early feature of Alzheimer's disease, a highly prevalent disorder whose causes remain only partially understood. To identify novel genetic predictors, we used an integrative genomics approach to perform the largest study to date of human memory (n=14 781). Using a genome-wide screen, we discovered a novel association of a polymorphism in the pro-apoptotic gene FASTKD2 (fas-activated serine/threonine kinase domains 2; rs7594645-G) with better memory performance and replicated this finding in independent samples. Consistent with a neuroprotective effect, rs7594645-G carriers exhibited increased hippocampal volume and gray matter density and decreased cerebrospinal fluid levels of apoptotic mediators. The MTOR (mechanistic target of rapamycin) gene and pathways related to endocytosis, cholinergic neurotransmission, epidermal growth factor receptor signaling and immune regulation, among others, also displayed association with memory. These findings nominate FASTKD2 as a target for modulating neurodegeneration and suggest potential mechanisms for therapies to combat memory loss in normal cognitive aging and dementia.
Collapse
Affiliation(s)
- Vijay K Ramanan
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA,Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA,Medical Scientist Training Program, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kwangsik Nho
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Li Shen
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA,Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Shannon L. Risacher
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sungeun Kim
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Brenna C. McDonald
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA,Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA,Indiana Alzheimer's Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Martin R. Farlow
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA,Indiana Alzheimer's Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Tatiana M. Foroud
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA,Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA,Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA,Indiana Alzheimer's Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sujuan Gao
- Indiana Alzheimer's Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA,Department of Biostatistics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Hilkka Soininen
- On behalf of the AddNeuroMed Consortium,Department of Neurology, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Iwona Kłoszewska
- On behalf of the AddNeuroMed Consortium,Medical University of Lodz, Lodz, Poland
| | - Patrizia Mecocci
- On behalf of the AddNeuroMed Consortium,Institute of Gerontology and Geriatrics, University of Perugia, Perugia, Italy
| | - Magda Tsolaki
- On behalf of the AddNeuroMed Consortium,3rd Department of Neurology, Aristotle University, Thessaloniki, Greece
| | - Bruno Vellas
- On behalf of the AddNeuroMed Consortium,INSERM U 558, University of Toulouse, Toulouse, France
| | - Simon Lovestone
- On behalf of the AddNeuroMed Consortium,University of Oxford, Department of Psychiatry, Oxford, UK
| | - Paul S. Aisen
- Department of Neuroscience, University of California-San Diego, San Diego, CA, USA
| | | | - Clifford R. Jack
- Department of Radiology, Mayo Clinic Minnesota, Rochester, MN, USA
| | - Leslie M. Shaw
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA,Institute on Aging, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - John Q. Trojanowski
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA,Institute on Aging, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Michael W. Weiner
- Departments of Radiology, Medicine, and Psychiatry, University of California-San Francisco, San Francisco, CA, USA,Department of Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Robert C. Green
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Arthur W. Toga
- Laboratory of Neuro Imaging, Keck School of Medicine, University of Southern California, Los Angeles, CA USA
| | - Philip L. De Jager
- Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Departments of Neurology and Psychiatry, Brigham and Women's Hospital, Boston, MA, USA,Harvard Medical School, Boston, MA, USA,Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Lei Yu
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - David A. Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Andrew J. Saykin
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA,Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA,Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA,Indiana Alzheimer's Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA,Correspondence to: Dr. Andrew J. Saykin, IU Health Neuroscience Center, Suite 4100 Indiana University School of Medicine 355 West 16th Street, Indianapolis, IN 46202, USA , Phone (317)963-7501, Fax (317)963-7547
| | | |
Collapse
|
21
|
Zhu J, Su J, Liu R, Yang J. Relationship between the FAS gene A-670G polymorphism and Alzheimer's disease: a meta-analysis. Aging Clin Exp Res 2015; 27:563-71. [PMID: 25809055 DOI: 10.1007/s40520-015-0351-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 02/16/2015] [Indexed: 12/28/2022]
Abstract
BACKGROUNDS AND AIMS The pathogenetic mechanism of Alzheimer's disease (AD) is still unknown; however, gene polymorphism may play a critical role in the pathogenesis of AD. The aim of this meta-analysis was to evaluate the association between FAS gene A-670G polymorphism and AD. METHODS We searched all related subjects in PubMed, Embase, Cochrane Library, SinoMed, and the China Knowledge Resource Integrated Database and identified seven studies reporting a relationship between the A-670G polymorphism in the FAS gene and AD. RESULTS A total of 1512 cases and 1707 controls were included in the seven studies. The meta-analyses results suggested no significant association between the A-670G polymorphism and AD in any genetic models. When a subgroup analysis was conducted by ethnicity, the A-670G polymorphism was also not relevant to AD. However, when stratified by age, the GG genotype increased the risk of early-onset AD. We also found that the A-670G polymorphism was related to patients with AD who carried the apolipoprotein-E ε4 allele in three genetic models. CONCLUSIONS To sum up, our data suggested that the FAS gene A-670G polymorphism may not be associated with AD. When a subgroup analysis was conducted by ethnicity, the A-670G polymorphism was also not related with AD in Asian and Caucasian population. However, the FAS-670 GG genotype may increase the risk of AD in the younger population (age, ≤65 years). Furthermore, we found that the A-670G polymorphism was related to patients with AD who carried the APOE4 allele in dominant, allele and homozygous models.
Collapse
|
22
|
Kärkkäinen V, Pomeshchik Y, Savchenko E, Dhungana H, Kurronen A, Lehtonen S, Naumenko N, Tavi P, Levonen AL, Yamamoto M, Malm T, Magga J, Kanninen KM, Koistinaho J. Nrf2 regulates neurogenesis and protects neural progenitor cells against Aβ toxicity. Stem Cells 2015; 32:1904-16. [PMID: 24753106 DOI: 10.1002/stem.1666] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2013] [Accepted: 01/15/2014] [Indexed: 12/25/2022]
Abstract
Neural stem/progenitor cells (NPCs) proliferate and produce new neurons in neurogenic areas throughout the lifetime. While these cells represent potential therapeutic treatment of neurodegenerative diseases, regulation of neurogenesis is not completely understood. We show that deficiency of nuclear factor erythroid 2-related factor (Nrf2), a transcription factor induced in response to oxidative stress, prevents the ischemia-induced increase in newborn neurons in the subgranular zone of the dentate gyrus. Consistent with this finding, the growth of NPC neurospheres was increased by lentivirus-mediated overexpression of Nrf2 gene or by treatment with pyrrolidine dithiocarbamate (PDTC), an Nrf2 activating compound. Also, neuronal differentiation of NPCs was increased by Nrf2 overexpression or PDTC treatment but reduced by Nrf2 deficiency. To investigate the impact of Nrf2 on NPCs in Alzheimer's disease (AD), we treated NPCs with amyloid beta (Aβ), a toxic peptide associated with neurodegeneration and cognitive abnormalities in AD. We found that Aβ1-42-induced toxicity and reduction in neurosphere proliferation were prevented by Nrf2 overexpression, while Nrf2 deficiency enhanced the Aβ1-42-induced reduction of neuronal differentiation. On the other hand, Aβ1-40 had no effect on neurosphere proliferation in wt NPCs but increased the proliferation of Nrf2 overexpressing neurospheres and reduced it in Nrf2-deficient neurospheres. These results suggest that Nrf2 is essential for neuronal differentiation of NPCs, regulates injury-induced neurogenesis and provides protection against Aβ-induced NPC toxicity.
