1
|
Tandon R, Zhao L, Watson CM, Sarkar N, Elmor M, Heilman C, Sanders K, Hales CM, Yang H, Loring DW, Goldstein FC, Hanfelt JJ, Duong DM, Johnson ECB, Wingo AP, Wingo TS, Roberts BR, Seyfried NT, Levey AI, Lah JJ, Mitchell CS. Stratifying risk of Alzheimer's disease in healthy middle-aged individuals with machine learning. Brain Commun 2025; 7:fcaf121. [PMID: 40226382 PMCID: PMC11986205 DOI: 10.1093/braincomms/fcaf121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 02/23/2025] [Accepted: 03/24/2025] [Indexed: 04/15/2025] Open
Abstract
Alzheimer's disease has a prolonged asymptomatic phase during which pathological changes accumulate before clinical symptoms emerge. This study aimed to stratify the risk of clinical disease to inform future disease-modifying treatments. Cerebrospinal fluid analysis from participants in the Emory Healthy Brain Study was used to classify individuals based on amyloid beta 42 (Aβ42), total tau (tTau) and phosphorylated tau (pTau) levels. Cognitively normal (CN), biomarker-positive (CN)/BM+individuals were identified using a tTau: Aβ42 ratio > 0.24, determined by Gaussian mixture models. CN/BM+ individuals (n = 134) were classified as having asymptomatic Alzheimer's disease (AsymAD), while CN, biomarker-negative (CN/BM-) individuals served as controls (n = 134). Cognitively symptomatic, biomarker-positive individuals with an Alzheimer's disease diagnosis confirmed by the Emory Cognitive Neurology Clinic were labelled as Alzheimer's disease (n = 134). Study groups were matched for age, sex, race and education. Cerebrospinal fluid samples from these matched Emory Healthy Brain Study groups were analysed using targeted proteomics via selected reaction monitoring mass spectrometry. The targeted cerebrospinal fluid panel included 75 peptides from 58 unique proteins. Machine learning approaches identified a subset of eight peptides (ADQDTIR, AQALEQAK, ELQAAQAR, EPVAGDAVPGPK, IASNTQSR, LGADMEDVCGR, VVSSIEQK, YDNSLK) that distinguished between CN/BM- and symptomatic Alzheimer's disease samples with a binary classifier area under the curve performance of 0.98. Using these eight peptides, Emory Healthy Brain Study AsymAD cases were further stratified into 'Control-like' and 'Alzheimer's disease-like' subgroups, representing varying levels of risk for developing clinical disease. The eight peptides were evaluated in an independent dataset from the Alzheimer's Disease Neuroimaging Initiative, effectively distinguishing CN/BM- from symptomatic Alzheimer's disease cases (area under the curve = 0.89) and stratifying AsymAD individuals into control-like and Alzheimer's disease-like subgroups (area under the curve = 0.89). In the absence of matched longitudinal data, an established cross-sectional event-based disease progression model was employed to assess the generalizability of these peptides for risk stratification. In summary, results from two independent modelling methods and datasets demonstrate that the identified eight peptides effectively stratify the risk of progression from asymptomatic to symptomatic Alzheimer's disease.
Collapse
Affiliation(s)
- Raghav Tandon
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Center for Machine Learning, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Liping Zhao
- Department of Biostatistics and Bioinformatics, Emory School of Public Health, Atlanta, GA 30322, USA
- Emory Goizueta Alzheimer’s Disease Research Center, Atlanta, GA 30329, USA
| | - Caroline M Watson
- Emory Goizueta Alzheimer’s Disease Research Center, Atlanta, GA 30329, USA
- Department of Neurology, Emory School of Medicine, Atlanta, GA 30322, USA
| | - Neel Sarkar
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Morgan Elmor
- Emory Goizueta Alzheimer’s Disease Research Center, Atlanta, GA 30329, USA
- Department of Neurology, Emory School of Medicine, Atlanta, GA 30322, USA
| | - Craig Heilman
- Emory Goizueta Alzheimer’s Disease Research Center, Atlanta, GA 30329, USA
- Department of Neurology, Emory School of Medicine, Atlanta, GA 30322, USA
| | - Katherine Sanders
- Emory Goizueta Alzheimer’s Disease Research Center, Atlanta, GA 30329, USA
- Department of Neurology, Emory School of Medicine, Atlanta, GA 30322, USA
| | - Chadwick M Hales
- Emory Goizueta Alzheimer’s Disease Research Center, Atlanta, GA 30329, USA
- Department of Neurology, Emory School of Medicine, Atlanta, GA 30322, USA
- Center for Neurodegenerative Disease, Emory University, Atlanta, GA 30322, USA
| | - Huiying Yang
- Department of Biostatistics and Bioinformatics, Emory School of Public Health, Atlanta, GA 30322, USA
- Emory Goizueta Alzheimer’s Disease Research Center, Atlanta, GA 30329, USA
| | - David W Loring
- Emory Goizueta Alzheimer’s Disease Research Center, Atlanta, GA 30329, USA
- Department of Neurology, Emory School of Medicine, Atlanta, GA 30322, USA
| | - Felicia C Goldstein
- Emory Goizueta Alzheimer’s Disease Research Center, Atlanta, GA 30329, USA
- Department of Neurology, Emory School of Medicine, Atlanta, GA 30322, USA
| | - John J Hanfelt
- Department of Biostatistics and Bioinformatics, Emory School of Public Health, Atlanta, GA 30322, USA
- Emory Goizueta Alzheimer’s Disease Research Center, Atlanta, GA 30329, USA
| | - Duc M Duong
- Emory Goizueta Alzheimer’s Disease Research Center, Atlanta, GA 30329, USA
- Department of Neurology, Emory School of Medicine, Atlanta, GA 30322, USA
- Department of Biochemistry, Emory School of Medicine, Atlanta, GA 30322, USA
| | - Erik C B Johnson
- Emory Goizueta Alzheimer’s Disease Research Center, Atlanta, GA 30329, USA
- Department of Neurology, Emory School of Medicine, Atlanta, GA 30322, USA
- Center for Neurodegenerative Disease, Emory University, Atlanta, GA 30322, USA
| | - Aliza P Wingo
- Department of Psychiatry, Emory School of Medicine, Atlanta, GA 30322, USA
- Division of Mental Health, Atlanta VA Medical Center, Atlanta, GA 30033, USA
| | - Thomas S Wingo
- Emory Goizueta Alzheimer’s Disease Research Center, Atlanta, GA 30329, USA
- Department of Neurology, Emory School of Medicine, Atlanta, GA 30322, USA
- Center for Neurodegenerative Disease, Emory University, Atlanta, GA 30322, USA
| | - Blaine R Roberts
- Center for Neurodegenerative Disease, Emory University, Atlanta, GA 30322, USA
- Department of Biochemistry, Emory School of Medicine, Atlanta, GA 30322, USA
| | - Nicholas T Seyfried
- Emory Goizueta Alzheimer’s Disease Research Center, Atlanta, GA 30329, USA
- Center for Neurodegenerative Disease, Emory University, Atlanta, GA 30322, USA
- Department of Biochemistry, Emory School of Medicine, Atlanta, GA 30322, USA
| | - Allan I Levey
- Emory Goizueta Alzheimer’s Disease Research Center, Atlanta, GA 30329, USA
- Department of Neurology, Emory School of Medicine, Atlanta, GA 30322, USA
- Center for Neurodegenerative Disease, Emory University, Atlanta, GA 30322, USA
| | - James J Lah
- Emory Goizueta Alzheimer’s Disease Research Center, Atlanta, GA 30329, USA
- Department of Neurology, Emory School of Medicine, Atlanta, GA 30322, USA
- Center for Neurodegenerative Disease, Emory University, Atlanta, GA 30322, USA
| | - Cassie S Mitchell
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Center for Machine Learning, Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
2
|
Pashaei E, Pashaei E, Aydin N. Biomarker Identification for Alzheimer's Disease Using a Multi-Filter Gene Selection Approach. Int J Mol Sci 2025; 26:1816. [PMID: 40076442 PMCID: PMC11898513 DOI: 10.3390/ijms26051816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/09/2025] [Accepted: 02/17/2025] [Indexed: 03/14/2025] Open
Abstract
There is still a lack of effective therapies for Alzheimer's disease (AD), the leading cause of dementia and cognitive decline. Identifying reliable biomarkers and therapeutic targets is crucial for advancing AD research. In this study, we developed an aggregative multi-filter gene selection approach to identify AD biomarkers. This method integrates hub gene ranking techniques, such as degree and bottleneck, with feature selection algorithms, including Random Forest and Double Input Symmetrical Relevance, and applies ranking aggregation to improve accuracy and robustness. Five publicly available AD-related microarray datasets (GSE48350, GSE36980, GSE132903, GSE118553, and GSE5281), covering diverse brain regions like the hippocampus and frontal cortex, were analyzed, yielding 803 overlapping differentially expressed genes from 464 AD and 492 normal cases. An independent dataset (GSE109887) was used for external validation. The approach identified 50 prioritized genes, achieving an AUC of 86.8 in logistic regression on the validation dataset, highlighting their predictive value. Pathway analysis revealed involvement in critical biological processes such as synaptic vesicle cycles, neurodegeneration, and cognitive function. These findings provide insights into potential therapeutic targets for AD.
