1
|
Feng G, Zhong M, Huang H, Zhao P, Zhang X, Wang T, Gao H, Xu H. Identification of UBE2N as a biomarker of Alzheimer's disease by combining WGCNA with machine learning algorithms. Sci Rep 2025; 15:6479. [PMID: 39987324 PMCID: PMC11847011 DOI: 10.1038/s41598-025-90578-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 02/13/2025] [Indexed: 02/24/2025] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia, emphasizing the critical need for the development of biomarkers that facilitate accurate and objective assessment of disease progression for early detection and intervention to delay its onset. In our study, three AD datasets from the Gene Expression Omnibus (GEO) database were integrated for differential expression analysis, followed by a weighted gene co-expression network analysis (WGCNA), and potential AD biomarkers were screened. Our study identified UBE2N as a promising biomarker for AD. Functional enrichment analysis revealed that UBE2N is associated with synaptic vesicle cycling and T cell/B cell receptor signaling pathways. Notably, UBE2N expression levels were found to be significantly reduced in the cortex and hippocampus of the TauP301S mice. Furthermore, analysis of single-cell data from AD patients demonstrated the association of UBE2N and T cell function. These findings underscore the potential of UBE2N as a valuable biomarker for AD, offering important insights for diagnosis and targeted therapeutic strategies.
Collapse
Affiliation(s)
- Gangyi Feng
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Manli Zhong
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Hudie Huang
- Department of Anatomy, Histology and Embryology, School of Medicine, Shenzhen University, Shenzhen, China
| | - Pu Zhao
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Xiaoyu Zhang
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Tao Wang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Huiling Gao
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, China.
| | - He Xu
- Department of Anatomy, Histology and Embryology, School of Medicine, Shenzhen University, Shenzhen, China.
| |
Collapse
|
2
|
Li X, Gao Y, Li B, Zhao W, Cai Q, Yin W, Zeng S, Li X, Gao H, Cheng M. Integrated proteomics and metabolomics analysis of D-pinitol function during hippocampal damage in streptozocin-induced aging-accelerated mice. Front Mol Neurosci 2023; 16:1251513. [PMID: 38025258 PMCID: PMC10664147 DOI: 10.3389/fnmol.2023.1251513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 10/09/2023] [Indexed: 12/01/2023] Open
Abstract
Purpose Diabetes can cause hippocampal damage and lead to cognitive impairment. Diabetic cognitive impairment (DCI) is a chronic complication of diabetes associated with a high disability rate; however, its pathogenesis and therapeutic targets are unclear. We aimed to explore the mechanism of hippocampal damage during diabetes and evaluate the potential role of D-pinitol (DP) in protecting hippocampal tissue and improving cognitive dysfunction. Methods DP (150 mg/kg/day) was administered intragastrically to streptozocin-induced aging-accelerated mice for 8 weeks. Hippocampal tissues were examined using tandem mass tag (TMT)-based proteomics and liquid chromatography-mass spectrometry (LC-MS)/MS-based non-targeted metabolomic analysis. Differentially expressed proteins (DEPs) and differentially regulated metabolites (DRMs) were screened for further analysis, and some DEPs were verified using western blotting. Results Our results showed that 329 proteins had significantly altered hippocampal expression in untreated diabetic mice (DM), which was restored to normal after DP treatment in 72 cases. In total, 207 DRMs were identified in the DM group, and the expression of 32 DRMs was restored to normal post-DP treatment. These proteins and metabolites are involved in metabolic pathways (purine metabolism, arginine and proline metabolism, and histidine metabolism), actin cytoskeleton regulation, oxidative phosphorylation, and Rap1-mediated signaling. Conclusions Our study may help to better understand the mechanism of diabetic hippocampal damage and cognitive impairment and suggest a potential therapeutic target.
Collapse
Affiliation(s)
- Xiaoxia Li
- Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Jinan Clinical Research Center for Geriatric Medicine, Jinan, China
- Department of Diabetes, The Third People's Hospital of Gansu Province, Lanzhou, China
| | - Yuan Gao
- Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Jinan Clinical Research Center for Geriatric Medicine, Jinan, China
| | - Baoying Li
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Health Management Center (East Area), Qilu Hospital of Shandong University, Jinan, China
| | - Wenqian Zhao
- Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Jinan Clinical Research Center for Geriatric Medicine, Jinan, China
| | - Qian Cai
- Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Jinan Clinical Research Center for Geriatric Medicine, Jinan, China
| | - Wenbin Yin
- Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Jinan Clinical Research Center for Geriatric Medicine, Jinan, China
| | - Shudong Zeng
- Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Jinan Clinical Research Center for Geriatric Medicine, Jinan, China
| | - Xiaoli Li
- Department of Pharmacy, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Haiqing Gao
- Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Jinan Clinical Research Center for Geriatric Medicine, Jinan, China
| | - Mei Cheng
- Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Jinan Clinical Research Center for Geriatric Medicine, Jinan, China
| |
Collapse
|
3
|
Uras I, Karayel-Basar M, Sahin B, Baykal AT. Detection of early proteomic alterations in 5xFAD Alzheimer's disease neonatal mouse model via MALDI-MSI. Alzheimers Dement 2023; 19:4572-4589. [PMID: 36934297 DOI: 10.1002/alz.13008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 03/20/2023]
Abstract
Alzheimer's disease (AD) is a debilitating neurodegenerative disorder, characterized by memory deficit and dementia. AD is considered a multifactorial disorder where multiple processes like amyloid-beta and tau accumulation, axonal degeneration, synaptic plasticity, and autophagic processes plays an important role. In this study, the spatial proteomic differences in the neonatal 5xFAD brain tissue were investigated using MALDI-MSI coupled to LC-MS/MS, and the statistically significantly altered proteins were associated with AD. Thirty-five differentially expressed proteins (DEPs) between the brain tissues of neonatal 5xFAD and their littermate mice were detected via MALDI-MSI technique. Among the 35 proteins identified, 26 of them were directly associated with AD. Our results indicated a remarkable resemblance in the protein expression profiles of neonatal 5xFAD brain when compared to AD patient specimens or AD mouse models. These findings showed that the molecular alterations in the AD brain existed even at birth and that some proteins are neurodegenerative presages in neonatal AD brain. HIGHLIGHTS: Spatial proteomic alterations in the 5xFAD mouse brain compared to the littermate. 26 out of 35 differentially expressed proteins associated with Alzheimer's disease (AD). Molecular alterations and neurodegenerative presages in neonatal AD brain. Alterations in the synaptic function an early and common neurobiological thread.