Collapse
Affiliation(s)
- Virve Kärkkäinen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Krauthausen M, Kummer MP, Zimmermann J, Reyes-Irisarri E, Terwel D, Bulic B, Heneka MT, Müller M. CXCR3 promotes plaque formation and behavioral deficits in an Alzheimer's disease model. J Clin Invest 2014; 125:365-78. [PMID: 25500888 DOI: 10.1172/jci66771] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Accepted: 11/10/2014] [Indexed: 01/28/2023] Open
Abstract
Chemokines are important modulators of neuroinflammation and neurodegeneration. In the brains of Alzheimer's disease (AD) patients and in AD animal models, the chemokine CXCL10 is found in high concentrations, suggesting a pathogenic role for this chemokine and its receptor, CXCR3. Recent studies aimed at addressing the role of CXCR3 in neurological diseases indicate potent, but diverse, functions for CXCR3. Here, we examined the impact of CXCR3 in the amyloid precursor protein (APP)/presenilin 1 (PS1) transgenic mouse model of AD. We found that, compared with control APP/PSI animals, plaque burden and Aβ levels were strongly reduced in CXCR3-deficient APP/PS1 mice. Analysis of microglial phagocytosis in vitro and in vivo demonstrated that CXCR3 deficiency increased the microglial uptake of Aβ. Application of a CXCR3 antagonist increased microglial Aβ phagocytosis, which was associated with reduced TNF-α secretion. Moreover, in CXCR3-deficient APP/PS1 mice, microglia exhibited morphological activation and reduced plaque association, and brain tissue from APP/PS1 animals lacking CXCR3 had reduced concentrations of proinflammatory cytokines compared with controls. Further, loss of CXCR3 attenuated the behavioral deficits observed in APP/PS1 mice. Together, our data indicate that CXCR3 signaling mediates development of AD-like pathology in APP/PS1 mice and suggest that CXCR3 has potential as a therapeutic target for AD.
Collapse
|
24
|
Ettcheto M, Junyent F, de Lemos L, Pallas M, Folch J, Beas-Zarate C, Verdaguer E, Gómez-Sintes R, Lucas JJ, Auladell C, Camins A. Mice Lacking Functional Fas Death Receptors Are Protected from Kainic Acid-Induced Apoptosis in the Hippocampus. Mol Neurobiol 2014; 52:120-9. [PMID: 25119776 DOI: 10.1007/s12035-014-8836-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 07/25/2014] [Indexed: 01/08/2023]
Abstract
The Fas receptor (FasR)/Fas ligand (FasL) system plays a significant role in the process of neuronal loss in neurological disorders. Thus, in the present study, we used a real-time PCR array focused apoptosis (Mouse Apoptosis RT(2) PCR Array) to study the role of the Fas pathway in the apoptotic process that occurs in a kainic acid (KA) mice experimental model. In fact, significant changes in the transcriptional activity of a total of 23 genes were found in the hippocampus of wild-type C57BL/6 mice after 12 h of KA treatment compared to untreated mice. Among the up-regulated genes, we found key factors involved in the extrinsic apoptotic pathway, such as tnf, fas and fasL, and also in caspase genes (caspase -4, caspase-8 and caspase-3). To discern the importance of the FasR/FasL pathway, mice lacking the functional Fas death receptor (lpr) were also treated with KA. After 24 h of neurotoxin treatment, lpr mice exhibited a reduced number of apoptotic positive cells, determined by the terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL) method in different regions of the hippocampus, when compared to wild-type mice. In addition, treatment of lpr mice with KA did not produce significant changes in the transcriptional activity of genes related to apoptosis in the hippocampus, either in the fas and fas ligand genes or in caspase-4 and caspase-8 and the executioner caspase-3 genes, as occurred in wild-type C57BL/6 mice. Thus, these data provide direct evidence that Fas signalling plays a key role in the induction of apoptosis in the hippocampus following KA treatment, making the inhibition of the death receptor pathway a potentially suitable target for excitotoxicity neuroprotection in neurological conditions such as epilepsy.
Collapse
Affiliation(s)
- Miren Ettcheto
- Unitat de Farmacologia i Farmacognòsia Facultat de Farmàcia, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda/Diagonal 643, E-08028, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Neuronal apoptosis and motor deficits in mice with genetic inhibition of GSK-3 are Fas-dependent. PLoS One 2013; 8:e70952. [PMID: 23940673 PMCID: PMC3734180 DOI: 10.1371/journal.pone.0070952] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 06/24/2013] [Indexed: 11/19/2022] Open
Abstract
Glycogen synthase kinase-3 (GSK-3) inhibitors have been postulated as useful therapeutic tools for the treatment of chronic neurodegenerative and neuropsychiatric diseases. Nevertheless the clinical use of these inhibitors has been limited by their common side effects. Lithium, a non-selective GSK-3 inhibitor has been classically administered to treat bipolar patients but its prescription is decreasing due to its frequent side effects such as hand tremor. This toxicity seems to be higher in the elderly and a clinical trial with lithium for Alzheimer’s disease was stopped due to high rate of discontinuation. We have previously described a mechanism for the adverse effects of chronic lithium that involves neuronal apoptosis via Fas signaling. As lithium inhibits many other enzymatic activities such as inositol monophosphatase and histone deacetylase, here we aim to genetically test whether GSK-3 inhibition induces those adverse effects through Fas receptor. For this purpose we took advantage of a transgenic mouse line with decreased GSK-3 activity (Tet/DN-GSK-3 mice) that shows increased rate of neuronal apoptosis as well as motor deficits and brought it to a Fas deficient background (lpr mice). We found that apoptosis induced by GSK-3 inhibition was absent in Fas deficient background. Interestingly, motor deficits were also absent in Fas deficient Tet/DN-GSK-3 mice. These results demonstrate that Fas signaling contributes to the neurological toxicity of GSK-3 inhibition and suggest that a combination of GSK-3 inhibitors with blockers of Fas signaling could help to improve the application of GSK-3 inhibitors to clinics.