Collapse
Affiliation(s)
- Elnaz Pashaei
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Elham Pashaei
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Nizamettin Aydin
- Department of Computer Engineering, Faculty of Computer and Informatics Engineering, Istanbul Technical University, Istanbul 34467, Türkiye;
| |
Collapse
|
3
|
Rojas-Pirela M, Andrade-Alviárez D, Rojas V, Marcos M, Salete-Granado D, Chacón-Arnaude M, Pérez-Nieto MÁ, Kemmerling U, Concepción JL, Michels PAM, Quiñones W. Exploring glycolytic enzymes in disease: potential biomarkers and therapeutic targets in neurodegeneration, cancer and parasitic infections. Open Biol 2025; 15:240239. [PMID: 39904372 PMCID: PMC11793985 DOI: 10.1098/rsob.240239] [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: 08/26/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 02/06/2025] Open
Abstract
Glycolysis, present in most organisms, is evolutionarily one of the oldest metabolic pathways. It has great relevance at a physiological level because it is responsible for generating ATP in the cell through the conversion of glucose into pyruvate and reducing nicotinamide adenine dinucleotide (NADH) (that may be fed into the electron chain in the mitochondria to produce additional ATP by oxidative phosphorylation), as well as for producing intermediates that can serve as substrates for other metabolic processes. Glycolysis takes place through 10 consecutive chemical reactions, each of which is catalysed by a specific enzyme. Although energy transduction by glucose metabolism is the main function of this pathway, involvement in virulence, growth, pathogen-host interactions, immunomodulation and adaptation to environmental conditions are other functions attributed to this metabolic pathway. In humans, where glycolysis occurs mainly in the cytosol, the mislocalization of some glycolytic enzymes in various other subcellular locations, as well as alterations in their expression and regulation, has been associated with the development and progression of various diseases. In this review, we describe the role of glycolytic enzymes in the pathogenesis of diseases of clinical interest. In addition, the potential role of these enzymes as targets for drug development and their potential for use as diagnostic and prognostic markers of some pathologies are also discussed.
Collapse
Affiliation(s)
- Maura Rojas-Pirela
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca37007, Spain
- Unidad de Medicina Molecular, Departamento de Medicina, Universidad de Salamanca, Salamanca37007, Spain
- Servicio de Medicina Interna, Hospital Universitario de Salamanca, Salamanca37007, Spain
| | - Diego Andrade-Alviárez
- Laboratorio de Enzimología de Parásitos, Departamento de Biología, Facultad de Ciencias, Universidad de Los Andes, Mérida5101, Venezuela
| | - Verónica Rojas
- Instituto de Biología, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso2373223, Chile
| | - Miguel Marcos
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca37007, Spain
- Unidad de Medicina Molecular, Departamento de Medicina, Universidad de Salamanca, Salamanca37007, Spain
- Servicio de Medicina Interna, Hospital Universitario de Salamanca, Salamanca37007, Spain
| | - Daniel Salete-Granado
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca37007, Spain
- Unidad de Medicina Molecular, Departamento de Medicina, Universidad de Salamanca, Salamanca37007, Spain
| | - Marirene Chacón-Arnaude
- Laboratorio de Enzimología de Parásitos, Departamento de Biología, Facultad de Ciencias, Universidad de Los Andes, Mérida5101, Venezuela
| | - María Á. Pérez-Nieto
- Unidad de Medicina Molecular, Departamento de Medicina, Universidad de Salamanca, Salamanca37007, Spain
- Fundación Instituto de Estudios de Ciencias de la Salud de Castilla y León, Soria42002, Spain
| | - Ulrike Kemmerling
- Instituto de Ciencias Biomédicas, Universidad de Chile, Facultad de Medicina, Santiago de Chile8380453, Chile
| | - Juan Luis Concepción
- Laboratorio de Enzimología de Parásitos, Departamento de Biología, Facultad de Ciencias, Universidad de Los Andes, Mérida5101, Venezuela
| | - Paul A. M. Michels
- School of Biological Sciences, University of Edinburgh, The King’s Buildings, EdinburghEH9 3FL, UK
| | - Wilfredo Quiñones
- Laboratorio de Enzimología de Parásitos, Departamento de Biología, Facultad de Ciencias, Universidad de Los Andes, Mérida5101, Venezuela
| |
Collapse
|
4
|
Markovich SW, Frey BL, Scalf M, Shortreed MR, Smith LM. Dehydroamino acids and their crosslinks in Alzheimer's disease aggregates. Brain Commun 2025; 7:fcaf019. [PMID: 39882022 PMCID: PMC11775630 DOI: 10.1093/braincomms/fcaf019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/16/2024] [Accepted: 01/13/2025] [Indexed: 01/31/2025] Open
Abstract
Alzheimer's disease (AD) is characterized by the accumulation of protein aggregates, which are thought to be influenced by posttranslational modifications (PTMs). Dehydroamino acids (DHAAs) are rarely observed PTMs that contain an electrophilic alkene capable of forming protein-protein crosslinks, which may lead to protein aggregation. We report here the discovery of DHAAs in the protein aggregates from AD, constituting an unknown and previously unsuspected source of extensive proteomic complexity. We used mass spectrometry-based proteomics to discover 404 sites of DHAA formation in 171 proteins from protein aggregate-enriched human brain samples, 6-fold more sites than observed in the soluble protein fractions. The DHAA modifications are observed both directly and in the form of conjugates after reacting with abundant cellular nucleophiles or crosslinking to nucleophilic amino acid residues. We report 11 such crosslinks, including three in the Tau protein, which are 10-fold more abundant in AD samples compared with age-matched controls. Many of the proteins found to contain DHAAs and their conjugates are involved in protein aggregation or pathways dysregulated in AD. DHAAs are prevalent modifications in the AD brain proteome and give rise to protein crosslinks that may contribute to protein aggregation.
Collapse
Affiliation(s)
- Samuel W Markovich
- Department of Chemistry, University of Wisconsin-Madison; Madison, WI 53706, USA
| | - Brian L Frey
- Department of Chemistry, University of Wisconsin-Madison; Madison, WI 53706, USA
| | - Mark Scalf
- Department of Chemistry, University of Wisconsin-Madison; Madison, WI 53706, USA
| | - Michael R Shortreed
- Department of Chemistry, University of Wisconsin-Madison; Madison, WI 53706, USA
| | - Lloyd M Smith
- Department of Chemistry, University of Wisconsin-Madison; Madison, WI 53706, USA
| |
Collapse
|
5
|
Zhang Z, Luo G, Ma Y, Wu Z, Peng S, Chen S, Wu Y. GraphkmerDTA: integrating local sequence patterns and topological information for drug-target binding affinity prediction and applications in multi-target anti-Alzheimer's drug discovery. Mol Divers 2025:10.1007/s11030-024-11065-7. [PMID: 39792322 DOI: 10.1007/s11030-024-11065-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 11/22/2024] [Indexed: 01/12/2025]
Abstract
Identifying drug-target binding affinity (DTA) plays a critical role in early-stage drug discovery. Despite the availability of various existing methods, there are still two limitations. Firstly, sequence-based methods often extract features from fixed length protein sequences, requiring truncation or padding, which can result in information loss or the introduction of unwanted noise. Secondly, structure-based methods prioritize extracting topological information but struggle to effectively capture sequence features. To address these challenges, we propose a novel deep learning model named GraphkmerDTA, which integrates Kmer features with structural topology. Specifically, GraphkmerDTA utilizes graph neural networks to extract topological features from both molecules and proteins, while fully connected networks learn local sequence patterns from the Kmer features of proteins. Experimental results indicate that GraphkmerDTA outperforms existing methods on benchmark datasets. Furthermore, a case study on lung cancer demonstrates the effectiveness of GraphkmerDTA, as it successfully identifies seven known EGFR inhibitors from a screening library of over two thousand compounds. To further assess the practical utility of GraphkmerDTA, we integrated it with network pharmacology to investigate the mechanisms underlying the therapeutic effects of Lonicera japonica flower in treating Alzheimer's disease. Through this interdisciplinary approach, three potential compounds were identified and subsequently validated through molecular docking studies. In conclusion, we present not only a novel AI model for the DTA task but also demonstrate its practical application in drug discovery by integrating modern AI approaches with traditional drug discovery methodologies.