Collapse
Affiliation(s)
- Irep Uras
- Department of Biochemistry and Molecular Biology, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Merve Karayel-Basar
- Department of Biochemistry and Molecular Biology, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Betul Sahin
- Acibadem Labmed Clinical Laboratories, Istanbul, Turkey
| | - Ahmet Tarik Baykal
- Department of Medical Biochemistry, Faculty of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| |
Collapse
|
4
|
Vrettou S, Wirth B. S-Glutathionylation and S-Nitrosylation in Mitochondria: Focus on Homeostasis and Neurodegenerative Diseases. Int J Mol Sci 2022; 23:15849. [PMID: 36555492 PMCID: PMC9779533 DOI: 10.3390/ijms232415849] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/24/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022] Open
Abstract
Redox post-translational modifications are derived from fluctuations in the redox potential and modulate protein function, localization, activity and structure. Amongst the oxidative reversible modifications, the S-glutathionylation of proteins was the first to be characterized as a post-translational modification, which primarily protects proteins from irreversible oxidation. However, a growing body of evidence suggests that S-glutathionylation plays a key role in core cell processes, particularly in mitochondria, which are the main source of reactive oxygen species. S-nitrosylation, another post-translational modification, was identified >150 years ago, but it was re-introduced as a prototype cell-signaling mechanism only recently, one that tightly regulates core processes within the cell’s sub-compartments, especially in mitochondria. S-glutathionylation and S-nitrosylation are modulated by fluctuations in reactive oxygen and nitrogen species and, in turn, orchestrate mitochondrial bioenergetics machinery, morphology, nutrients metabolism and apoptosis. In many neurodegenerative disorders, mitochondria dysfunction and oxidative/nitrosative stresses trigger or exacerbate their pathologies. Despite the substantial amount of research for most of these disorders, there are no successful treatments, while antioxidant supplementation failed in the majority of clinical trials. Herein, we discuss how S-glutathionylation and S-nitrosylation interfere in mitochondrial homeostasis and how the deregulation of these modifications is associated with Alzheimer’s, Parkinson’s, amyotrophic lateral sclerosis and Friedreich’s ataxia.
Collapse
Affiliation(s)
- Sofia Vrettou
- Institute of Human Genetics, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
| | - Brunhilde Wirth
- Institute of Human Genetics, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany
- Center for Rare Diseases, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
| |
Collapse
|
5
|
Shi W, Hu R, Zhao R, Zhu J, Shen H, Li H, Wang L, Yang Z, Jiang Q, Qiao Y, Jiang G, Cheng J, Wan X. Transcriptome analysis of hepatopancreas and gills of Palaemon gravieri under salinity stress. Gene 2022; 851:147013. [DOI: 10.1016/j.gene.2022.147013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 09/01/2022] [Accepted: 10/25/2022] [Indexed: 11/04/2022]
|
6
|
Tsumagari K, Sato Y, Shimozawa A, Aoyagi H, Okano H, Kuromitsu J. Co-expression network analysis of human tau-transgenic mice reveals protein modules associated with tau-induced pathologies. iScience 2022; 25:104832. [PMID: 35992067 PMCID: PMC9382322 DOI: 10.1016/j.isci.2022.104832] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 06/03/2022] [Accepted: 07/20/2022] [Indexed: 12/04/2022] Open
Abstract
Abnormally accumulated tau protein aggregates are one of the hallmarks of neurodegenerative diseases, including Alzheimer's disease (AD). In order to investigate proteomic alteration driven by tau aggregates, we implemented quantitative proteomics to analyze disease model mice expressing human MAPT P301S transgene (hTau-Tg) and quantified more than 9,000 proteins in total. We applied the weighted gene co-expression analysis (WGCNA) algorithm to the datasets and explored protein co-expression modules that were associated with the accumulation of tau aggregates and were preserved in proteomes of AD brains. This led us to identify four modules with functions related to neuroinflammatory responses, mitochondrial energy production processes (including the tricarboxylic acid cycle and oxidative phosphorylation), cholesterol biosynthesis, and postsynaptic density. Furthermore, a phosphoproteomics study uncovered phosphorylation sites that were highly correlated with these modules. Our datasets represent resources for understanding the molecular basis of tau-induced neurodegeneration, including AD.
Collapse
Affiliation(s)
- Kazuya Tsumagari
- Center for Integrated Medical Research, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- RIKEN Center for Integrative Medical Sciences, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Yoshiaki Sato
- Eisai-Keio Innovation Laboratory for Dementia, hhc Data Creation Center, Eisai Co., Ltd., Shinjuku-ku, Tokyo 160-8582, Japan
| | - Aki Shimozawa
- Center for Integrated Medical Research, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hirofumi Aoyagi
- Eisai-Keio Innovation Laboratory for Dementia, hhc Data Creation Center, Eisai Co., Ltd., Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Junro Kuromitsu
- Eisai-Keio Innovation Laboratory for Dementia, hhc Data Creation Center, Eisai Co., Ltd., Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
7
|
Schwab K, Melis V, Harrington CR, Wischik CM, Magbagbeolu M, Theuring F, Riedel G. Proteomic Analysis of Hydromethylthionine in the Line 66 Model of Frontotemporal Dementia Demonstrates Actions on Tau-Dependent and Tau-Independent Networks. Cells 2021; 10:2162. [PMID: 34440931 PMCID: PMC8391171 DOI: 10.3390/cells10082162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 08/16/2021] [Accepted: 08/19/2021] [Indexed: 12/21/2022] Open
Abstract
Abnormal aggregation of tau is the pathological hallmark of tauopathies including frontotemporal dementia (FTD). We have generated tau-transgenic mice that express the aggregation-prone P301S human tau (line 66). These mice present with early-onset, high tau load in brain and FTD-like behavioural deficiencies. Several of these behavioural phenotypes and tau pathology are reversed by treatment with hydromethylthionine but key pathways underlying these corrections remain elusive. In two proteomic experiments, line 66 mice were compared with wild-type mice and then vehicle and hydromethylthionine treatments of line 66 mice were compared. The brain proteome was investigated using two-dimensional electrophoresis and mass spectrometry to identify protein networks and pathways that were altered due to tau overexpression or modified by hydromethylthionine treatment. Overexpression of mutant tau induced metabolic/mitochondrial dysfunction, changes in synaptic transmission and in stress responses, and these functions were recovered by hydromethylthionine. Other pathways, such as NRF2, oxidative phosphorylation and protein ubiquitination were activated by hydromethylthionine, presumably independent of its function as a tau aggregation inhibitor. Our results suggest that hydromethylthionine recovers cellular activity in both a tau-dependent and a tau-independent fashion that could lead to a wide-spread improvement of homeostatic function in the FTD brain.
Collapse
Affiliation(s)
- Karima Schwab
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK; (K.S.); (V.M.); (C.R.H.); (C.M.W.)
- Charité—Universitätsmedizin Berlin, Hessische Str. 3-4, 10115 Berlin, Germany; (M.M.); (F.T.)
| | - Valeria Melis
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK; (K.S.); (V.M.); (C.R.H.); (C.M.W.)
| | - Charles R. Harrington
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK; (K.S.); (V.M.); (C.R.H.); (C.M.W.)