Collapse
|
26
|
Wong HKA, Veremeyko T, Patel N, Lemere CA, Walsh DM, Esau C, Vanderburg C, Krichevsky AM. De-repression of FOXO3a death axis by microRNA-132 and -212 causes neuronal apoptosis in Alzheimer's disease. Hum Mol Genet 2013; 22:3077-92. [PMID: 23585551 DOI: 10.1093/hmg/ddt164] [Citation(s) in RCA: 218] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Alzheimer's disease (AD) is a multifactorial and fatal neurodegenerative disorder for which the mechanisms leading to profound neuronal loss are incompletely recognized. MicroRNAs (miRNAs) are recently discovered small regulatory RNA molecules that repress gene expression and are increasingly acknowledged as prime regulators involved in human brain pathologies. Here we identified two homologous miRNAs, miR-132 and miR-212, downregulated in temporal cortical areas and CA1 hippocampal neurons of human AD brains. Sequence-specific inhibition of miR-132 and miR-212 induces apoptosis in cultured primary neurons, whereas their overexpression is neuroprotective against oxidative stress. Using primary neurons and PC12 cells, we demonstrate that miR-132/212 controls cell survival by direct regulation of PTEN, FOXO3a and P300, which are all key elements of AKT signaling pathway. Silencing of these three target genes by RNAi abrogates apoptosis caused by the miR-132/212 inhibition. We further demonstrate that mRNA and protein levels of PTEN, FOXO3a, P300 and most of the direct pro-apoptotic transcriptional targets of FOXO3a are significantly elevated in human AD brains. These results indicate that the miR-132/miR-212/PTEN/FOXO3a signaling pathway contributes to AD neurodegeneration.
Collapse
Affiliation(s)
- Hon-Kit Andus Wong
- Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Harvard Institutes of Medicine, Boston, MA 02115, USA
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Region-specific expression of Dickkopf-like1 in the adult brain. Abbreviated title: Dkkl1 in the adult brain. Neurosci Lett 2013; 535:84-9. [PMID: 23295900 DOI: 10.1016/j.neulet.2012.12.040] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 12/15/2012] [Accepted: 12/20/2012] [Indexed: 11/20/2022]
Abstract
In the adult, the dickkopf family member Dickkopf-like1 (Dkkl1) has been described as a testicular protein involved in the regulation of spermatocyte apoptosis. However, microarray studies additionally suggested that Dkkl1 regulation is involved in various tumors including high grade gliomas. Since investigations of Dkkl1 in the adult central nervous system are lacking, we analyzed Dkkl1 expression in the adult mouse brain and found a region specific expression pattern with a profoundly high cortical expression. Analysis of transgenic mice in which the lacZ gene was inserted into the Dkkl1 locus further pointed to NeuN-positive neurons as the main source of Dkkl1 in the normal adult brain. In Dkkl1(-/-) mutant mice, gross brain morphology as well as hippocampal and cortical lamination appeared normal. Similarly, neuronal density in cortical layer V was not altered. Thus, Dkkl1 may not be essential for normal brain organization, but could exert import functions during pathological conditions such as tumorigenesis and cancer progression.
Collapse
|
28
|
Almeida A. Genetic determinants of neuronal vulnerability to apoptosis. Cell Mol Life Sci 2013; 70:71-88. [PMID: 22695677 PMCID: PMC11113535 DOI: 10.1007/s00018-012-1029-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 04/22/2012] [Accepted: 05/07/2012] [Indexed: 12/18/2022]
Abstract
Apoptosis is a common mode of cell death that contributes to neuronal loss associated with neurodegeneration. Single-nucleotide polymorphisms (SNPs) in chromosomal DNA are contributing factors dictating natural susceptibility of humans to disease. Here, the most common SNPs affecting neuronal vulnerability to apoptosis are reviewed in the context of neurological disorders. Polymorphic variants in genes encoding apoptotic proteins, either from the extrinsic (FAS, TNF-α, CASP8) or the intrinsic (BAX, BCL2, CASP3, CASP9) pathways could be highly valuable in the diagnosis of neurodegenerative diseases and stroke. Interestingly, the Arg72Pro SNP in TP53, the gene encoding tumor suppressor p53, was recently revealed a biomarker of poor prognosis in stroke due to its ability to modulate neuronal apoptotic death. Search for new SNPs responsible for genetic variability to apoptosis will ensure the implementation of novel diagnostic and prognostic tools, as well as therapeutic strategies against neurological diseases.
Collapse
Affiliation(s)
- Angeles Almeida
- Instituto de Investigación Biomédica de Salamanca, Hospital Universitario de Salamanca, 37007, Salamanca, Spain.
| |
Collapse
|
29
|
Javier Miguel-Hidalgo J, Paul IA, Wanzo V, Banerjee PK. Memantine prevents cognitive impairment and reduces Bcl-2 and caspase 8 immunoreactivity in rats injected with amyloid β1–40. Eur J Pharmacol 2012; 692:38-45. [DOI: 10.1016/j.ejphar.2012.07.032] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Revised: 07/09/2012] [Accepted: 07/12/2012] [Indexed: 02/01/2023]
|
30
|
Renin inhibitor aliskiren exerts neuroprotection against amyloid beta-peptide toxicity in rat cortical neurons. Neurochem Int 2012; 61:369-77. [DOI: 10.1016/j.neuint.2012.05.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Revised: 04/24/2012] [Accepted: 05/09/2012] [Indexed: 12/20/2022]
|
31
|
Inhibition of the NFAT pathway alleviates amyloid β neurotoxicity in a mouse model of Alzheimer's disease. J Neurosci 2012; 32:3176-92. [PMID: 22378890 DOI: 10.1523/jneurosci.6439-11.2012] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Amyloid β (Aβ) peptides, the main pathological species associated with Alzheimer's disease (AD), disturb intracellular calcium homeostasis, which in turn activates the calcium-dependent phosphatase calcineurin (CaN). CaN activation induced by Aβ leads to pathological morphological changes in neurons, and overexpression of constitutively active calcineurin is sufficient to generate a similar phenotype, even without Aβ. Here, we tested the hypothesis that calcineurin mediates neurodegenerative effects via activation of the nuclear transcription factor of activated T-cells (NFAT). We found that both spine loss and dendritic branching simplification induced by Aβ exposure were mimicked by constitutively active NFAT, and abolished when NFAT activation was blocked using the genetically encoded inhibitor VIVIT. When VIVIT was specifically addressed to the nucleus, identical beneficial effects were observed, thus enforcing the role of NFAT transcriptional activity in Aβ-related neurotoxicity. In vivo, when VIVIT or its nuclear counterpart were overexpressed in a transgenic model of Alzheimer's disease via a gene therapy approach, the spine loss and neuritic abnormalities observed in the vicinity of amyloid plaques were blocked. Overall, these results suggest that NFAT/calcineurin transcriptional cascades contribute to Aβ synaptotoxicity, and may provide a new specific set of pathways for neuroprotective strategies.
Collapse
|
32
|
Mines MA, Beurel E, Jope RS. Regulation of cell survival mechanisms in Alzheimer's disease by glycogen synthase kinase-3. Int J Alzheimers Dis 2011; 2011:861072. [PMID: 21629713 PMCID: PMC3100684 DOI: 10.4061/2011/861072] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2011] [Accepted: 03/09/2011] [Indexed: 11/27/2022] Open
Abstract
A pivotal role has emerged for glycogen synthase kinase-3 (GSK3) as an important contributor to Alzheimer's disease pathology. Evidence for the involvement of GSK3 in Alzheimer's disease pathology and neuronal loss comes from studies of GSK3 overexpression, GSK3 localization studies, multiple relationships between GSK3 and amyloid β-peptide (Aβ), interactions between GSK3 and the microtubule-associated tau protein, and GSK3-mediated apoptotic cell death. Apoptotic signaling proceeds by either an intrinsic pathway or an extrinsic pathway. GSK3 is well established to promote intrinsic apoptotic signaling induced by many insults, several of which may contribute to neuronal loss in Alzheimer's disease. Particularly important is evidence that GSK3 promotes intrinsic apoptotic signaling induced by Aβ. GSK3 appears to promote intrinsic apoptotic signaling by modulating proteins in the apoptosis signaling pathway and by modulating transcription factors that regulate the expression of proteins involved in apoptosis. Thus, GSK3 appears to contribute to several neuropathological mechanisms in Alzheimer's disease, including apoptosis-mediated neuronal loss.