Collapse
Affiliation(s)
- Zuolong Zhang
- School of Software, Henan University, Kaifeng, 475000, Henan, China
| | - Gang Luo
- School of Mathematics and Computer Science, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Yixuan Ma
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases Ministry of Education, Jiangxi Province Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering, Gannan Medical University, Ganzhou, 341000, Jiangxi, China
| | - Zhaoqi Wu
- School of Basic Medicine Sciences, Gannan Medical University, Ganzhou, 341000, Jiangxi, China
| | - Shuo Peng
- Department of Computer Science, Jinggangshan University, Ji'an, 343009, Jiangxi, China
| | - Shengbo Chen
- Henan Engineering Research Center of Intelligent Technology and Application, Henan University, Kaifeng, 475000, Henan, China.
- School of Software, Nanchang University, Nanchang, 330031, Jiangxi, China.
| | - Yi Wu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases Ministry of Education, Jiangxi Province Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering, Gannan Medical University, Ganzhou, 341000, Jiangxi, China.
| |
Collapse
|
6
|
Muronets VI, Kudryavtseva SS, Kurochkina LP, Leisi EV, Stroylova YY, Schmalhausen EV. Factors Affecting Pathological Amyloid Protein Transformation: From Post-Translational Modifications to Chaperones. BIOCHEMISTRY. BIOKHIMIIA 2025; 90:S164-S192. [PMID: 40164158 DOI: 10.1134/s0006297924604003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/10/2024] [Accepted: 10/30/2024] [Indexed: 04/02/2025]
Abstract
The review discusses the influence of various factors (e.g., post-translational modifications and chaperones) on the pathological transformation of amyloidogenic proteins involved in the onset and development of neurodegenerative diseases (Alzheimer's and Parkinson's diseases) and spongiform encephalopathies of various origin with special focus on the role of α-synuclein, prion protein, and, to a lesser extent, beta-amyloid peptide. The factors investigated by the authors of this review are discussed in more detail, including posttranslational modifications (glycation and S-nitrosylation), cinnamic acid derivatives and dendrimers, and chaperonins (eukaryotic, bacterial, and phage). A special section is devoted to the role of the gastrointestinal microbiota in the pathogenesis of amyloid neurodegenerative diseases, in particular, its involvement in the transformation of infectious prions and possibly other proteins capable of prion-like transmission of amyloidogenic diseases.
Collapse
Affiliation(s)
- Vladimir I Muronets
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia.
- Butlerov Institute of Chemistry, Kazan (Volga Region) Federal University, Kazan, 420008, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Sofiya S Kudryavtseva
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Lidia P Kurochkina
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Evgeniia V Leisi
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Yulia Yu Stroylova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Elena V Schmalhausen
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| |
Collapse
|
7
|
Khan NS, Choudhary S, Ali M, Shawaz M, Lohnes BJ, Poddar NK. Unveiling biomarker detection in Alzheimer's disease: a computational approach to microarray analysis. 3 Biotech 2024; 14:311. [PMID: 39606011 PMCID: PMC11589038 DOI: 10.1007/s13205-024-04159-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 11/10/2024] [Indexed: 11/29/2024] Open
Abstract
Alzheimer's disease (AD) is a major neurodegenerative condition that affects a significant number of people around the world, making understanding the underlying molecular mechanisms fundamental for identifying predictive biomarkers and therapeutic targets for treating AD. Analysis of the gene expression profile GSE5281, consisting of 161 samples (87 AD and 74 control samples) revealed differentially expressed genes (DEGs) used for KEGG screening to connect dysregulated genes to metabolic pathways or other neurological diseases including Parkinson's, prion, and Huntington's and construction of a protein interaction network. Protein-protein interaction (PPI) network and module analysis uncovered the hub genes ACTB, ACTG1, ATP5A1, CCT2, CDC42, EGFR, FN1, GAPDH, GFAP, GRIA1, HSP90AB1, MAPK1, PSMA3, PSMD14, SNAP25, SNCA, SOD1, SOX2, TPI1, and YWHAZ. The analysis revealed a link between dysregulated genes and processes in AD pathology, including the promotion of osteoporosis, an altered nucleotide metabolism, microtubule stability, and the dysfunctionality of the blood-brain barrier (BBB). These targets might be used as predictive biomarkers or to develop curative and preventive therapeutic approaches for treating AD.
Collapse
Affiliation(s)
- Noor Saba Khan
- Biomedical Informatics Centre, ICMR-National Institute of Pathology, New Delhi, 110029 India
| | - Saumya Choudhary
- Biomedical Informatics Centre, ICMR-National Institute of Pathology, New Delhi, 110029 India
| | - Mohd. Ali
- Ram-Eesh Institute of Vocational & Technical Education, Gautam Budh Nagar, Greater Noida, Uttar Pradesh 201310 India
| | - Mohd. Shawaz
- Ram-Eesh Institute of Vocational & Technical Education, Gautam Budh Nagar, Greater Noida, Uttar Pradesh 201310 India
| | - Benedikt Jakob Lohnes
- Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany
| | - Nitesh Kumar Poddar
- Department of Biosciences, Manipal University Jaipur, Near GVK Toll Plaza, Jaipur-Ajmer Express Highway, Dehmi Kalan, Jaipur, Rajasthan 303007 India
| |
Collapse
|
8
|
Górska AM, Santos-García I, Eiriz I, Brüning T, Nyman T, Pahnke J. Evaluation of cerebrospinal fluid (CSF) and interstitial fluid (ISF) mouse proteomes for the validation and description of Alzheimer's disease biomarkers. J Neurosci Methods 2024; 411:110239. [PMID: 39102902 DOI: 10.1016/j.jneumeth.2024.110239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/22/2024] [Accepted: 07/26/2024] [Indexed: 08/07/2024]
Abstract
BACKGROUND Mass spectrometry (MS)-based cerebrospinal fluid (CSF) proteomics is an important method for discovering biomarkers of neurodegenerative diseases. CSF serves as a reservoir for interstitial fluid (ISF), and extensive communication between the two fluid compartments helps to remove waste products from the brain. NEW METHOD We performed proteomic analyses of both CSF and ISF fluid compartments using intracerebral microdialysis to validate and detect novel biomarkers of Alzheimer's disease (AD) in APPtg and C57Bl/6J control mice. RESULTS We identified up to 625 proteins in ISF and 4483 proteins in CSF samples. By comparing the biofluid profiles of APPtg and C57Bl/6J mice, we detected 37 and 108 significantly up- and downregulated candidates, respectively. In ISF, 7 highly regulated proteins, such as Gfap, Aldh1l1, Gstm1, and Txn, have already been implicated in AD progression, whereas in CSF, 9 out of 14 highly regulated proteins, such as Apba2, Syt12, Pgs1 and Vsnl1, have also been validated to be involved in AD pathogenesis. In addition, we also detected new interesting regulated proteins related to the control of synapses and neurotransmission (Kcna2, Cacng3, and Clcn6) whose roles as AD biomarkers should be further investigated. COMPARISON WITH EXISTING METHODS This newly established combined protocol provides better insight into the mutual communication between ISF and CSF as an analysis of tissue or CSF compartments alone. CONCLUSIONS The use of multiple fluid compartments, ISF and CSF, for the detection of their biological communication enables better detection of new promising AD biomarkers.
Collapse
Affiliation(s)
- Anna Maria Górska
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO) and Section of Neuropathology Research, Department of Pathology, Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, Oslo NO-0372, Norway.
| | - Irene Santos-García
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO) and Section of Neuropathology Research, Department of Pathology, Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, Oslo NO-0372, Norway.
| | - Ivan Eiriz
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO) and Section of Neuropathology Research, Department of Pathology, Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, Oslo NO-0372, Norway.
| | - Thomas Brüning
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO) and Section of Neuropathology Research, Department of Pathology, Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, Oslo NO-0372, Norway.
| | - Tuula Nyman
- Proteomics Core Facility, Department of Immunology, Oslo University Hospital (OUS) and University of Oslo (UiO), Faculty of Medicine, Sognsvannsveien 20, Oslo NO-0372, Norway.
| | - Jens Pahnke
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO) and Section of Neuropathology Research, Department of Pathology, Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, Oslo NO-0372, Norway; Institute of Nutritional Medicine (INUM) and Lübeck Institute of Dermatology (LIED), University of Lübeck (UzL) and University Medical Center Schleswig-Holstein (UKSH), Ratzeburger Allee 160, Lübeck D-23538, Germany; Department of Pharmacology, Faculty of Medicine and Life Sciences, University of Latvia, Jelgavas iela 3, Rīga LV-1004, Latvia; School of Neurobiology, Biochemistry and Biophysics, The Georg S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv IL-6997801, Israel.