- TauRx Therapeutics Ltd., 395 King Street, Aberdeen AB24 5RP, UK
| | - Claude M. Wischik
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK; (K.S.); (V.M.); (C.R.H.); (C.M.W.)
- TauRx Therapeutics Ltd., 395 King Street, Aberdeen AB24 5RP, UK
| | - Mandy Magbagbeolu
- Charité—Universitätsmedizin Berlin, Hessische Str. 3-4, 10115 Berlin, Germany; (M.M.); (F.T.)
| | - Franz Theuring
- Charité—Universitätsmedizin Berlin, Hessische Str. 3-4, 10115 Berlin, Germany; (M.M.); (F.T.)
| | - Gernot Riedel
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK; (K.S.); (V.M.); (C.R.H.); (C.M.W.)
| |
Collapse
|
8
|
Santos-Pereira C, Rodrigues LR, Côrte-Real M. Emerging insights on the role of V-ATPase in human diseases: Therapeutic challenges and opportunities. Med Res Rev 2021; 41:1927-1964. [PMID: 33483985 DOI: 10.1002/med.21782] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/05/2020] [Accepted: 01/05/2021] [Indexed: 12/13/2022]
Abstract
The control of the intracellular pH is vital for the survival of all organisms. Membrane transporters, both at the plasma and intracellular membranes, are key players in maintaining a finely tuned pH balance between intra- and extracellular spaces, and therefore in cellular homeostasis. V-ATPase is a housekeeping ATP-driven proton pump highly conserved among prokaryotes and eukaryotes. This proton pump, which exhibits a complex multisubunit structure based on cell type-specific isoforms, is essential for pH regulation and for a multitude of ubiquitous and specialized functions. Thus, it is not surprising that V-ATPase aberrant overexpression, mislocalization, and mutations in V-ATPase subunit-encoding genes have been associated with several human diseases. However, the ubiquitous expression of this transporter and the high toxicity driven by its off-target inhibition, renders V-ATPase-directed therapies very challenging and increases the need for selective strategies. Here we review emerging evidence linking V-ATPase and both inherited and acquired human diseases, explore the therapeutic challenges and opportunities envisaged from recent data, and advance future research avenues. We highlight the importance of V-ATPases with unique subunit isoform molecular signatures and disease-associated isoforms to design selective V-ATPase-directed therapies. We also discuss the rational design of drug development pipelines and cutting-edge methodological approaches toward V-ATPase-centered drug discovery. Diseases like cancer, osteoporosis, and even fungal infections can benefit from V-ATPase-directed therapies.
Collapse
Affiliation(s)
- Cátia Santos-Pereira
- Department of Biology, Centre of Molecular and Environmental Biology (CBMA), University of Minho, Braga, Portugal.,Department of Biological Engineering, Centre of Biological Engineering (CEB), University of Minho, Braga, Portugal
| | - Lígia R Rodrigues
- Department of Biological Engineering, Centre of Biological Engineering (CEB), University of Minho, Braga, Portugal
| | - Manuela Côrte-Real
- Department of Biology, Centre of Molecular and Environmental Biology (CBMA), University of Minho, Braga, Portugal
| |
Collapse
|
9
|
Gao Y, Zhao H, Xu T, Tian J, Qin X. Identification of Crucial Genes and Diagnostic Value Analysis in Major Depressive Disorder Using Bioinformatics Analysis. Comb Chem High Throughput Screen 2020; 25:13-20. [PMID: 33238838 DOI: 10.2174/1386207323999201124204413] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/27/2020] [Accepted: 11/01/2020] [Indexed: 11/22/2022]
Abstract
AIM AND OBJECTIVE Despite the prevalence and burden of major depressive disorder (MDD), our current understanding of the pathophysiology is still incomplete. Therefore, this paper aims to explore genes and evaluate their diagnostic ability in the pathogenesis of MDD. METHODS Firstly, the expression profiles of mRNA and microRNA were downloaded from the gene expression database and analyzed by the GEO2R online tool to identify differentially expressed genes (DEGs) and differentially expressed microRNAs (DEMs). Then, the DAVID tool was used for functional enrichment analysis. Secondly, the comprehensive protein- protein interaction (PPI) network was analyzed using Cytoscape, and the network MCODE was applied to explore hub genes. Thirdly, the receiver operating characteristic (ROC) curve of the core gene was drawn to evaluate clinical diagnostic ability. Finally, mirecords was used to predict the target genes of DEMs. RESULTS A total of 154 genes were identified as DEGs, and 14 microRNAs were identified as DEMs. Pathway enrichment analysis showed that DEGs were mainly involved in hematopoietic cell lineage, PI3K-Akt signaling pathway, cytokinecytokine receptor interaction, chemokine signaling pathway, and JAK-STAT signaling pathway. Three important modules are identified and selected by the MCODE clustering algorithm. The top 12 hub genes including CXCL16, CXCL1, GNB5, GNB4, OPRL1, SSTR2, IL7R, MYB, CSF1R, GSTM1, GSTM2, and GSTP1 were identified as important genes for subsequent analysis. Among these important hub genes, GSTM2, GNB4, GSTP1 and CXCL1 have good diagnostic ability. Finally, by combining these four genes, the diagnostic ability of MDD can be improved to 0.905, which is of great significance for the clinical diagnosis of MDD. CONCLUSION Our results indicate that GSTM2, GNB4, GSTP1 and CXCL1 have potential diagnostic markers and are of great significance in clinical research and diagnostic application of MDD. This result needs a large sample study to further confirm the pathogenesis of MDD.
Collapse
Affiliation(s)
- Yao Gao
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan,. China
| | - Huiliang Zhao
- Shanxi Key Laboratory of Active Constituents Research and Utilization of TCM, Shanxi University, Taiyuan,. China
| | - Teng Xu
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan,. China
| | - Junsheng Tian
- Shanxi Key Laboratory of Active Constituents Research and Utilization of TCM, Shanxi University, Taiyuan,. China
| | - Xuemei Qin
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan,. China
| |
Collapse
|
10
|
Jiang S, Bhaskar K. Degradation and Transmission of Tau by Autophagic-Endolysosomal Networks and Potential Therapeutic Targets for Tauopathy. Front Mol Neurosci 2020; 13:586731. [PMID: 33177989 PMCID: PMC7596180 DOI: 10.3389/fnmol.2020.586731] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 09/24/2020] [Indexed: 01/21/2023] Open
Abstract
Tauopathies are a class of neurodegenerative diseases, including Alzheimer’s disease (AD), Frontotemporal Dementia (FTD), Progressive Supranuclear Palsy (PSP), Corticobasal Degeneration (CBD), and many others where microtubule-associated protein tau (MAPT or tau) is hyperphosphorylated and aggregated to form insoluble paired helical filaments (PHFs) and ultimately neurofibrillary tangles (NFTs). Autophagic-endolysosomal networks (AELN) play important roles in tau clearance. Excessive soluble neurotoxic forms of tau and tau hyperphosphorylated at specific sites are cleared through the ubiquitin-proteasome system (UPS), Chaperon-mediated Autophagy (CMA), and endosomal microautophagy (e-MI). On the other hand, intra-neuronal insoluble tau aggregates are often degraded within lysosomes by macroautophagy. AELN defects have been observed in AD, FTD, CBD, and PSP, and lysosomal dysfunction was shown to promote the cleavage and neurotoxicity of tau. Moreover, several AD risk genes (e.g., PICALM, GRN, and BIN1) have been associated with dysregulation of AELN in the late-onset sporadic AD. Conversely, tau dissociation from microtubules interferes with retrograde transport of autophagosomes to lysosomes, and that tau fragments can also lead to lysosomal dysfunction. Recent studies suggest that tau is not merely an intra-neuronal protein, but it can be released to brain parenchyma via extracellular vesicles, like exosomes and ectosomes, and thus spread between neurons. Extracellular tau can also be taken up by microglial cells and astrocytes, either being degraded through AELN or propagated via exosomes. This article reviews the complex roles of AELN in the degradation and transmission of tau, potential diagnostic/therapeutic targets and strategies based on AELN-mediated tau clearance and propagation, and the current state of drug development targeting AELN and tau against tauopathies.