Collapse
Affiliation(s)
- Marjelo A Mines
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Sparks Center 1057, 1720 Seventh Avenue South, Birmingham, AL 35294-0017, USA
| | | | | |
Collapse
|
33
|
Craig-Schapiro R, Kuhn M, Xiong C, Pickering EH, Liu J, Misko TP, Perrin RJ, Bales KR, Soares H, Fagan AM, Holtzman DM. Multiplexed immunoassay panel identifies novel CSF biomarkers for Alzheimer's disease diagnosis and prognosis. PLoS One 2011; 6:e18850. [PMID: 21526197 PMCID: PMC3079734 DOI: 10.1371/journal.pone.0018850] [Citation(s) in RCA: 172] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2010] [Accepted: 03/21/2011] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Clinicopathological studies suggest that Alzheimer's disease (AD) pathology begins ∼10-15 years before the resulting cognitive impairment draws medical attention. Biomarkers that can detect AD pathology in its early stages and predict dementia onset would, therefore, be invaluable for patient care and efficient clinical trial design. We utilized a targeted proteomics approach to discover novel cerebrospinal fluid (CSF) biomarkers that can augment the diagnostic and prognostic accuracy of current leading CSF biomarkers (Aβ42, tau, p-tau181). METHODS AND FINDINGS Using a multiplexed Luminex platform, 190 analytes were measured in 333 CSF samples from cognitively normal (Clinical Dementia Rating [CDR] 0), very mildly demented (CDR 0.5), and mildly demented (CDR 1) individuals. Mean levels of 37 analytes (12 after Bonferroni correction) were found to differ between CDR 0 and CDR>0 groups. Receiver-operating characteristic curve analyses revealed that small combinations of a subset of these markers (cystatin C, VEGF, TRAIL-R3, PAI-1, PP, NT-proBNP, MMP-10, MIF, GRO-α, fibrinogen, FAS, eotaxin-3) enhanced the ability of the best-performing established CSF biomarker, the tau/Aβ42 ratio, to discriminate CDR>0 from CDR 0 individuals. Multiple machine learning algorithms likewise showed that the novel biomarker panels improved the diagnostic performance of the current leading biomarkers. Importantly, most of the markers that best discriminated CDR 0 from CDR>0 individuals in the more targeted ROC analyses were also identified as top predictors in the machine learning models, reconfirming their potential as biomarkers for early-stage AD. Cox proportional hazards models demonstrated that an optimal panel of markers for predicting risk of developing cognitive impairment (CDR 0 to CDR>0 conversion) consisted of calbindin, Aβ42, and age. CONCLUSIONS/SIGNIFICANCE Using a targeted proteomic screen, we identified novel candidate biomarkers that complement the best current CSF biomarkers for distinguishing very mildly/mildly demented from cognitively normal individuals. Additionally, we identified a novel biomarker (calbindin) with significant prognostic potential.
Collapse
Affiliation(s)
- Rebecca Craig-Schapiro
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Max Kuhn
- Neuroscience Research Unit, Pfizer Global Research and Development, Groton, Connecticut, United States of America
- Neuroscience Research Unit, Pfizer Global Research and Development, St. Louis, Missouri, United States of America
| | - Chengjie Xiong
- The Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Eve H. Pickering
- Neuroscience Research Unit, Pfizer Global Research and Development, Groton, Connecticut, United States of America
- Neuroscience Research Unit, Pfizer Global Research and Development, St. Louis, Missouri, United States of America
| | - Jingxia Liu
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Thomas P. Misko
- Neuroscience Research Unit, Pfizer Global Research and Development, Groton, Connecticut, United States of America
- Neuroscience Research Unit, Pfizer Global Research and Development, St. Louis, Missouri, United States of America
| | - Richard J. Perrin
- The Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Division of Neuropathology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Kelly R. Bales
- Neuroscience Research Unit, Pfizer Global Research and Development, Groton, Connecticut, United States of America
- Neuroscience Research Unit, Pfizer Global Research and Development, St. Louis, Missouri, United States of America
| | - Holly Soares
- Neuroscience Research Unit, Pfizer Global Research and Development, Groton, Connecticut, United States of America
- Neuroscience Research Unit, Pfizer Global Research and Development, St. Louis, Missouri, United States of America
| | - Anne M. Fagan
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- The Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - David M. Holtzman
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- The Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri, United States of America
| |
Collapse
|
34
|
Braskie MN, Ringman JM, Thompson PM. Neuroimaging measures as endophenotypes in Alzheimer's disease. Int J Alzheimers Dis 2011; 2011:490140. [PMID: 21547229 PMCID: PMC3087508 DOI: 10.4061/2011/490140] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Revised: 01/08/2011] [Accepted: 02/07/2011] [Indexed: 01/06/2023] Open
Abstract
Late onset Alzheimer's disease (AD) is moderately to highly heritable. Apolipoprotein E allele ε4 (APOE4) has been replicated consistently as an AD risk factor over many studies, and recently confirmed variants in other genes such as CLU, CR1, and PICALM each increase the lifetime risk of AD. However, much of the heritability of AD remains unexplained. AD is a complex disease that is diagnosed largely through neuropsychological testing, though neuroimaging measures may be more sensitive for detecting the incipient disease stages. Difficulties in early diagnosis and variable environmental contributions to the disease can obscure genetic relationships in traditional case-control genetic studies. Neuroimaging measures may be used as endophenotypes for AD, offering a reliable, objective tool to search for possible genetic risk factors. Imaging measures might also clarify the specific mechanisms by which proposed risk factors influence the brain.