| |
Collapse
|
9
|
Ma X, Sun Y, Li C, Wang M, Zang Q, Zhang X, Wang F, Niu Y, Hua J. Novel Insights Into DLAT's Role in Alzheimer's Disease-Related Copper Toxicity Through Microglial Exosome Dynamics. CNS Neurosci Ther 2024; 30:e70064. [PMID: 39428563 PMCID: PMC11491298 DOI: 10.1111/cns.70064] [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] [Revised: 08/10/2024] [Accepted: 09/03/2024] [Indexed: 10/22/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a complex neurodegenerative disorder, with recent research emphasizing the roles of microglia and their secreted extracellular vesicles in AD pathology. However, the involvement of specific molecular pathways contributing to neuronal death in the context of copper toxicity remains largely unexplored. OBJECTIVE This study investigates the interaction between pyruvate kinase M2 (PKM2) and dihydrolipoamide S-acetyltransferase (DLAT), particularly focusing on copper-induced neuronal death in Alzheimer's disease. METHODS Gene expression datasets were analyzed to identify key factors involved in AD-related copper toxicity. The role of DLAT was validated using 5xFAD transgenic mice, while in vitro experiments were conducted to assess the impact of microglial exosomes on neuronal PKM2 transfer and DLAT expression. The effects of inhibiting the PKM2 transfer via microglial exosomes on DLAT expression and copper-induced neuronal death were also evaluated. RESULTS DLAT was identified as a critical factor in the pathology of AD, particularly in copper toxicity. In 5xFAD mice, increased DLAT expression was linked to hippocampal damage and cognitive decline. In vitro, microglial exosomes were shown to facilitate the transfer of PKM2 to neurons, leading to upregulation of DLAT expression and increased copper-induced neuronal death. Inhibition of PKM2 transfer via exosomes resulted in a significant reduction in DLAT expression, mitigating neuronal death and slowing AD progression. CONCLUSION This study uncovers a novel pathway involving microglial exosomes and the PKM2-DLAT interaction in copper-induced neuronal death, providing potential therapeutic targets for Alzheimer's disease. Blocking PKM2 transfer could offer new strategies for reducing neuronal damage and slowing disease progression in AD.
Collapse
Affiliation(s)
- Xiang Ma
- Laboratory of Biochemistry and PharmacyTaiyuan Institute of TechnologyTaiyuanP. R. China
| | - Yusheng Sun
- Laboratory of Biochemistry and PharmacyTaiyuan Institute of TechnologyTaiyuanP. R. China
| | - Changchun Li
- Department of Chemistry and Chemical EngineeringTaiyuan Institute of TechnologyTaiyuanP. R. China
| | - Man Wang
- Laboratory of Biochemistry and PharmacyTaiyuan Institute of TechnologyTaiyuanP. R. China
| | - Qijiao Zang
- Laboratory of Biochemistry and PharmacyTaiyuan Institute of TechnologyTaiyuanP. R. China
| | - Xuxia Zhang
- Laboratory of Biochemistry and PharmacyTaiyuan Institute of TechnologyTaiyuanP. R. China
| | - Feng Wang
- Department of Chemistry and Chemical EngineeringTaiyuan Institute of TechnologyTaiyuanP. R. China
| | - Yulan Niu
- Department of Chemistry and Chemical EngineeringTaiyuan Institute of TechnologyTaiyuanP. R. China
| | - Jiai Hua
- Laboratory of Biochemistry and PharmacyTaiyuan Institute of TechnologyTaiyuanP. R. China
| |
Collapse
|
10
|
Schmalhausen EV, Medvedeva MV, Muronetz VI. Glyceraldehyde-3-phosphate dehydrogenase is involved in the pathogenesis of Alzheimer's disease. Arch Biochem Biophys 2024; 758:110065. [PMID: 38906311 DOI: 10.1016/j.abb.2024.110065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 06/23/2024]
Abstract
One of important characteristics of Alzheimer's disease is a persistent oxidative/nitrosative stress caused by pro-oxidant properties of amyloid-beta peptide (Aβ) and chronic inflammation in the brain. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) is easily oxidized under oxidative stress. Numerous data indicate that oxidative modifications of GAPDH in vitro and in cell cultures stimulate GAPDH denaturation and aggregation, and the catalytic cysteine residue Cys152 is important for these processes. Both intracellular and extracellular GAPDH aggregates are toxic for the cells. Interaction of denatured GAPDH with soluble Aβ results in mixed insoluble aggregates with increased toxicity. The above-described properties of GAPDH (sensitivity to oxidation and propensity to form aggregates, including mixed aggregates with Aβ) determine its role in the pathogenesis of Alzheimer's disease.
Collapse
Affiliation(s)
- E V Schmalhausen
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskie Gory 1, Bld 40, 119991, Moscow, Russia.
| | - M V Medvedeva
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Leninskie Gory 1, Bld 73, 119991, Moscow, Russia
| | - V I Muronetz
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskie Gory 1, Bld 40, 119991, Moscow, Russia; Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Leninskie Gory 1, Bld 73, 119991, Moscow, Russia
| |
Collapse
|
11
|
Adeoye T, Shah SI, Ullah G. Systematic Analysis of Biological Processes Reveals Gene Co-expression Modules Driving Pathway Dysregulation in Alzheimer's Disease. Aging Dis 2024:AD.2024.0429. [PMID: 38913039 DOI: 10.14336/ad.2024.0429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 06/12/2024] [Indexed: 06/25/2024] Open
Abstract
Alzheimer's disease (AD) manifests as a complex systems pathology with intricate interplay among various genes and biological processes. Traditional differential gene expression (DEG) analysis, while commonly employed to characterize AD-driven perturbations, does not sufficiently capture the full spectrum of underlying biological processes. Utilizing single-nucleus RNA-sequencing data from postmortem brain samples across key regions-middle temporal gyrus, superior frontal gyrus, and entorhinal cortex-we provide a comprehensive systematic analysis of disrupted processes in AD. We go beyond the DEG-centric analysis by integrating pathway activity analysis with weighted gene co-expression patterns to comprehensively map gene interconnectivity, identifying region- and cell-type-specific drivers of biological processes associated with AD. Our analysis reveals profound modular heterogeneity in neurons and glia as well as extensive AD-related functional disruptions. Co-expression networks highlighted the extended involvement of astrocytes and microglia in biological processes beyond neuroinflammation, such as calcium homeostasis, glutamate regulation, lipid metabolism, vesicle-mediated transport, and TOR signaling. We find limited representation of DEGs within dysregulated pathways across neurons and glial cells, suggesting that differential gene expression alone may not adequately represent the disease complexity. Further dissection of inferred gene modules revealed distinct dynamics of hub DEGs in neurons versus glia, suggesting that DEGs exert more impact on neurons compared to glial cells in driving modular dysregulations underlying perturbed biological processes. Interestingly, we observe an overall downregulation of astrocyte and microglia modules across all brain regions in AD, indicating a prevailing trend of functional repression in glial cells across these regions. Notable genes from the CALM and HSP90 families emerged as hub genes across neuronal modules in all brain regions, suggesting conserved roles as drivers of synaptic dysfunction in AD. Our findings demonstrate the importance of an integrated, systems-oriented approach combining pathway and network analysis to comprehensively understand the cell-type-specific roles of genes in AD-related biological processes.
Collapse
|
12
|
Adeoye T, Shah SI, Ullah G. Systematic Analysis of Biological Processes Reveals Gene Co-expression Modules Driving Pathway Dysregulation in Alzheimer's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.15.585267. [PMID: 38559218 PMCID: PMC10980062 DOI: 10.1101/2024.03.15.585267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Alzheimer's disease (AD) manifests as a complex systems pathology with intricate interplay among various genes and biological processes. Traditional differential gene expression (DEG) analysis, while commonly employed to characterize AD-driven perturbations, does not sufficiently capture the full spectrum of underlying biological processes. Utilizing single-nucleus RNA-sequencing data from postmortem brain samples across key regions-middle temporal gyrus, superior frontal gyrus, and entorhinal cortex-we provide a comprehensive systematic analysis of disrupted processes in AD. We go beyond the DEG-centric analysis by integrating pathway activity analysis with weighted gene co-expression patterns to comprehensively map gene interconnectivity, identifying region- and cell-type-specific drivers of biological processes associated with AD. Our analysis reveals profound modular heterogeneity in neurons and glia as well as extensive AD-related functional disruptions. Co-expression networks highlighted the extended involvement of astrocytes and microglia in biological processes beyond neuroinflammation, such as calcium homeostasis, glutamate regulation, lipid metabolism, vesicle-mediated transport, and TOR signaling. We find limited representation of DEGs within dysregulated pathways across neurons and glial cells, indicating that differential gene expression alone may not adequately represent the disease complexity. Further dissection of inferred gene modules revealed distinct dynamics of hub DEGs in neurons versus glia, highlighting the differential impact of DEGs on neurons compared to glial cells in driving modular dysregulations underlying perturbed biological processes. Interestingly, we note an overall downregulation of both astrocyte and microglia modules in AD across all brain regions, suggesting a prevailing trend of functional repression in glial cells across these regions. Notable genes, including those of the CALM and HSP90 family genes emerged as hub genes across neuronal modules in all brain regions, indicating conserved roles as drivers of synaptic dysfunction in AD. Our findings demonstrate the importance of an integrated, systems-oriented approach combining pathway and network analysis for a comprehensive understanding of the cell-type-specific roles of genes in AD-related biological processes.