Collapse
Affiliation(s)
- Shanya Jiang
- Department of Molecular Genetics and Microbiology, The University of New Mexico, Albuquerque, NM, United States
| | - Kiran Bhaskar
- Department of Molecular Genetics and Microbiology, The University of New Mexico, Albuquerque, NM, United States
| |
Collapse
|
11
|
Ojo JO, Crynen G, Algamal M, Vallabhaneni P, Leary P, Mouzon B, Reed JM, Mullan M, Crawford F. Unbiased Proteomic Approach Identifies Pathobiological Profiles in the Brains of Preclinical Models of Repetitive Mild Traumatic Brain Injury, Tauopathy, and Amyloidosis. ASN Neuro 2020; 12:1759091420914768. [PMID: 32241177 PMCID: PMC7132820 DOI: 10.1177/1759091420914768] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
No concerted investigation has been conducted to explore overlapping and distinct
pathobiological mechanisms between repetitive mild traumatic brain injury
(r-mTBI) and tau/amyloid proteinopathies considering the long history of
association between TBI and Alzheimer’s disease. We address this problem by
using unbiased proteomic approaches to generate detailed time-dependent brain
molecular profiles of response to repetitive mTBI in C57BL/6 mice and in mouse
models of amyloidosis (with amyloid precursor protein KM670/671NL (Swedish) and
Presenilin 1 M146L mutations [PSAPP]) and tauopathy (hTau). Brain tissues from
animals were collected at different timepoints after injuries (24 hr–12 months
post-injury) and at different ages for tau or amyloid transgenic models (3, 9,
and 15 months old), encompassing the pre-, peri-, and post-“onset” of cognitive
and pathological phenotypes. We identified 30 hippocampal and 47 cortical
proteins that were significantly modulated over time in the r-mTBI compared with
sham mice. These proteins identified TBI-dependent modulation of
phosphatidylinositol-3-kinase/AKT signaling, protein kinase A signaling, and
PPARα/RXRα activation in the hippocampus and protein kinase A signaling,
gonadotropin-releasing hormone signaling, and B cell receptor signaling in the
cortex. Previously published neuropathological studies of our mTBI model showed
a lack of amyloid and tau pathology. In PSAPP mice, we identified 19 proteins
significantly changing in the cortex and only 7 proteins in hTau mice versus
wild-type littermates. When we explored the overlap between our r-mTBI model and
the PSAPP/hTau models, a fairly small coincidental change was observed involving
only eight significantly regulated proteins. This work suggests a very distinct
TBI neurodegeneration and also that other factors are needed to drive
pathologies such as amyloidosis and tauopathy postinjury.
Collapse
Affiliation(s)
- Joseph O Ojo
- Experimental Neuropathology and Proteomic Laboratory, Roskamp Institute, Sarasota, Florida, United States.,James A. Haley Veterans' Hospital, Tampa, Florida, United States.,School of Life, Health and Chemical Sciences, The Open University, Milton Keynes, United Kingdom
| | - Gogce Crynen
- Experimental Neuropathology and Proteomic Laboratory, Roskamp Institute, Sarasota, Florida, United States.,School of Life, Health and Chemical Sciences, The Open University, Milton Keynes, United Kingdom
| | - Moustafa Algamal
- Experimental Neuropathology and Proteomic Laboratory, Roskamp Institute, Sarasota, Florida, United States.,School of Life, Health and Chemical Sciences, The Open University, Milton Keynes, United Kingdom
| | - Prashanti Vallabhaneni
- Experimental Neuropathology and Proteomic Laboratory, Roskamp Institute, Sarasota, Florida, United States
| | - Paige Leary
- Experimental Neuropathology and Proteomic Laboratory, Roskamp Institute, Sarasota, Florida, United States
| | - Benoit Mouzon
- Experimental Neuropathology and Proteomic Laboratory, Roskamp Institute, Sarasota, Florida, United States.,James A. Haley Veterans' Hospital, Tampa, Florida, United States.,School of Life, Health and Chemical Sciences, The Open University, Milton Keynes, United Kingdom
| | - Jon M Reed
- Experimental Neuropathology and Proteomic Laboratory, Roskamp Institute, Sarasota, Florida, United States.,Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut, United States
| | - Michael Mullan
- Experimental Neuropathology and Proteomic Laboratory, Roskamp Institute, Sarasota, Florida, United States.,School of Life, Health and Chemical Sciences, The Open University, Milton Keynes, United Kingdom
| | - Fiona Crawford
- Experimental Neuropathology and Proteomic Laboratory, Roskamp Institute, Sarasota, Florida, United States.,James A. Haley Veterans' Hospital, Tampa, Florida, United States.,School of Life, Health and Chemical Sciences, The Open University, Milton Keynes, United Kingdom
| |
Collapse
|
12
|
Sullivan SE, Liao M, Smith RV, White C, Lagomarsino VN, Xu J, Taga M, Bennett DA, De Jager PL, Young-Pearse TL. Candidate-based screening via gene modulation in human neurons and astrocytes implicates FERMT2 in Aβ and TAU proteostasis. Hum Mol Genet 2020; 28:718-735. [PMID: 30371777 DOI: 10.1093/hmg/ddy376] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 10/15/2018] [Accepted: 10/16/2018] [Indexed: 11/13/2022] Open
Abstract
Large-scale 'omic' studies investigating the pathophysiological processes that lead to Alzheimer's disease (AD) dementia have identified an increasing number of susceptibility genes, many of which are poorly characterized and have not previously been implicated in AD. Here, we evaluated the utility of human induced pluripotent stem cell-derived neurons and astrocytes as tools to systematically test AD-relevant cellular phenotypes following perturbation of candidate genes identified by genome-wide studies. Lentiviral-mediated delivery of shRNAs was used to modulate expression of 66 genes in astrocytes and 52 genes in induced neurons. Five genes (CNN2, GBA, GSTP1, MINT2 and FERMT2) in neurons and nine genes (CNN2, ITGB1, MINT2, SORL1, VLDLR, NPC1, NPC2, PSAP and SCARB2) in astrocytes significantly altered extracellular amyloid-β (Aβ) levels. Knockdown of AP3M2, CNN2, GSTP1, NPC1, NPC2, PSAP and SORL1 reduced interleukin-6 levels in astrocytes. Only knockdown of FERMT2 led to a reduction in the proportion of TAU that is phosphorylated. Further, CRISPR-Cas9 targeting of FERMT2 in both familial AD (fAD) and fAD-corrected human neurons validated the findings of reduced extracellular Aβ. Interestingly, FERMT2 reduction had no effect on the Aβ42:40 ratio in corrected neurons and a reduction of phospho-tau, but resulted in an elevation in Aβ42:40 ratio and no reduction in phospho-tau in fAD neurons. Taken together, this study has prioritized 15 genes as being involved in contributing to Aβ accumulation, phosphorylation of tau and/or cytokine secretion, and, as illustrated with FERMT2, it sets the stage for further cell-type-specific dissection of the role of these genes in AD.