Collapse
Affiliation(s)
- Meredith N Braskie
- Laboratory of Neuro Imaging, Department of Neurology, UCLA School of Medicine, 635 Charles Young Drive South, Suite 225, Los Angeles, CA 90095, USA
| | | | | |
Collapse
|
35
|
Rocha de Paula M, Gómez Ravetti M, Berretta R, Moscato P. Differences in abundances of cell-signalling proteins in blood reveal novel biomarkers for early detection of clinical Alzheimer's disease. PLoS One 2011; 6:e17481. [PMID: 21479255 PMCID: PMC3063784 DOI: 10.1371/journal.pone.0017481] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2010] [Accepted: 02/06/2011] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND In November 2007 a study published in Nature Medicine proposed a simple test based on the abundance of 18 proteins in blood to predict the onset of clinical symptoms of Alzheimer's Disease (AD) two to six years before these symptoms manifest. Later, another study, published in PLoS ONE, showed that only five proteins (IL-1, IL-3, EGF, TNF- and G-CSF) have overall better prediction accuracy. These classifiers are based on the abundance of 120 proteins. Such values were standardised by a Z-score transformation, which means that their values are relative to the average of all others. METHODOLOGY The original datasets from the Nature Medicine paper are further studied using methods from combinatorial optimisation and Information Theory. We expand the original dataset by also including all pair-wise differences of z-score values of the original dataset ("metafeatures"). Using an exact algorithm to solve the resulting Feature Set problem, used to tackle the feature selection problem, we found signatures that contain either only features, metafeatures or both, and evaluated their predictive performance on the independent test set. CONCLUSIONS It was possible to show that a specific pattern of cell signalling imbalance in blood plasma has valuable information to distinguish between NDC and AD samples. The obtained signatures were able to predict AD in patients that already had a Mild Cognitive Impairment (MCI) with up to 84% of sensitivity, while maintaining also a strong prediction accuracy of 90% on a independent dataset with Non Demented Controls (NDC) and AD samples. The novel biomarkers uncovered with this method now confirms ANG-2, IL-11, PDGF-BB, CCL15/MIP-1; and supports the joint measurement of other signalling proteins not previously discussed: GM-CSF, NT-3, IGFBP-2 and VEGF-B.
Collapse
Affiliation(s)
- Mateus Rocha de Paula
- Centre for Bioinformatics, Biomarker Discovery & Information-Based Medicine, The University of Newcastle, Callaghan, Australia
| | - Martín Gómez Ravetti
- Departamento de Engenharia de Produção, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Regina Berretta
- Centre for Bioinformatics, Biomarker Discovery & Information-Based Medicine, The University of Newcastle, Callaghan, Australia
| | - Pablo Moscato
- Centre for Bioinformatics, Biomarker Discovery & Information-Based Medicine, The University of Newcastle, Callaghan, Australia
| |
Collapse
|
36
|
Couturier J, Morel M, Pontcharraud R, Gontier V, Fauconneau B, Paccalin M, Page G. Interaction of double-stranded RNA-dependent protein kinase (PKR) with the death receptor signaling pathway in amyloid beta (Abeta)-treated cells and in APPSLPS1 knock-in mice. J Biol Chem 2010; 285:1272-82. [PMID: 19889624 PMCID: PMC2801255 DOI: 10.1074/jbc.m109.041954] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2009] [Revised: 10/27/2009] [Indexed: 01/05/2023] Open
Abstract
For 10 years, research has focused on signaling pathways controlling translation to explain neuronal death in Alzheimer Disease (AD). Previous studies demonstrated in different cellular and animal models and AD patients that translation is down-regulated by the activation of double-stranded RNA-dependent protein kinase (PKR). Among downstream factors of PKR, the Fas-associated protein with a death domain (FADD) and subsequent activated caspase-8 are responsible for PKR-induced apoptosis in recombinant virus-infected cells. However, no studies have reported the role of PKR in death receptor signaling in AD. The aim of this project is to determine physical and functional interactions of PKR with FADD in amyloid-beta peptide (Abeta) neurotoxicity and in APP(SL)PS1 KI transgenic mice. In SH-SY5Y cells, results showed that Abeta42 induced a large increase in phosphorylated PKR and FADD levels and a physical interaction between PKR and FADD in the nucleus, also observed in the cortex of APP(SL)PS1 KI mice. However, PKR gene silencing or treatment with a specific PKR inhibitor significantly prevented the increase in pT(451)-PKR and pS(194)-FADD levels in SH-SY5Y nuclei and completely inhibited activities of caspase-3 and -8. The contribution of PKR in neurodegeneration through the death receptor signaling pathway may support the development of therapeutics targeting PKR to limit neuronal death in AD.
Collapse
Affiliation(s)
- Julien Couturier
- From the Research Group on Brain Aging, GReViC EA 3808, University of Poitiers, 6 rue de la Milétrie, BP 199, 86034 Poitiers Cedex, France
| | - Milena Morel
- From the Research Group on Brain Aging, GReViC EA 3808, University of Poitiers, 6 rue de la Milétrie, BP 199, 86034 Poitiers Cedex, France
| | - Raymond Pontcharraud
- From the Research Group on Brain Aging, GReViC EA 3808, University of Poitiers, 6 rue de la Milétrie, BP 199, 86034 Poitiers Cedex, France
| | - Virginie Gontier
- From the Research Group on Brain Aging, GReViC EA 3808, University of Poitiers, 6 rue de la Milétrie, BP 199, 86034 Poitiers Cedex, France
| | - Bernard Fauconneau
- From the Research Group on Brain Aging, GReViC EA 3808, University of Poitiers, 6 rue de la Milétrie, BP 199, 86034 Poitiers Cedex, France
| | - Marc Paccalin
- From the Research Group on Brain Aging, GReViC EA 3808, University of Poitiers, 6 rue de la Milétrie, BP 199, 86034 Poitiers Cedex, France
- the Department of Geriatrics, Poitiers University Hospital, 2 rue de la Milétrie, BP 577, 86021 Poitiers Cedex, France, and
- the Clinical Investigation Center, CIC INSERM 802, Poitiers University Hospital, Poitiers, France
| | - Guylène Page
- From the Research Group on Brain Aging, GReViC EA 3808, University of Poitiers, 6 rue de la Milétrie, BP 199, 86034 Poitiers Cedex, France
| |
Collapse
|
37
|
The FAS gene, brain volume, and disease progression in Alzheimer's disease. Alzheimers Dement 2009; 6:118-24. [PMID: 19766542 DOI: 10.1016/j.jalz.2009.05.663] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2009] [Revised: 05/04/2009] [Accepted: 05/04/2009] [Indexed: 11/23/2022]
Abstract
OBJECTIVE We sought to identify single-nucleotide polymorphisms (SNPs) associated with Alzheimer's disease (AD) progression and brain volume. METHODS Ninety-seven SNPs were genotyped in 243 subjects from a longitudinal study of healthy aging. Subjects who received a diagnosis of cognitive impairment (CI) at any study visit (before their most recent visit) and had DNA in the study's DNA bank were included. Progression of AD was defined as the duration from onset of CI to diagnosis of AD. Association of each of the 97 SNPs with AD progression was tested via Cox model. Those SNPs meeting a criterion of nominal significance (P < 0.05) for association with AD progression were reassessed to account for multiple testing by repeating the marker selection process in 10,000 random permutations. Next, the association between the one SNP that survived the multiple-testing adjustment and brain volume was determined by multiple regression analysis in a subgroup of subjects for whom magnetic-resonance imaging (MRI)-derived brain-volume data were available. Brain volumes were adjusted for age at MRI, gender, and time from MRI to onset of CI. RESULTS The minor allele of rs1468063 in the FAS gene, which is member 6 of the tumor necrosis factor receptor superfamily, was significantly associated with faster AD progression after adjustment for multiple testing (P(permutation) = 0.049). The same allele in rs1468063 was associated with smaller brain volumes and larger ventricular volumes (P = 0.02 and 0.04, respectively). CONCLUSIONS The FAS gene, which plays a role in apoptosis, may be associated with AD by modulating the apoptosis and neuronal loss secondary to AD neuropathology.