Collapse
Affiliation(s)
- Temitope Adeoye
- Department of Physics, University of South Florida, Tampa, FL 33620
| | - Syed I Shah
- Department of Physics, University of South Florida, Tampa, FL 33620
| | - Ghanim Ullah
- Department of Physics, University of South Florida, Tampa, FL 33620
| |
Collapse
|
13
|
Samy MVG, Perumal S. Systems pharmacology and multi-scale mechanism of Enicostema axillare bioactives in treating Alzheimer disease. Inflammopharmacology 2024; 32:575-593. [PMID: 37845599 DOI: 10.1007/s10787-023-01348-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 09/19/2023] [Indexed: 10/18/2023]
Abstract
As a progressive neurological disease with increased morbidity and mortality, Alzheimer Disease (AD) is characterized by neuron damage that controls memory and mental functions. Enicostema axillare (EA), an herb with a history of combativeness and effectiveness in treating Rheumatoid Arthritis, Cancer, and Diabetes, is used in Indian folk medicine from a holistic point of view. Though the herb is used for many illnesses, the molecular mechanism of its bioactive on AD has not been deciphered by intricate research. A unique pharmacology approach based on ADME drug screening and targeting, pathway enrichment (GO and KEGG), and network pharmacology, was established to explore the molecular mechanisms of E. axillare (EA) bioactive compounds for the treatment of AD. In brief, we bring to light the three active compounds of EA and seven potential molecular targets of AD, which are mainly implicated in four signaling pathways, i.e., MAPK, Apoptosis, neurodegeneration, and the TNF pathway. Moreover, the network analysis of the active compounds, molecular targets, and their pathways reveals the pharmacological nature of the compounds. Further, molecular docking studies were carried out to explore the interactions between the EA bioactive compounds and the targets and examine the binding affinity. The outcome of the work reflects the potential therapeutic effects of the compounds for treating AD through the modulation of the key proteins, which further corroborates the reliability of our network pharmacology analysis. This study not only helps in understanding the molecular mechanism of the drugs but also helps in finding and sorting new drugs for the treatment of AD, and other complex diseases through modern medicine.
Collapse
Affiliation(s)
| | - Sasidharan Perumal
- Cell and Molecular Biology Division, Biome Live Analytical Center, Karaikudi, Tamil Nadu, India.
| |
Collapse
|
14
|
Wang Z, Zhang D, Cheng C, Lin Z, Zhou D, Sun Y, Li W, Yan J, Luo S, Qian Z, Li Z, Huang G. Supplementation of Medium-Chain Triglycerides Combined with Docosahexaenoic Acid Inhibits Amyloid Beta Protein Deposition by Improving Brain Glucose Metabolism in APP/PS1 Mice. Nutrients 2023; 15:4244. [PMID: 37836528 PMCID: PMC10574179 DOI: 10.3390/nu15194244] [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: 08/17/2023] [Revised: 09/26/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
The deterioration of brain glucose metabolism predates the clinical onset of Alzheimer's disease (AD). Medium-chain triglycerides (MCTs) and docosahexaenoic acid (DHA) positively improve brain glucose metabolism and decrease the expression of AD-related proteins. However, the effects of the combined intervention are unclear. The present study explored the effects of the supplementation of MCTs combined with DHA in improving brain glucose metabolism and decreasing AD-related protein expression levels in APP/PS1 mice. The mice were assigned into four dietary treatment groups: the control group, MCTs group, DHA group, and MCTs + DHA group. The corresponding diet of the respective groups was fed to mice from the age of 3 to 11 months. The results showed that the supplementation of MCTs combined with DHA could increase serum octanoic acid (C8:0), decanoic acid (C10:0), DHA, and β-hydroxybutyrate (β-HB) levels; improve glucose metabolism; and reduce nerve cell apoptosis in the brain. Moreover, it also aided with decreasing the expression levels of amyloid beta protein (Aβ), amyloid precursor protein (APP), β-site APP cleaving enzyme-1 (BACE1), and presenilin-1 (PS1) in the brain. Furthermore, the supplementation of MCTs + DHA was significantly more beneficial than that of MCTs or DHA alone. In conclusion, the supplementation of MCTs combined with DHA could improve energy metabolism in the brain of APP/PS1 mice, thus decreasing nerve cell apoptosis and inhibiting the expression of Aβ.
Collapse
Affiliation(s)
- Zehao Wang
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin 300070, China; (Z.W.); (C.C.); (Z.L.); (D.Z.); (Y.S.); (W.L.); (S.L.)
| | - Dalong Zhang
- Department of Toxicology, Tianjin Centers for Disease Control and Prevention, Tianjin 300011, China; (D.Z.); (Z.Q.)
| | - Cheng Cheng
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin 300070, China; (Z.W.); (C.C.); (Z.L.); (D.Z.); (Y.S.); (W.L.); (S.L.)
| | - Zhenzhen Lin
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin 300070, China; (Z.W.); (C.C.); (Z.L.); (D.Z.); (Y.S.); (W.L.); (S.L.)
| | - Dezheng Zhou
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin 300070, China; (Z.W.); (C.C.); (Z.L.); (D.Z.); (Y.S.); (W.L.); (S.L.)
| | - Yue Sun
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin 300070, China; (Z.W.); (C.C.); (Z.L.); (D.Z.); (Y.S.); (W.L.); (S.L.)
| | - Wen Li
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin 300070, China; (Z.W.); (C.C.); (Z.L.); (D.Z.); (Y.S.); (W.L.); (S.L.)
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin 300070, China;
| | - Jing Yan
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin 300070, China;
- Department of Social Medicine and Health Administration, School of Public Health, Tianjin Medical University, Tianjin 300070, China
| | - Suhui Luo
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin 300070, China; (Z.W.); (C.C.); (Z.L.); (D.Z.); (Y.S.); (W.L.); (S.L.)
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin 300070, China;
| | - Zhiyong Qian
- Department of Toxicology, Tianjin Centers for Disease Control and Prevention, Tianjin 300011, China; (D.Z.); (Z.Q.)
| | - Zhenshu Li
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin 300070, China; (Z.W.); (C.C.); (Z.L.); (D.Z.); (Y.S.); (W.L.); (S.L.)
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin 300070, China;
| | - Guowei Huang
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin 300070, China; (Z.W.); (C.C.); (Z.L.); (D.Z.); (Y.S.); (W.L.); (S.L.)
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin 300070, China;
- Department of Critical Care Medicine and Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
15
|
Naletova I, Schmalhausen E, Tomasello B, Pozdyshev D, Attanasio F, Muronetz V. The role of sperm-specific glyceraldehyde-3-phosphate dehydrogenase in the development of pathologies-from asthenozoospermia to carcinogenesis. Front Mol Biosci 2023; 10:1256963. [PMID: 37711387 PMCID: PMC10499166 DOI: 10.3389/fmolb.2023.1256963] [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: 07/14/2023] [Accepted: 08/21/2023] [Indexed: 09/16/2023] Open
Abstract
The review considers various aspects of the influence of the glycolytic enzyme, sperm-specific glyceraldehyde-3-phosphate dehydrogenase (GAPDS) on the energy metabolism of spermatozoa and on the occurrence of several pathologies both in spermatozoa and in other cells. GAPDS is a unique enzyme normally found only in mammalian spermatozoa. GAPDS provides movement of the sperm flagellum through the ATP formation in glycolytic reactions. Oxidation of cysteine residues in GAPDS results in inactivation of the enzyme and decreases sperm motility. In particular, reduced sperm motility in diabetes can be associated with GAPDS oxidation by superoxide anion produced during glycation reactions. Mutations in GAPDS gene lead in the loss of motility, and in some cases, disrupts the formation of the structural elements of the sperm flagellum, in which the enzyme incorporates during spermiogenesis. GAPDS activation can be used to increase the spermatozoa fertility, and inhibitors of this enzyme are being tried as contraceptives. A truncated GAPDS lacking the N-terminal fragment of 72 amino acids that attaches the enzyme to the sperm flagellum was found in melanoma cell lines and then in specimens of melanoma and other tumors. Simultaneous production of the somatic form of GAPDH and sperm-specific GAPDS in cancer cells leads to a reorganization of their energy metabolism, which is accompanied by a change in the efficiency of metastasis of certain forms of cancer. Issues related to the use of GAPDS for the diagnosis of cancer, as well as the possibility of regulating the activity of this enzyme to prevent metastasis, are discussed.