Collapse
Affiliation(s)
- Sarah E Sullivan
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Meichen Liao
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Robert V Smith
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Charles White
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Valentina N Lagomarsino
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jishu Xu
- Cell Circuits Program, Broad Institute, Cambridge, MA, USA
| | - Mariko Taga
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Philip L De Jager
- Cell Circuits Program, Broad Institute, Cambridge, MA, USA.,Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Tracy L Young-Pearse
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
13
|
Pires G, McElligott S, Drusinsky S, Halliday G, Potier MC, Wisniewski T, Drummond E. Secernin-1 is a novel phosphorylated tau binding protein that accumulates in Alzheimer's disease and not in other tauopathies. Acta Neuropathol Commun 2019; 7:195. [PMID: 31796108 PMCID: PMC6892024 DOI: 10.1186/s40478-019-0848-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 11/14/2019] [Indexed: 12/13/2022] Open
Abstract
We recently identified Secernin-1 (SCRN1) as a novel amyloid plaque associated protein using localized proteomics. Immunohistochemistry studies confirmed that SCRN1 was present in plaque-associated dystrophic neurites and also revealed distinct and abundant co-localization with neurofibrillary tangles (NFTs). Little is known about the physiological function of SCRN1 and its role in Alzheimer's disease (AD) and other neurodegenerative diseases has not been studied. Therefore, we performed a comprehensive study of SCRN1 distribution in neurodegenerative diseases. Immunohistochemistry was used to map SCRN1 accumulation throughout the progression of AD in a cohort of 58 patients with a range of NFT pathology (Abundant NFT, n = 21; Moderate NFT, n = 22; Low/No NFT, n = 15), who were clinically diagnosed as having AD, mild cognitive impairment or normal cognition. SCRN1 accumulation was also examined in two cases with both Frontotemporal Lobar Degeneration (FTLD)-Tau and AD-related neuropathology, cases of Down Syndrome (DS) with AD (n = 5), one case of hereditary cerebral hemorrhage with amyloidosis - Dutch type (HCHWA-D) and other non-AD tauopathies including: primary age-related tauopathy (PART, [n = 5]), Corticobasal Degeneration (CBD, [n = 5]), Progressive Supranuclear Palsy (PSP, [n = 5]) and Pick's disease (PiD, [n = 4]). Immunohistochemistry showed that SCRN1 was a neuronal protein that abundantly accumulated in NFTs and plaque-associated dystrophic neurites throughout the progression of AD. Quantification of SCRN1 immunohistochemistry confirmed that SCRN1 preferentially accumulated in NFTs in comparison to surrounding non-tangle containing neurons at both early and late stages of AD. Similar results were observed in DS with AD and PART. However, SCRN1 did not co-localize with phosphorylated tau inclusions in CBD, PSP or PiD. Co-immunoprecipitation revealed that SCRN1 interacted with phosphorylated tau in human AD brain tissue. Together, these results suggest that SCRN1 is uniquely associated with tau pathology in AD, DS and PART. As such, SCRN1 has potential as a novel therapeutic target and could serve as a useful biomarker to distinguish AD from other tauopathies.
Collapse
Affiliation(s)
- Geoffrey Pires
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, 435 East 30th Street, Rm 1017, New York, NY, 10016, USA
| | - Sacha McElligott
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, 435 East 30th Street, Rm 1017, New York, NY, 10016, USA
| | - Shiron Drusinsky
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, 435 East 30th Street, Rm 1017, New York, NY, 10016, USA
| | - Glenda Halliday
- Brain & Mind Centre and Central Clinical School, Faculty of Medicine and Health, University of Sydney, 94 Mallett Street, Camperdown, NSW, 2050, Australia
| | - Marie-Claude Potier
- Institut du Cerveau et de la Moelle épinière, CNRS UMR7225, INSERM U1127, UPMC, Hôpital de la Pitié-Salpêtrière, 47 Bd de l'Hôpital, Paris, France
| | - Thomas Wisniewski
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, 435 East 30th Street, Rm 1017, New York, NY, 10016, USA.
- Departments of Pathology and Psychiatry, New York University School of Medicine, 435 East 30th Street, Rm 1017, New York, NY, 10016, USA.
| | - Eleanor Drummond
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, 435 East 30th Street, Rm 1017, New York, NY, 10016, USA.
- Brain & Mind Centre and Central Clinical School, Faculty of Medicine and Health, University of Sydney, 94 Mallett Street, Camperdown, NSW, 2050, Australia.