Collapse
|
38
|
Erten-Lyons D, Woltjer RL, Dodge H, Nixon R, Vorobik R, Calvert JF, Leahy M, Montine T, Kaye J. Factors associated with resistance to dementia despite high Alzheimer disease pathology. Neurology 2009; 72:354-60. [PMID: 19171833 DOI: 10.1212/01.wnl.0000341273.18141.64] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Autopsy series have shown that some elderly people remain with normal cognitive function during life despite having high burdens of pathologic lesions associated with Alzheimer disease (AD) at death. Understanding why these individuals show no cognitive decline, despite high AD pathologic burdens, may be key to discovery of neuroprotective mechanisms. METHODS A total of 36 subjects who on autopsy had Braak stage V or VI and moderate or frequent neuritic plaque scores based on Consortium to Establish a Registry for Alzheimer's Disease (CERAD) standards were included. Twelve had normal cognitive function and 24 a diagnosis of AD before death. Demographic characteristics, clinical and pathologic data, as well as antemortem brain volumes were compared between the groups. RESULTS In multiple regression analysis, antemortem hippocampal and total brain volumes were significantly larger in the group with normal cognitive function after adjusting for gender, age at MRI, time from MRI to death, Braak stage, CERAD neuritic plaque score, and overall presence of vascular disease. CONCLUSION Larger brain and hippocampal volumes were associated with preserved cognitive function during life despite a high burden of Alzheimer disease (AD) pathologic lesions at death. A better understanding of processes that lead to preservation of brain volume may provide important clues for the discovery of mechanisms that protect the elderly from AD.
Collapse
|
39
|
Jantas D, Lason W. Protective effect of memantine against Doxorubicin toxicity in primary neuronal cell cultures: influence a development stage. Neurotox Res 2009; 15:24-37. [PMID: 19384585 DOI: 10.1007/s12640-009-9002-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2008] [Revised: 09/10/2008] [Accepted: 09/10/2008] [Indexed: 01/09/2023]
Abstract
One of the serious unwanted effects of the anthracycline anticancer drug doxorubicin (Dox, adriamycin) is its neurotoxicity, which can be evoked by the activation of extracellular (FAS/CD95/Apo-1) pathway of apoptosis in cells. Since memantine, a clinically used N-methyl-D: -aspartic acid (NMDA) receptor antagonist, shows antiapoptotic action in several models of neuronal cell damage, in this study we evaluated the effect of memantine on the cell death induced by Dox in primary neuronal cell cultures. First, we investigated the effect of different concentrations of Dox (0.1-5 microM) on mouse neocortical, hippocampal, striatal, and cerebellar neurons on 7- and 12-day in vitro (DIV). The 7 DIV neuronal cell cultures were more prone to Dox-induced cell death than 12 DIV cultures. The cerebellar neurons were the most resistant to Dox-induced apoptosis in comparison to neuronal cell cultures derived from the forebrain. Memantine (0.1-2 microM) attenuated the Dox-evoked lactate dehydrogenase release in 7 DIV neuronal cell cultures with no significant effect on 12 DIV cultures. The ameliorating effect of memantine on Dox-mediated cell death was also confirmed by an increase in cell viability measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide reduction assay. There was no effect of memantine on Dox-induced caspase-8 and -3 activity and Dox-evoked decrease in mitochondrial potential, although attenuation in the number of cells with apoptotic DNA fragmentation was observed. We also showed that the antiapoptotic effect of memantine in our model was NMDA receptor-independent, since two other antagonists of this receptor, MK-801 and AP-5, did not attenuate Dox-induced cell death. Furthermore, memantine did not influence the Dox-evoked increase in cytoplasmic Ca2+ level. The obtained data suggest developmental regulation of both, the Dox-mediated neurotoxicity and efficacy of memantine in alleviating the Dox-induced cell damage in neuronal cell cultures. Moreover, this neuroprotective effect of memantine seems not to be dependent on caspase-3 activity and on the antagonistic action on NMDA receptor.
Collapse
Affiliation(s)
- D Jantas
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Smetna 12, 31-343, Krakow, Poland,
| | | |
Collapse
|
40
|
Riley DE, Krieger JN. Embryonic nervous system genes predominate in searches for dinucleotide simple sequence repeats flanked by conserved sequences. Gene 2008; 429:74-9. [PMID: 18952158 DOI: 10.1016/j.gene.2008.09.025] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2008] [Revised: 09/18/2008] [Accepted: 09/23/2008] [Indexed: 12/15/2022]
Abstract
To study evolution of dinucleotide simple sequence repeats (diSSRs) we searched recently available mammalian genomes for UTR-localized diSSRs with conserved upstream flanking sequences (CFS). There were 252 reported Homo sapiens genes containing the repeats (AC)n, (GT)n, (AG)n or (CT)n in their UTRs including 22 (8.7%) with diSSR-upstream flanking sequences conserved comparing divergent mammalian lineages represented by Homo sapiens and the marsupial, Monodelphis domestica. Of these 22 genes, 19 had known functions including 18 (95%) that proved critical for mammalian nervous systems (Fishers exact test, P<0.0001). The remaining gene, Cd2ap, proved critical for development of kidney podocytes, cells that have multiple similarities to neurons. Gene functions included voltage and chloride channels, synapse-associated proteins, neurotransmitter receptors, axon and dendrite pathfinders, a NeuroD potentiator and other neuronal activities. Repeat length polymorphism was confirmed for 68% of CFS diSSRs even though these repeats were nestled among highly conserved sequences. This finding supports a hypothesis that SSR polymorphism has functional implications. A parallel study was performed on the self-complementary diSSRs (AT)n and (GC)n. When flanked by conserved sequences, the self-complementary diSSR (AT)n was also associated with genes expressed in the developing nervous system. Our findings implicate functional roles for diSSRs in nervous system development.
Collapse
Affiliation(s)
- Donald E Riley
- Department of Urology, University of Washington, Seattle, WA 98195, USA.
| | | |
Collapse
|
41
|
Death receptor Fas (CD95) signaling in the central nervous system: tuning neuroplasticity? Trends Neurosci 2008; 31:478-86. [DOI: 10.1016/j.tins.2008.06.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2008] [Revised: 06/26/2008] [Accepted: 06/27/2008] [Indexed: 12/20/2022]
|
42
|
Mao G, Tan J, Gao W, Shi Y, Cui MZ, Xu X. Both the N-terminal fragment and the protein-protein interaction domain (PDZ domain) are required for the pro-apoptotic activity of presenilin-associated protein PSAP. Biochim Biophys Acta Gen Subj 2008; 1780:696-708. [PMID: 18291114 DOI: 10.1016/j.bbagen.2008.01.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2007] [Revised: 01/16/2008] [Accepted: 01/17/2008] [Indexed: 11/25/2022]
Abstract
Presenilin-associated protein (PSAP) was originally identified as a PS1-associated, PDZ domain protein. In a subsequent study, PSAP was found to be a mitochondrial apoptotic molecule. In this study, we cloned the PSAP gene and found that it is composed of 12 exons and localizes on chromosome 6. To better understand the structure and function of PSAP, we have generated a series of antibodies that recognize different regions of PSAP. Using these antibodies, we found that PSAP is expressed in four isoforms as a result of differential splicing of exon 8 in addition to the use of either the first or the second ATG codon as the start codon. We also found that all these isoforms are localized in the mitochondria and are pro-apoptotic. Furthermore, our data revealed that the PDZ domain and N-terminal fragment are required for the pro-apoptotic activity of PSAP.