Collapse
Affiliation(s)
- Irina Naletova
- Institute of Crystallography, National Council of Research, Catania, Italy
| | - Elena Schmalhausen
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Barbara Tomasello
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
| | - Denis Pozdyshev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | | | - Vladimir Muronetz
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
- Butlerov Chemical Institute, Kazan Federal University, Kazan, Russia
| |
Collapse
|
16
|
Tandon R, Zhao L, Watson CM, Elmor M, Heilman C, Sanders K, Hales CM, Yang H, Loring DW, Goldstein FC, Hanfelt JJ, Duong DM, Johnson EC, Wingo AP, Wingo TS, Roberts BR, Seyfried NT, Levey AI, Mitchell CS, Lah JJ. Predictors of Cognitive Decline in Healthy Middle-Aged Individuals with Asymptomatic Alzheimer's Disease. RESEARCH SQUARE 2023:rs.3.rs-2577025. [PMID: 36909654 PMCID: PMC10002814 DOI: 10.21203/rs.3.rs-2577025/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Alzheimer's disease (AD) progresses through a lengthy asymptomatic period during which pathological changes accumulate prior to development of clinical symptoms. As disease-modifying treatments are developed, tools to stratify risk of clinical disease will be required to guide their use. In this study, we examine the relationship of AD biomarkers in healthy middle-aged individuals to health history, family history, and neuropsychological measures and identify cerebrospinal fluid (CSF) biomarkers to stratify risk of progression from asymptomatic to symptomatic AD. CSF from cognitively normal (CN) individuals (N=1149) in the Emory Healthy Brain Study were assayed for Aβ42, total Tau (tTau), and phospho181-Tau (pTau), and a subset of 134 cognitively normal, but biomarker-positive, individuals were identified with asymptomatic AD (AsymAD) based on a locally-determined cutoff value for ratio of tTau to Aβ42. These AsymAD cases were matched for demographic features with 134 biomarker-negative controls (CN/BM-) and compared for differences in medical comorbidities and family history. Dyslipidemia emerged as a distinguishing feature between AsymAD and CN/BM-groups with significant association with personal and family history of dyslipidemia. A weaker relationship was seen with diabetes, but there was no association with hypertension. Examination of the full cohort by median regression revealed a significant relationship of CSF Aβ42 (but not tTau or pTau) with dyslipidemia and diabetes. On neuropsychological tests, CSF Aβ42 was not correlated with performance on any measures, but tTau and pTau were strongly correlated with visuospatial perception and visual episodic memory. In addition to traditional CSF AD biomarkers, a panel of AD biomarker peptides derived from integrating brain and CSF proteomes were evaluated using machine learning strategies to identify a set of 8 peptides that accurately classified CN/BM- and symptomatic AD CSF samples with AUC of 0.982. Using these 8 peptides in a low dimensional t-distributed Stochastic Neighbor Embedding analysis and k-Nearest Neighbor (k=5) algorithm, AsymAD cases were stratified into "Control-like" and "AD-like" subgroups based on their proximity to CN/BM- or AD CSF profiles. Independent analysis of these cases using a Joint Mutual Information algorithm selected a set of 5 peptides with 81% accuracy in stratifying cases into AD-like and Control-like subgroups. Performance of both sets of peptides was evaluated and validated in an independent data set from the Alzheimer's Disease Neuroimaging Initiative. Based on our findings, we conclude that there is an important role of lipid metabolism in asymptomatic stages of AD. Visuospatial perception and visual episodic memory may be more sensitive than language-based abilities to earliest stages of cognitive decline in AD. Finally, candidate CSF peptides show promise as next generation biomarkers for predicting progression from asymptomatic to symptomatic stages of AD.
Collapse
Affiliation(s)
- Raghav Tandon
- Department of Biomedical Engineering, Georgia Institute of Technology
- Center for Machine Learning, Georgia Institute of Technology
| | - Liping Zhao
- Department of Biostatistics and Bioinformatics, Emory School of Public Health
- Emory Goizueta Alzheimer’s Disease Research Center
| | - Caroline M. Watson
- Emory Goizueta Alzheimer’s Disease Research Center
- Department of Neurology, Emory School of Medicine
| | - Morgan Elmor
- Emory Goizueta Alzheimer’s Disease Research Center
- Department of Neurology, Emory School of Medicine
| | - Craig Heilman
- Emory Goizueta Alzheimer’s Disease Research Center
- Department of Neurology, Emory School of Medicine
| | - Katherine Sanders
- Emory Goizueta Alzheimer’s Disease Research Center
- Department of Neurology, Emory School of Medicine
| | - Chadwick M. Hales
- Emory Goizueta Alzheimer’s Disease Research Center
- Department of Neurology, Emory School of Medicine
- Center for Neurodegenerative Disease, Emory University
| | - Huiying Yang
- Department of Biostatistics and Bioinformatics, Emory School of Public Health
- Emory Goizueta Alzheimer’s Disease Research Center
| | - David W. Loring
- Emory Goizueta Alzheimer’s Disease Research Center
- Department of Neurology, Emory School of Medicine
| | - Felicia C. Goldstein
- Emory Goizueta Alzheimer’s Disease Research Center
- Department of Neurology, Emory School of Medicine
| | - John J. Hanfelt
- Department of Biostatistics and Bioinformatics, Emory School of Public Health
- Emory Goizueta Alzheimer’s Disease Research Center
| | - Duc M. Duong
- Emory Goizueta Alzheimer’s Disease Research Center
- Department of Neurology, Emory School of Medicine
- Department of Biochemistry, Emory School of Medicine
| | - Erik C.B. Johnson
- Emory Goizueta Alzheimer’s Disease Research Center
- Department of Neurology, Emory School of Medicine
- Center for Neurodegenerative Disease, Emory University
| | | | - Aliza P. Wingo
- Department of Psychiatry, Emory School of Medicine
- Division of Mental Health, Atlanta VA Medical Center, GA, USA
| | - Thomas S. Wingo
- Emory Goizueta Alzheimer’s Disease Research Center
- Department of Neurology, Emory School of Medicine
- Center for Neurodegenerative Disease, Emory University
| | - Blaine R. Roberts
- Center for Neurodegenerative Disease, Emory University
- Department of Biochemistry, Emory School of Medicine
| | - Nicholas T. Seyfried
- Emory Goizueta Alzheimer’s Disease Research Center
- Center for Neurodegenerative Disease, Emory University
- Department of Biochemistry, Emory School of Medicine
| | - Allan I. Levey
- Emory Goizueta Alzheimer’s Disease Research Center
- Department of Neurology, Emory School of Medicine
- Center for Neurodegenerative Disease, Emory University
| | - Cassie S. Mitchell
- Department of Biomedical Engineering, Georgia Institute of Technology
- Center for Machine Learning, Georgia Institute of Technology
| | - James J. Lah
- Emory Goizueta Alzheimer’s Disease Research Center
- Department of Neurology, Emory School of Medicine
- Center for Neurodegenerative Disease, Emory University
| |
Collapse
|
17
|
Lazarev VF, Dutysheva EA, Kanunikov IE, Guzhova IV, Margulis BA. Protein Interactome of Amyloid-β as a Therapeutic Target. Pharmaceuticals (Basel) 2023; 16:312. [PMID: 37259455 PMCID: PMC9965366 DOI: 10.3390/ph16020312] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/27/2023] [Accepted: 02/08/2023] [Indexed: 04/12/2024] Open
Abstract
The amyloid concept of Alzheimer's disease (AD) assumes the β-amyloid peptide (Aβ) as the main pathogenic factor, which injures neural and other brain cells, causing their malfunction and death. Although Aβ has been documented to exert its cytotoxic effect in a solitary manner, there is much evidence to claim that its toxicity can be modulated by other proteins. The list of such Aβ co-factors or interactors includes tau, APOE, transthyretin, and others. These molecules interact with the peptide and affect the ability of Aβ to form oligomers or aggregates, modulating its toxicity. Thus, the list of potential substances able to reduce the harmful effects of the peptide should include ones that can prevent the pathogenic interactions by specifically binding Aβ and/or its partners. In the present review, we discuss the data on Aβ-based complexes in AD pathogenesis and on the compounds directly targeting Aβ or the destructors of its complexes with other polypeptides.