| |
Collapse
|
14
|
Liu J, Li Y. Upregulation of MAPK10, TUBB2B and RASL11B may contribute to the development of neuroblastoma. Mol Med Rep 2019; 20:3475-3486. [PMID: 31432180 PMCID: PMC6755162 DOI: 10.3892/mmr.2019.10589] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 11/07/2018] [Indexed: 01/01/2023] Open
Abstract
The present study aimed to investigate genes and transcription factors (TFs) that may contribute to neuroblastoma (NB) development. The GSE78061 dataset that included 25 human NB cell lines and four retinal pigment epithelial cell lines was used to analyze differentially expressed genes (DEGs) between groups. Functional enrichment analysis and protein-protein interaction (PPI) network analysis were performed for the identified DEGs. Additionally, submodule analysis and TF-target regulatory networks were conducted. The relative mRNA expression levels of mitogen-activated protein kinase 10 (MAPK10), tubulin β 2B class IIb (TUBB2B), RAS like family 11 member B (RASL11B) and integrin subunit α 2 (ITGA2) in the NB cell line SH-SY5Y were compared with retinal pigment epithelial cell lines. A set of 386 upregulated and 542 downregulated DEGs were obtained. Upregulated DEGs were significantly associated with the ‘neuron migration’ and ‘dopaminergic synapse signaling’ pathways, whereas, downregulated DEGs were primarily involved in ‘focal adhesion’ such as ITGA2 and ITGA3. In the PPI networks analyzed, MAPK10, dopa decarboxylase (DDC), G protein subunit γ 2 (GNG2), paired like homeobox 2B (PHOX2B), TUBB2B, RASL11B, and ITGA2 were hub genes with high connectivity degrees. Additionally, PHOX2B was predicted to be a TF regulating TUBB2B in the regulatory network. The expressions of MAPK10, TUBB2B, RASL11B and ITGA2 were detected by reverse transcription-quantitative polymerase chain reaction in the NB cell line SH-SY5Y, and were consistent with the present bioinformatics results, suggesting that MAPK10, DDC, GNG2, PHOX2B, TUBB2B, RASL11B, ITGA2 and ITGA3 may contribute to NB development. Additionally, the present study identified a novel significant association between the increased expression levels of MAPK10, TUBB2B and RASL11B, and NB cells.
Collapse
Affiliation(s)
- Jiangtao Liu
- The Key Laboratory of Pathobiology, Ministry of Education, Department of Pathobiology, College of Basic Medical Sciences, Jilin University, Chuangchun, Jilin 130000, P.R. China
| | - Yulin Li
- The Key Laboratory of Pathobiology, Ministry of Education, Department of Pathobiology, College of Basic Medical Sciences, Jilin University, Chuangchun, Jilin 130000, P.R. China
| |
Collapse
|
15
|
Mazi AR, Arzuman AS, Gurel B, Sahin B, Tuzuner MB, Ozansoy M, Baykal AT. Neonatal Neurodegeneration in Alzheimer's Disease Transgenic Mouse Model. J Alzheimers Dis Rep 2018; 2:79-91. [PMID: 30480251 PMCID: PMC6159732 DOI: 10.3233/adr-170049] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive disorder characterized by a variety of molecular pathologies causing cortical dementia with a prominent memory deficit. Formation of the pathology, which begins decades before the diagnosis of the disease, is highly correlated with the clinical symptoms. Several proteomics studies were performed using animal models to monitor the alterations of the brain tissue proteome at different stages of AD. However, proteome changes in the brain regions of newborn transgenic mouse model have not been investigated yet. To this end, we analyzed protein expression alterations in cortex, hippocampus and cerebellum of transgenic mice carrying five familial AD mutations (5XFAD) at neonatal day-1. Our results indicate a remarkable difference in protein expression profile of newborn 5XFAD brain with region specific variations. Additionally, the proteins, which show similar expression alteration pattern in postmortem human AD brains, were determined. Bioinformatics analysis showed that the molecular alterations were mostly related to the cell morphology, cellular assembly and organization, and neuroinflammation. Moreover, morphological analysis revealed that there is an increase in neurite number of 5XFAD mouse neurons in vitro. We suggest that, molecular alterations in the AD brain exist even at birth, and perhaps the disease is silenced until older ages when the brain becomes vulnerable.
Collapse
Affiliation(s)
- Aise Rumeysa Mazi
- Regenerative and Restorative Medicine Research Center, REMER, Istanbul, Turkey.,Institute of Health Science, Istanbul Medipol University, Istanbul, Turkey.,Department of Medical Biochemistry, Acibadem Mehmet Ali Aydinlar University, School of Medicine, Istanbul, Turkey
| | - Aysegul Sumeyye Arzuman
- Regenerative and Restorative Medicine Research Center, REMER, Istanbul, Turkey.,Institute of Health Science, Istanbul Medipol University, Istanbul, Turkey.,Department of Medical Biochemistry, Acibadem Mehmet Ali Aydinlar University, School of Medicine, Istanbul, Turkey
| | - Busra Gurel
- Regenerative and Restorative Medicine Research Center, REMER, Istanbul, Turkey.,Institute of Health Science, Istanbul Medipol University, Istanbul, Turkey.,Department of Medical Biochemistry, Acibadem Mehmet Ali Aydinlar University, School of Medicine, Istanbul, Turkey
| | - Betul Sahin
- Acibadem Labmed R&D Laboratory, Istanbul, Turkey
| | | | - Mehmet Ozansoy
- Regenerative and Restorative Medicine Research Center, REMER, Istanbul, Turkey.,Department of Physiology, International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Ahmet Tarik Baykal
- Acibadem Labmed R&D Laboratory, Istanbul, Turkey.,Department of Medical Biochemistry, Acibadem Mehmet Ali Aydinlar University, School of Medicine, Istanbul, Turkey
| |
Collapse
|
16
|
Nie L, Wei G, Peng S, Qu Z, Yang Y, Yang Q, Huang X, Liu J, Zhuang Z, Yang X. Melatonin ameliorates anxiety and depression-like behaviors and modulates proteomic changes in triple transgenic mice of Alzheimer's disease. Biofactors 2017; 43:593-611. [PMID: 28608594 DOI: 10.1002/biof.1369] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 04/06/2017] [Accepted: 05/01/2017] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disease accompanied by neuropsychiatric symptoms, such as anxiety and depression. The levels of melatonin decrease in brains of AD patients. The potential effect of melatonin on anxiety and depression behaviors in AD and the underlying mechanisms remain unclear. In this study, we treated 10-month-old triple transgenic mice of AD (3xTg-AD) with melatonin (10 mg/kg body weight/day) for 1 month and explored the effects of melatonin on anxiety and depression-like behaviors in 3xTg-AD mice and the protein expression of hippocampal tissues. The behavioral test showed that melatonin ameliorated anxiety and depression-like behaviors of 3xTg-AD mice as measured by open field test, elevated plus maze test, forced swimming test, and tail suspension test. By carrying out two-dimensional fluorescence difference gel electrophoresis (2D-DIGE) coupled with mass spectrometry, we revealed a total of 46 differentially expressed proteins in hippocampus between the wild-type (WT) mice and non-treated 3xTg-AD mice. A total of 21 differentially expressed proteins were revealed in hippocampus between melatonin-treated and non-treated 3xTg-AD mice. Among these differentially expressed proteins, glutathione S-transferase P 1 (GSTP1) (an anxiety-associated protein) and complexin-1 (CPLX1) (a depression-associated protein) were significantly down-regulated in hippocampus of 3xTg-AD mice compared with the WT mice. The expression of these two proteins was modulated by melatonin treatment. Our study suggested that melatonin could be used as a potential candidate drug to improve the neuropsychiatric behaviors in AD via modulating the expression of the proteins (i.e. GSTP1 and CPLX1) involved in anxiety and depression behaviors. © 2017 BioFactors, 43(4):593-611, 2017.