Collapse
Affiliation(s)
- Guozhang Mao
- Department of Pathobiology, College of Veterinary Medicine, The University of Tennessee, 2407 River Dr., Knoxville, TN 37996, USA
| | | | | | | | | | | |
Collapse
|
43
|
Zhang S, Liu H, Yu H, Cooper GJS. Fas-associated death receptor signaling evoked by human amylin in islet beta-cells. Diabetes 2008; 57:348-56. [PMID: 17977957 DOI: 10.2337/db07-0849] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE Aggregation of human amylin (hA) into beta-sheet-containing oligomers is linked to islet beta-cell dysfunction and the pathogenesis of type 2 diabetes. Here, we investigated possible contributions of Fas-associated death-receptor signaling to the mechanism of hA-evoked beta-cell apoptosis. RESEARCH DESIGN AND METHODS We measured responses to hA in isolated mouse islets and two insulinoma cell lines, wherein we measured Fas/Fas ligand (FasL) and Fas-associated death domain (FADD) expression by quantitative RT-PCR, Western blotting, and immunofluorescence staining. We used two anti-Fas/FasL blocking antibodies and the Fas/FasL antagonist Kp7-6 to probe roles of Fas interactions in the regulation of apoptosis in hA-treated beta-cells and measured Kp7-6-mediated effects on beta-sheet formation and aggregation using circular dichroism and thioflavin-T binding. RESULTS hA treatment stimulated Fas and FADD expression in beta-cells. Both blocking antibodies suppressed hA-evoked apoptosis but did not modify its aggregation. Therefore, Fas receptor interactions played a critical role in induction of this pathway. Interestingly, hA-evoked beta-cell apoptosis was suppressed and rescued by Kp7-6, which also impaired hA beta-sheet formation. CONCLUSIONS This is the first report linking hA-evoked induction and activation of Fas and FADD to beta-cell apoptosis. We have identified a Fas/FasL antagonist, Kp7-6, as a potent inhibitor of hA aggregation and related beta-cell death. These results also support an interaction between hA and Fas on the surface of apoptotic beta-cells. Increased expression and activation of Fas in beta-cells could constitute a molecular event common to the pathogenesis of both type 1 and type 2 diabetes, although the mode of pathway activation may differ between these common forms of diabetes.
Collapse
Affiliation(s)
- Shaoping Zhang
- School of Biological Sciences, University of Auckland, Level 4, 3A Symonds St., Private Bag 92019, Auckland 1142, New Zealand
| | | | | | | |
Collapse
|
44
|
Caspase activation in fetal rat brain following experimental intrauterine inflammation. Brain Res 2008; 1200:138-45. [PMID: 18289516 DOI: 10.1016/j.brainres.2008.01.045] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2007] [Revised: 01/01/2008] [Accepted: 01/03/2008] [Indexed: 11/21/2022]
Abstract
Intrauterine inflammation has been implicated in developmental brain injuries, including the development of periventricular leukomalacia (PVL) and cerebral palsy (CP). Previous studies in our rat model of intrauterine inflammation demonstrated apoptotic cell death in fetal brains within the first 5 days after lipopolysaccharide (LPS) administration to mothers and eventual dysmyelination. Cysteine-containing, aspartate-specific proteases, or caspases, are proteins involved with apoptosis through both intracellular (intrinsic pathway) and extracellular (extrinsic pathway) mechanisms. We hypothesized that cell death in our model would occur mainly via activation of the extrinsic pathway. We further hypothesized that Fas, a member of the tumor necrosis factor receptor (TNFR) superfamily, would be increased and the death inducing signaling complex (DISC) would be detectable. Pregnant rats were injected intracervically with LPS at E15 and immunoblotting, immunohistochemical and immunoprecipitation analyses were performed. The presence of the activated form of the effector caspase (caspase-3) was observed 24 h after LPS administration. Caspase activity assays demonstrated rapid increases in (i) caspases-9 and -10 within 1 h, (ii) caspase-8 at 2 h and (iii) caspase-3 at 4 h. At 24 h after LPS, activated caspase-3(+)/Fas(+) cells were observed within the developing white matter. Lastly, the DISC complex (caspase-8, Fas and Fas-associated death domain (FADD)) was observed within 30 min by immunoprecipitation. Apoptosis in our model occurs via both extrinsic and intrinsic pathways, and activation of Fas may play a role. Understanding the mechanisms of cell death in models of intrauterine inflammation may affect development of future strategies to mitigate these injuries in children.
Collapse
|
45
|
Sajan FD, Martiniuk F, Marcus DL, Frey WH, Hite R, Bordayo EZ, Freedman ML. Apoptotic gene expression in Alzheimer's disease hippocampal tissue. Am J Alzheimers Dis Other Demen 2007; 22:319-28. [PMID: 17712163 PMCID: PMC10846078 DOI: 10.1177/1533317507302447] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Alzheimer's disease (AD) is the major cause of dementia, accounting for 50% to 70% of the late-onset patients, with 17 to 20 million affected. It is characterized by neurofibrillary tangles, neuronal loss, and amyloid plaques in tissues of the cortex, hippocampus, and amygdala. Apoptosis or programmed cell death appears in the progression of AD. In this study, we investigated the gene expression of 14 apoptotic genes (E2F1, p21/WAF, ICE-LAP3, Fas Antigen, CPP-32, GADD153, ICE-beta, c-Fos, c-Jun, Bax-alpha, Bcl-2, Bcl-(x)L, BAK, and p53) in 5 normal and 6 AD human hippocampal tissues, using reverse transcription-polymerase chain reaction. Our results show an upregulation of gene expression in AD patients for c-Fos and BAK. ICE-beta, c-Jun, Bax-alpha, Bcl-x(L), p53, and GADD153 were found to be upregulated in some AD samples but were not detected or downregulated in other AD or normal samples. No gene expression was found for E2F1 , p21/WAF, ICE-LAP3, Fas Antigen, CPP32, or Bcl-2. These results indicate significant increases in c-Fos , c-Jun, and Bak; therefore, we suggest that these genes may be critical in the apoptotic cascades of AD.