Collapse
Affiliation(s)
- Vladimir F. Lazarev
- Institute of Cytology of the Russian Academy of Sciences, 194064 Saint Petersburg, Russia
| | - Elizaveta A. Dutysheva
- Institute of Cytology of the Russian Academy of Sciences, 194064 Saint Petersburg, Russia
| | - Igor E. Kanunikov
- Biological Faculty, St. Petersburg State University, 199034 Saint Petersburg, Russia
| | - Irina V. Guzhova
- Institute of Cytology of the Russian Academy of Sciences, 194064 Saint Petersburg, Russia
| | - Boris A. Margulis
- Institute of Cytology of the Russian Academy of Sciences, 194064 Saint Petersburg, Russia
| |
Collapse
|
18
|
Ahmad I, Singh R, Pal S, Prajapati S, Sachan N, Laiq Y, Husain H. Exploring the Role of Glycolytic Enzymes PFKFB3 and GAPDH in the Modulation of Aβ and Neurodegeneration and Their Potential of Therapeutic Targets in Alzheimer's Disease. Appl Biochem Biotechnol 2023:10.1007/s12010-023-04340-0. [PMID: 36692648 DOI: 10.1007/s12010-023-04340-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/10/2023] [Indexed: 01/25/2023]
Abstract
Alzheimer's disease (AD) is presently the 6th major cause of mortality across the globe. However, it is expected to rise rapidly, following cancer and heart disease, as a leading cause of death among the elderly peoples. AD is largely characterized by metabolic changes linked to glucose metabolism and age-induced mitochondrial failure. Recent research suggests that the glycolytic pathway is required for a range of neuronal functions in the brain including synaptic transmission, energy production, and redox balance; however, alteration in glycolytic pathways may play a significant role in the development of AD. Moreover, it is hypothesized that targeting the key enzymes involved in glucose metabolism may help to prevent or reduce the risk of neurodegenerative disorders. One of the major pro-glycolytic enzyme is 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase-3 (PFKFB3); it is normally absent in neurons but abundant in astrocytes. Similarly, another key of glycolysis is glyceraldehyde-3-phosphate dehydrogenase (GAPDH) which catalyzes the conversion of aldolase and glyceraldehyde 3 phosphates to 1,3 bisphosphoglycerate. GAPDH has been reported to interact with various neurodegenerative disease-associated proteins, including the amyloid-β protein precursor (AβPP). These findings indicate PFKFB3 and GAPDH as a promising therapeutic target to AD. Current review highlight the contributions of PFKFB3 and GAPDH in the modulation of Aβand AD pathogenesis and further explore the potential of PFKFB3 and GAPDH as therapeutic targets in AD.
Collapse
Affiliation(s)
- Imran Ahmad
- Department of Biochemistry, King George's Medical University, Lucknow, 226003, Uttar Pradesh, India.
| | - Ranjana Singh
- Department of Biochemistry, King George's Medical University, Lucknow, 226003, Uttar Pradesh, India.
| | - Saurabh Pal
- Department of Biotechnology, Era's Lucknow Medical College & Hospital, Era University, Lucknow, 226003, Uttar Pradesh, India
| | - Soni Prajapati
- Department of Biochemistry, King George's Medical University, Lucknow, 226003, Uttar Pradesh, India
| | - Nidhi Sachan
- Cell and Neurobiology Laboratory, Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| | - Yusra Laiq
- Department of Biochemistry, King George's Medical University, Lucknow, 226003, Uttar Pradesh, India
| | - Hadiya Husain
- Department of Zoology, University of Lucknow, Lucknow, 226007, Uttar Pradesh, India
| |
Collapse
|
19
|
Ma S, Xia T, Wang X, Wang H. Identification and validation of biomarkers based on cellular senescence in mild cognitive impairment. Front Aging Neurosci 2023; 15:1139789. [PMID: 37187578 PMCID: PMC10176455 DOI: 10.3389/fnagi.2023.1139789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 03/30/2023] [Indexed: 05/17/2023] Open
Abstract
Background Mild cognitive impairment (MCI), a syndrome defined as decline of cognitive function greater than expected for an individual's age and education level, occurs in up to 22.7% of elderly patients in United States, causing the heavy psychological and economic burdens to families and society. Cellular senescence (CS) is a stress response that accompanies permanent cell-cycle arrest, which has been reported to be a fundamental pathological mechanism of many age-related diseases. This study aims to explore biomarkers and potential therapeutic targets in MCI based on CS. Methods The mRNA expression profiles of peripheral blood samples from patients in MCI and non-MCI group were download from gene expression omnibus (GEO) database (GSE63060 for training and GSE18309 for external validation), CS-related genes were obtained from CellAge database. Weighted gene co-expression network analysis (WGCNA) was conducted to discover the key relationships behind the co-expression modules. The differentially expressed CS-related genes would be obtained through overlapping among the above datasets. Then, pathway and GO enrichment analyses were performed to further elucidate the mechanism of MCI. The protein-protein interaction network was used to extract hub genes and the logistic regression was performed to distinguish the MCI patients from controls. The hub gene-drug network, hub gene-miRNA network as well as transcription factor-gene regulatory network were used to analyze potential therapeutic targets for MCI. Results Eight CS-related genes were identified as key gene signatures in MCI group, which were mainly enriched in the regulation of response to DNA damage stimulus, Sin3 complex and transcription corepressor activity. The receiver operating characteristic curves of logistic regression diagnostic model were constructed and presented great diagnostic value in both training and validation set. Conclusion Eight CS-related hub genes - SMARCA4, GAPDH, SMARCB1, RUNX1, SRC, TRIM28, TXN, and PRPF19 - serve as candidate biomarkers for MCI and display the excellent diagnostic value. Furthermore, we also provide a theoretical basis for targeted therapy against MCI through the above hub genes.
Collapse
Affiliation(s)
- Songmei Ma
- Department of Anesthesiology, The Third Central Clinical College of Tianjin Medical University, Tianjin, China
- Department of Anesthesiology, The First People’s Hospital of Shangqiu, Shangqiu, Henan, China
| | - Tong Xia
- Department of Anesthesiology, The Third Central Clinical College of Tianjin Medical University, Tianjin, China
| | - Xinyi Wang
- Department of Anesthesiology, The Third Central Clinical College of Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
- Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Haiyun Wang
- Department of Anesthesiology, The Third Central Clinical College of Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
- Tianjin Institute of Hepatobiliary Disease, Tianjin, China
- *Correspondence: Haiyun Wang,
| |
Collapse
|
20
|
Li Y, Lan X, Wang S, Cui Y, Song S, Zhou H, Li Q, Dai L, Zhang J. Serial five-membered lactone ring ions in the treatment of Alzheimer's diseases-comprehensive profiling of arctigenin metabolites and network analysis. Front Pharmacol 2022; 13:1065654. [PMID: 36605392 PMCID: PMC9807626 DOI: 10.3389/fphar.2022.1065654] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 12/01/2022] [Indexed: 12/24/2022] Open
Abstract
Arctigenin is a phenylpropanoid dibenzylbutyro lactone lignan compound with multiple biological functions. Previous studies have shown that arctigenin have neuroprotective effects in Alzheimer's disease (AD) models both in vivo and in vitro; however, its metabolism in vivo has not been studied. Most traditional analytical methods only partially characterize drug metabolite prototypes, so there is an urgent need for a research strategy that can fully characterize drug metabolites. In the present study, ions fishing with a serial five-membered lactone ring as a fishhook strategy based on ultrahigh-performance liquid chromatography-Q-Exactive Orbitrap mass spectrometry (UHPLC-Q-Exactive Orbitrap MS) was utilised to characterise the metabolism of arctigenin, and the establishment of this strategy also solved the challenge of creating a comprehensive metabolic profile of neolignan. Based on the proposed strategy, a total of 105 metabolites were detected and characterised, 76 metabolites of which were found in rats and 49 metabolites in liver microsomes. These metabolites were postulated to be produced through oxidation, reduction, hydrolysis, and complex reactions. Subsequently, network pharmacology was utilized to elucidate the mechanism of arctigenin and its main metabolites against Alzheimer's disease, screening 381 potential targets and 20 major signaling pathways. The study on the comprehensive metabolism of arctigenin provides a holistic metabolic profile, which will help to better understand the mechanism of arctigenin in the treatment of Alzheimer's disease (AD) and also provide a basis for the safe administration of arctigenin.