Collapse
Affiliation(s)
- Lulin Nie
- College of Chemistry, Xiangtan University, Xiangtan, 411105, China
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China
| | - Gang Wei
- Thyroid & Breast Surgery Department, Hubei Maternal & Children Hospital, Wuhan, 430070, China
| | - Shengming Peng
- College of Chemistry, Xiangtan University, Xiangtan, 411105, China
| | - Zhongsen Qu
- Department of Neurology, Shanghai Jiaotong University Affiliated the Sixth Hospital, Shanghai, 200233, China
| | - Ying Yang
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430070, China
| | - Qian Yang
- Department of cell biology and Anatomy, LSU Health Sciences Center, New Orleans, LA, 70112
| | - Xinfeng Huang
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China
| | - Jianjun Liu
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China
| | - Zhixiong Zhuang
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China
| | - Xifei Yang
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China
| |
Collapse
|
17
|
Colacurcio DJ, Nixon RA. Disorders of lysosomal acidification-The emerging role of v-ATPase in aging and neurodegenerative disease. Ageing Res Rev 2016; 32:75-88. [PMID: 27197071 DOI: 10.1016/j.arr.2016.05.004] [Citation(s) in RCA: 352] [Impact Index Per Article: 39.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 05/02/2016] [Accepted: 05/13/2016] [Indexed: 12/21/2022]
Abstract
Autophagy and endocytosis deliver unneeded cellular materials to lysosomes for degradation. Beyond processing cellular waste, lysosomes release metabolites and ions that serve signaling and nutrient sensing roles, linking the functions of the lysosome to various pathways for intracellular metabolism and nutrient homeostasis. Each of these lysosomal behaviors is influenced by the intraluminal pH of the lysosome, which is maintained in the low acidic range by a proton pump, the vacuolar ATPase (v-ATPase). New reports implicate altered v-ATPase activity and lysosomal pH dysregulation in cellular aging, longevity, and adult-onset neurodegenerative diseases, including forms of Parkinson disease and Alzheimer disease. Genetic defects of subunits composing the v-ATPase or v-ATPase-related proteins occur in an increasingly recognized group of familial neurodegenerative diseases. Here, we review the expanding roles of the v-ATPase complex as a platform regulating lysosomal hydrolysis and cellular homeostasis. We discuss the unique vulnerability of neurons to persistent low level lysosomal dysfunction and review recent clinical and experimental studies that link dysfunction of the v-ATPase complex to neurodegenerative diseases across the age spectrum.
Collapse
|
18
|
Ayyadevara S, Balasubramaniam M, Parcon PA, Barger SW, Griffin WST, Alla R, Tackett AJ, Mackintosh SG, Petricoin E, Zhou W, Shmookler Reis RJ. Proteins that mediate protein aggregation and cytotoxicity distinguish Alzheimer's hippocampus from normal controls. Aging Cell 2016; 15:924-39. [PMID: 27448508 PMCID: PMC5013017 DOI: 10.1111/acel.12501] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2016] [Indexed: 12/14/2022] Open
Abstract
Neurodegenerative diseases are distinguished by characteristic protein aggregates initiated by disease‐specific ‘seed’ proteins; however, roles of other co‐aggregated proteins remain largely unexplored. Compact hippocampal aggregates were purified from Alzheimer's and control‐subject pools using magnetic‐bead immunoaffinity pulldowns. Their components were fractionated by electrophoretic mobility and analyzed by high‐resolution proteomics. Although total detergent‐insoluble aggregates from Alzheimer's and controls had similar protein content, within the fractions isolated by tau or Aβ1–42 pulldown, the protein constituents of Alzheimer‐derived aggregates were more abundant, diverse, and post‐translationally modified than those from controls. Tau‐ and Aβ‐containing aggregates were distinguished by multiple components, and yet shared >90% of their protein constituents, implying similar accretion mechanisms. Alzheimer‐specific protein enrichment in tau‐containing aggregates was corroborated for individuals by three analyses. Five proteins inferred to co‐aggregate with tau were confirmed by precise in situ methods, including proximity ligation amplification that requires co‐localization within 40 nm. Nematode orthologs of 21 proteins, which showed Alzheimer‐specific enrichment in tau‐containing aggregates, were assessed for aggregation‐promoting roles in C. elegans by RNA‐interference ‘knockdown’. Fifteen knockdowns (71%) rescued paralysis of worms expressing muscle Aβ, and 12 (57%) rescued chemotaxis disrupted by neuronal Aβ expression. Proteins identified in compact human aggregates, bound by antibody to total tau, were thus shown to play causal roles in aggregation based on nematode models triggered by Aβ1–42. These observations imply shared mechanisms driving both types of aggregation, and/or aggregate‐mediated cross‐talk between tau and Aβ. Knowledge of protein components that promote protein accrual in diverse aggregate types implicates common mechanisms and identifies novel targets for drug intervention.
Collapse
Affiliation(s)
- Srinivas Ayyadevara
- McClellan Veterans Medical Center Central Arkansas Veterans Healthcare Service Little Rock AR 72205 USA
- Department of Geriatrics University of Arkansas for Medical Sciences Little Rock AR 72205 USA
| | - Meenakshisundaram Balasubramaniam
- Department of Geriatrics University of Arkansas for Medical Sciences Little Rock AR 72205 USA
- BioInformatics Program University of Arkansas for Medical Sciences and University of Arkansas at Little Rock Little Rock AR 72205 USA
| | - Paul A. Parcon
- Department of Geriatrics University of Arkansas for Medical Sciences Little Rock AR 72205 USA
| | - Steven W. Barger
- McClellan Veterans Medical Center Central Arkansas Veterans Healthcare Service Little Rock AR 72205 USA
- Department of Geriatrics University of Arkansas for Medical Sciences Little Rock AR 72205 USA
| | - W. Sue T. Griffin
- McClellan Veterans Medical Center Central Arkansas Veterans Healthcare Service Little Rock AR 72205 USA
- Department of Geriatrics University of Arkansas for Medical Sciences Little Rock AR 72205 USA
| | - Ramani Alla
- McClellan Veterans Medical Center Central Arkansas Veterans Healthcare Service Little Rock AR 72205 USA
- Department of Geriatrics University of Arkansas for Medical Sciences Little Rock AR 72205 USA
| | - Alan J. Tackett
- Department of Biochemistry & Molecular Biology University of Arkansas for Medical Sciences Little Rock AR 72205 USA
| | - Samuel G. Mackintosh
- Department of Biochemistry & Molecular Biology University of Arkansas for Medical Sciences Little Rock AR 72205 USA
| | - Emanuel Petricoin
- Center for Applied Proteomics and Molecular Medicine George Mason University Manassas VA 20110 USA
| | - Weidong Zhou
- Center for Applied Proteomics and Molecular Medicine George Mason University Manassas VA 20110 USA
| | - Robert J. Shmookler Reis
- McClellan Veterans Medical Center Central Arkansas Veterans Healthcare Service Little Rock AR 72205 USA
- Department of Geriatrics University of Arkansas for Medical Sciences Little Rock AR 72205 USA
- Department of Biochemistry & Molecular Biology University of Arkansas for Medical Sciences Little Rock AR 72205 USA
| |
Collapse
|
19
|
Wang HY, Lin ZL, Yu XF, Bao Y, Cui XS, Kim NH. Computational Prediction of Alzheimer's and Parkinson's Disease MicroRNAs in Domestic Animals. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2015; 29:782-92. [PMID: 26954182 PMCID: PMC4852244 DOI: 10.5713/ajas.15.0413] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 07/23/2015] [Accepted: 08/14/2015] [Indexed: 12/27/2022]
Abstract
As the most common neurodegenerative diseases, Alzheimer’s disease (AD) and Parkinson’s disease (PD) are two of the main health concerns for the elderly population. Recently, microRNAs (miRNAs) have been used as biomarkers of infectious, genetic, and metabolic diseases in humans but they have not been well studied in domestic animals. Here we describe a computational biology study in which human AD- and PD-associated miRNAs (ADM and PDM) were utilized to predict orthologous miRNAs in the following domestic animal species: dog, cow, pig, horse, and chicken. In this study, a total of 121 and 70 published human ADM and PDM were identified, respectively. Thirty-seven miRNAs were co-regulated in AD and PD. We identified a total of 105 unrepeated human ADM and PDM that had at least one 100% identical animal homolog, among which 81 and 54 showed 100% sequence identity with 241 and 161 domestic animal miRNAs, respectively. Over 20% of the total mature horse miRNAs (92) showed perfect matches to AD/PD-associated miRNAs. Pigs, dogs, and cows have similar numbers of AD/PD-associated miRNAs (63, 62, and 59). Chickens had the least number of perfect matches (34). Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses suggested that humans and dogs are relatively similar in the functional pathways of the five selected highly conserved miRNAs. Taken together, our study provides the first evidence for better understanding the miRNA-AD/PD associations in domestic animals, and provides guidance to generate domestic animal models of AD/PD to replace the current rodent models.