Collapse
Affiliation(s)
- Farrah D Sajan
- Division of Geriatrics, New York University School of Medicne, New York, NY 10016, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Fernández M, Segura MF, Solé C, Colino A, Comella JX, Ceña V. Lifeguard/neuronal membrane protein 35 regulates Fas ligand-mediated apoptosis in neurons via microdomain recruitment. J Neurochem 2007; 103:190-203. [PMID: 17635665 DOI: 10.1111/j.1471-4159.2007.04767.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Fas ligand (FasL)-receptor system plays an essential role in regulating cell death in the developing nervous system, and it has been implicated in neurodegenerative and inflammatory responses in the CNS. Lifeguard (LFG) is a protein highly expressed in the hippocampus and the cerebellum, and it shows a particularly interesting regulation by being up-regulated during postnatal development and in the adult. We show that over-expression of LFG protected cortical neurons from FasL-induced apoptosis and decreased caspase-activation. Reduction of endogenous LFG expression by small interfering RNA sensitized cerebellar granular neurons to FasL-induced cell death and caspase-8 activation, and also increased sensitivity of cortical neurons. In differentiated cerebellar granular neurons, protection from FasL-induced cell death could be attributed exclusively to LFG and appears to be independent of FLICE inhibitor protein. Thus, LFG is an endogenous inhibitor of FasL-mediated neuronal death and it mediates the FasL resistance of CNS differentiated neurons. Finally, we also demonstrate that LFG is detected in lipid rafts microdomains, where it may interact with Fas receptor and regulate FasL-activated signaling pathways.
Collapse
Affiliation(s)
- Miriam Fernández
- Unidad Asociada Neurodeath, UCLM-CSIC, Departamento de Ciencias Médicas, Universidad de Castilla-La Mancha, Albacete, Spain
| | | | | | | | | | | |
Collapse
|
47
|
Shin JH, Cho SI, Lim HR, Lee JK, Lee YA, Noh JS, Joo IS, Kim KW, Gwag BJ. Concurrent administration of Neu2000 and lithium produces marked improvement of motor neuron survival, motor function, and mortality in a mouse model of amyotrophic lateral sclerosis. Mol Pharmacol 2007; 71:965-75. [PMID: 17105868 DOI: 10.1124/mol.106.030676] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The Fas pathway and oxidative stress mediate neuronal death in stroke and may contribute to neurodegenerative disease. We tested the hypothesis that these two factors synergistically produce spinal motor neuron degeneration in amyotrophic lateral sclerosis (ALS). Levels of reactive oxygen species were increased in motor neurons from ALS mice compared with wild-type mice at age 10 weeks, before symptom onset. The proapoptotic proteins Fas, Fas-associated death domain, caspase 8, and caspase 3 were also elevated. Oral administration of 2-hydroxy-5-(2,3,5,6-tetrafluoro-4-trifluoromethyl-benzylamino)-benzoic acid (Neu2000), a potent antioxidant, blocked the increase in reactive oxygen species but only slightly reduced activation of proapoptotic proteins. Administration of lithium carbonate (Li(+)), a mood stabilizer that prevents apoptosis, blocked the apoptosis machinery without preventing oxidative stress. Neu2000 or Li(+) alone significantly enhanced survival time and motor function and together had an additive effect. These findings provide evidence that jointly targeting oxidative stress and Fas-mediated apoptosis can prevent neuronal loss and motor dysfunction in ALS.
Collapse
Affiliation(s)
- Jin Hee Shin
- Department of Pharmacology, Ajou University School of Medicine, Sawon, Korea 442-749
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Takagi Y, Kikuta KI, Nozaki K, Fujimoto M, Hayashi J, Hashimoto N. Neuronal expression of Fas-associated death domain protein and caspase-8 in the perinidal parenchyma of cerebral arteriovenous malformations. J Neurosurg 2007; 106:275-82. [PMID: 17410712 DOI: 10.3171/jns.2007.106.2.275] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Object
The expression and localization of phosphorylated Fas-associated death domain protein (pFADD) and cleaved caspase-8 was examined in human cerebral arteriovenous malformations (AVMs). The authors focused on the perinidal parenchyma to clarify the effect of AVMs on perinidal brain tissue.
Methods
Seventeen cerebral AVMs were analyzed using immunohistochemical methods. Specimens were removed from patients during surgical procedures. The characteristics of the areas that stained positively for pFADD or cleaved caspase-8 were also assessed using an image analysis system. Eleven (65%) of the 17 lesions demonstrated anti-pFADD immunoreactivity and 12 (71%) showed anti–cleaved caspase-8 immunoreactivity. The immunoreactive cells in the perinidal parenchyma demonstrated obvious neuronal morphological characteristics.
The characteristics of pFADD-positive and cleaved caspase-8–positive areas were assessed using the image analysis system. The mean distance from the nidus adjacent to either area was not affected by preoperative hemorrhage. The neuronal densities of pFADD-positive and cleaved caspase-8–positive areas were analyzed using the same system. The density of the control area (samples that were pFADD-negative and cleaved caspase-8 negative) was significantly higher when compared with that of pFADD-positive and cleaved caspase-8–positive areas (p < 0.05). The expressions of cleaved caspase-9, cleaved poly(adenosine diphosphate–ribose) polymerase, and apoptotic cells were analyzed using the terminal deoxynucleotidyl transferase–mediated deoxyuridine triphosphate nick-end labeling method.
Conclusions
Neuronal areas that stained positively for pFADD and cleaved caspase-8 existed around the nidus of AVMs. In these areas, the neuronal density was lower than that in the other parenchyma around the AVM. Neuronal loss around the nidus may be the origin of brain dysfunction around AVMs.
Collapse
Affiliation(s)
- Yasushi Takagi
- Department of Neurosurgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | | | | | | | | |
Collapse
|
49
|
Pulliam L, Sun B, Rempel H, Martinez PM, Hoekman JD, Rao RJ, Frey WH, Hanson LR. Intranasal Tat Alters Gene Expression in the Mouse Brain. J Neuroimmune Pharmacol 2007; 2:87-92. [DOI: 10.1007/s11481-006-9053-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2006] [Accepted: 12/05/2006] [Indexed: 10/23/2022]
|
50
|
Shen Y, He P, Zhong Z, McAllister C, Lindholm K. Distinct destructive signal pathways of neuronal death in Alzheimer's disease. Trends Mol Med 2006; 12:574-9. [PMID: 17055782 DOI: 10.1016/j.molmed.2006.10.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2006] [Revised: 09/14/2006] [Accepted: 10/10/2006] [Indexed: 11/17/2022]
Abstract
Abundant neuron loss is a major feature of Alzheimer's disease (AD). Hypotheses for this loss include abnormal amyloid precursor protein processing (i.e. excess Abeta production, protein aggregation or misfolding), oxidative stress, excitotoxicity and inflammation. Neuron loss is a major cause of dementia in AD; however, it seems that there is no definitive pathway that causes cell death in the AD brain. Here, we examine the hypotheses for neuron loss in AD and pose the argument that the means by which neurons degenerate is irrelevant for cognitive decline. The best treatment for cognitive decline is to prevent the toxicity that first sets the neuron on its path to destruction, which is the production of Abeta peptide.
Collapse
Affiliation(s)
- Yong Shen
- Haldeman Laboratory of Molecular and Cellular Neurobiology, Sun Health Research Institute 3501, West Santa Fe Drive, Sun City, AZ 85351, USA.
| | | | | | | | | |
Collapse
|