Collapse
Affiliation(s)
- Hai Yang Wang
- Department of Animal Sciences, Chungbuk National University, Cheongju 362-763, Korea
| | - Zi Li Lin
- Department of Animal Sciences, Chungbuk National University, Cheongju 362-763, Korea
| | - Xian Feng Yu
- College of Animal Sciences, Jilin University, Changchun, Jilin 130062, China
| | - Yuan Bao
- College of Animal Sciences, Jilin University, Changchun, Jilin 130062, China
| | - Xiang-Shun Cui
- Department of Animal Sciences, Chungbuk National University, Cheongju 362-763, Korea
| | - Nam-Hyung Kim
- Department of Animal Sciences, Chungbuk National University, Cheongju 362-763, Korea.,College of Animal Sciences, Jilin University, Changchun, Jilin 130062, China
| |
Collapse
|
20
|
Lee JH, Yoo JY, You YA, Kwon WS, Lee SM, Pang MG, Kim YJ. Proteomic analysis of fetal programming-related obesity markers. Proteomics 2015; 15:2669-77. [PMID: 25886259 DOI: 10.1002/pmic.201400359] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 02/16/2015] [Accepted: 04/14/2015] [Indexed: 12/14/2022]
Abstract
The objectives of this study were to analyze fetal programming in rat brain using proteomic analysis and to identify fetal programming-related obesity markers. Sprague-Dawley rats were divided into four feeding groups: (i) the Ad Libitum (AdLib)/AdLib group was given a normal diet during pregnancy and the lactation period; (ii) the AdLib/maternal food restriction group (FR) was subjected to 50% FR during the lactation period; (iii) the FR/AdLib group was subjected to 50% FR during pregnancy; and (iv) the FR/FR group was subjected to 50% FR during pregnancy and the lactation period. Offspring from each group were sacrificed at 3 weeks of age and whole brains were dissected. To obtain a maximum number of protein markers related to obesity, 2DE and Pathway Studio bioinformatics analysis were performed. The identities of the markers among the selected and candidate proteins were confirmed by Western blotting and immunohistochemistry. Proteomic and bioinformatics analyses revealed that expression of ubiquitin carboxy-terminal hydrolase L1 (UCHL1) and Secernin 1 (SCRN1) were significantly different in the FR/AdLib group compared with the AdLib/AdLib group for both male and female offspring. These findings suggest that UCHL1 and SCRN1 may be used as fetal programming-related obesity markers.
Collapse
Affiliation(s)
- Ji Hye Lee
- Department of Obstetrics and Gynecology, Ewha Womans University, Seoul, South Korea
| | - Jae Young Yoo
- Department of Obstetrics and Gynecology, Ewha Womans University, Seoul, South Korea
| | - Young-Ah You
- Department of Obstetrics and Gynecology, Ewha Womans University, Seoul, South Korea
| | - Woo-Sung Kwon
- Department of Animal Science and Technology, Chung-Ang University, Anseong, Gyeonggi-Do 456-756, South Korea
| | - Sang Mi Lee
- Department of Obstetrics and Gynecology, Ewha Womans University, Seoul, South Korea
| | - Myung-Geol Pang
- Department of Animal Science and Technology, Chung-Ang University, Anseong, Gyeonggi-Do 456-756, South Korea
| | - Young Ju Kim
- Department of Obstetrics and Gynecology, Ewha Womans University, Seoul, South Korea
| |
Collapse
|
21
|
The Potential of Proteomics in Understanding Neurodegeneration. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2015; 121:25-58. [DOI: 10.1016/bs.irn.2015.05.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
22
|
Chang RYK, Etheridge N, Dodd PR, Nouwens AS. Targeted quantitative analysis of synaptic proteins in Alzheimer’s disease brain. Neurochem Int 2014; 75:66-75. [DOI: 10.1016/j.neuint.2014.05.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 05/22/2014] [Accepted: 05/26/2014] [Indexed: 11/15/2022]
|
23
|
Jung IS, Kim HJ, Noh R, Kim SC, Kim CW. Effects of extremely low frequency magnetic fields on NGF induced neuronal differentiation of PC12 cells. Bioelectromagnetics 2014; 35:459-69. [DOI: 10.1002/bem.21861] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 04/25/2014] [Indexed: 12/21/2022]
Affiliation(s)
- In-Soo Jung
- College of Life Sciences and Biotechnology; Korea University; Seoul Korea
| | - Hyun-Jung Kim
- College of Life Sciences and Biotechnology; Korea University; Seoul Korea
| | - Ran Noh
- College of Life Sciences and Biotechnology; Korea University; Seoul Korea
| | - Soo-Chan Kim
- Department of Electrical Electronic and Control Engineering; Hankyong National University; Anseong-si Kyonggi-do Korea
| | - Chan-Wha Kim
- College of Life Sciences and Biotechnology; Korea University; Seoul Korea
| |
Collapse
|