Collapse
Affiliation(s)
- Yanan Li
- School of Pharmacy, Binzhou Medical University, Yantai, China,School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xianming Lan
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Shaoping Wang
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Yifang Cui
- School of Pharmacy, Binzhou Medical University, Yantai, China,School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shuyi Song
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Hongyan Zhou
- School of Pharmacy, Binzhou Medical University, Yantai, China,School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qiyan Li
- Shandong Provincial Institute for Food and Drug Control, Jinan, China,*Correspondence: Jiayu Zhang, ; Long Dai, ; Qiyan Li,
| | - Long Dai
- School of Pharmacy, Binzhou Medical University, Yantai, China,*Correspondence: Jiayu Zhang, ; Long Dai, ; Qiyan Li,
| | - Jiayu Zhang
- School of Pharmacy, Binzhou Medical University, Yantai, China,*Correspondence: Jiayu Zhang, ; Long Dai, ; Qiyan Li,
| |
Collapse
|
21
|
Hyslop PA, Chaney MO. Mechanism of GAPDH Redox Signaling by H 2O 2 Activation of a Two-Cysteine Switch. Int J Mol Sci 2022; 23:4604. [PMID: 35562998 PMCID: PMC9102624 DOI: 10.3390/ijms23094604] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 02/06/2023] Open
Abstract
Oxidation of glyceraldehyde-3-phosphate dehydrogenase (GAPDH) by reactive oxygen species such as H2O2 activate pleiotropic signaling pathways is associated with pathophysiological cell fate decisions. Oxidized GAPDH binds chaperone proteins with translocation of the complex to the nucleus and mitochondria initiating autophagy and cellular apoptosis. In this study, we establish the mechanism by which H2O2-oxidized GAPDH subunits undergo a subunit conformational rearrangement. H2O2 oxidizes both the catalytic cysteine and a vicinal cysteine (four residues downstream) to their respective sulfenic acids. A 'two-cysteine switch' is activated, whereby the sulfenic acids irreversibly condense to an intrachain thiosulfinic ester resulting in a major metastable subunit conformational rearrangement. All four subunits of the homotetramer are uniformly and independently oxidized by H2O2, and the oxidized homotetramer is stabilized at low temperatures. Over time, subunits unfold forming disulfide-linked aggregates with the catalytic cysteine oxidized to a sulfinic acid, resulting from thiosulfinic ester hydrolysis via the highly reactive thiosulfonic ester intermediate. Molecular Dynamic Simulations provide additional mechanistic insights linking GAPDH subunit oxidation with generating a putative signaling conformer. The low-temperature stability of the H2O2-oxidized subunit conformer provides an operable framework to study mechanisms associated with gain-of-function activities of oxidized GAPDH to identify novel targets for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Paul A. Hyslop
- Arkley Research Labs, Arkley BioTek, LLC, 4444 Decatur Blvd., Indianapolis, IN 46241, USA
| | - Michael O. Chaney
- Eli Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN 46285, USA;
| |
Collapse
|
22
|
Canarelli SE, Swalm BM, Larson ET, Morrison MJ, Weerapana E. Monitoring GAPDH activity and inhibition with cysteine-reactive chemical probes. RSC Chem Biol 2022; 3:972-982. [PMID: 35866162 PMCID: PMC9257626 DOI: 10.1039/d2cb00091a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/30/2022] [Indexed: 11/21/2022] Open
Abstract
Glyceraldehyde 3-phosphate dehydrogenase (GAPDH) is a central enzyme in glycolysis that regulates the Warburg effect in cancer cells. In addition to its role in metabolism, GAPDH is also implicated in diverse cellular processes, including transcription and apoptosis. Dysregulated GAPDH activity is associated with a variety of pathologies, and GAPDH inhibitors have demonstrated therapeutic potential as anticancer and immunomodulatory agents. Given the critical role of GAPDH in pathophysiology, it is important to have access to tools that enable rapid monitoring of GAPDH activity and inhibition within a complex biological system. Here, we report an electrophilic peptide-based probe, SEC1, which covalently modifies the active-site cysteine, C152, of GAPDH to directly report on GAPDH activity within a proteome. We demonstrate the utility of SEC1 to assess changes in GAPDH activity in response to oncogenic transformation, reactive oxygen species (ROS) and small-molecule GAPDH inhibitors, including Koningic acid (KA). We then further evaluated KA, to determine the detailed mechanism of inhibition. Our mechanistic studies confirm that KA is a highly effective irreversible inhibitor of GAPDH, which acts through a NAD+-uncompetitive and G3P-competitive mechanism. Proteome-wide evaluation of the cysteine targets of KA demonstrated high selectivity for the active-site cysteine of GAPDH over other reactive cysteines within the proteome. Lastly, the therapeutic potential of KA was investigated in an autoimmune model, where treatment with KA resulted in decreased cytokine production by Th1 effector cells. Together, these studies describe methods to evaluate GAPDH activity and inhibition within a proteome, and report on the high potency and selectivity of KA as an irreversible inhibitor of GAPDH. Cysteine-reactive chemical probes can covalently modify the active-site cysteine of GAPDH.![]()
Collapse
Affiliation(s)
- Sarah E. Canarelli
- Department of Chemistry, Boston College, Chestnut Hill, Massachusetts 02467, USA
| | | | - Eric T. Larson
- Rheos Medicines, Inc, Cambridge, Massachusetts 02142, USA
| | | | - Eranthie Weerapana
- Department of Chemistry, Boston College, Chestnut Hill, Massachusetts 02467, USA
| |
Collapse
|
23
|
Liang L, Yan J, Huang X, Zou C, Chen L, Li R, Xie J, Pan M, Zou D, Liu Y. Identification of molecular signatures associated with sleep disorder and Alzheimer's disease. Front Psychiatry 2022; 13:925012. [PMID: 35990086 PMCID: PMC9386361 DOI: 10.3389/fpsyt.2022.925012] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 07/20/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) and sleep disorders are both neurodegenerative conditions characterized by impaired or absent sleep. However, potential common pathogenetic mechanisms of these diseases are not well characterized. METHODS Differentially expressed genes (DEGs) were identified using publicly available human gene expression profiles GSE5281 for AD and GSE40562 for sleep disorder. DEGs common to the two datasets were used for enrichment analysis, and we performed multi-scale embedded gene co-expression network analysis (MEGENA) for common DEGs. Fast gene set enrichment analysis (fGSEA) was used to obtain common pathways, while gene set variation analysis (GSVA) was applied to quantify those pathways. Subsequently, we extracted the common genes between module genes identified by MEGENA and genes of the common pathways, and we constructed protein-protein interaction (PPI) networks. The top 10 genes with the highest degree of connectivity were classified as hub genes. Common genes were used to perform Metascape enrichment analysis for functional enrichment. Furthermore, we quantified infiltrating immune cells in patients with AD or sleep disorder and in controls. RESULTS DEGs common to the two disorders were involved in the citrate cycle and the HIF-1 signaling pathway, and several common DEGs were related to signaling pathways regulating the pluripotency of stem cells, as well as 10 other pathways. Using MEGENA, we identified 29 modules and 1,498 module genes in GSE5281, and 55 modules and 1,791 module genes in GSE40562. Hub genes involved in AD and sleep disorder were ATP5A1, ATP5B, COX5A, GAPDH, NDUFA9, NDUFS3, NDUFV2, SOD1, UQCRC1, and UQCRC2. Plasmacytoid dendritic cells and T helper 17 cells had the most extensive infiltration in both AD and sleep disorder. CONCLUSION AD pathology and pathways of neurodegeneration participate in processes contributing in AD and sleep disorder. Hub genes may be worth exploring as potential candidates for targeted therapy of AD and sleep disorder.
Collapse
Affiliation(s)
- Lucong Liang
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jing Yan
- Department of Geriatrics, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiaohua Huang
- Department of Neurology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Chun Zou
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Liechun Chen
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Rongjie Li
- Department of Geriatrics, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, China.,Department of Geriatrics, The First People's Hospital of Nanning, Nanning, China
| | - Jieqiong Xie
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Mika Pan
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Donghua Zou
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ying Liu
- Department of Geriatrics, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, China.,Department of Geriatrics, The First People's Hospital of Nanning, Nanning, China
| |
Collapse
|
24
|
Muronetz VI, Medvedeva MV, Sevostyanova IA, Schmalhausen EV. Modification of Glyceraldehyde-3-Phosphate Dehydrogenase with Nitric Oxide: Role in Signal Transduction and Development of Apoptosis. Biomolecules 2021; 11:1656. [PMID: 34827652 PMCID: PMC8615796 DOI: 10.3390/biom11111656] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/01/2021] [Accepted: 11/04/2021] [Indexed: 01/07/2023] Open
Abstract
This review focuses on the consequences of GAPDH S-nitrosylation at the catalytic cysteine residue. The widespread hypothesis according to which S-nitrosylation causes a change in GAPDH structure and its subsequent binding to the Siah1 protein is considered in detail. It is assumed that the GAPDH complex with Siah1 is transported to the nucleus by carrier proteins, interacts with nuclear proteins, and induces apoptosis. However, there are several conflicting and unproven elements in this hypothesis. In particular, there is no direct confirmation of the interaction between the tetrameric GAPDH and Siah1 caused by S-nitrosylation of GAPDH. The question remains as to whether the translocation of GAPDH into the nucleus is caused by S-nitrosylation or by some other modification of the catalytic cysteine residue. The hypothesis of the induction of apoptosis by oxidation of GAPDH is considered. This oxidation leads to a release of the coenzyme NAD+ from the active center of GAPDH, followed by the dissociation of the tetramer into subunits, which move to the nucleus due to passive transport and induce apoptosis. In conclusion, the main tasks are summarized, the solutions to which will make it possible to more definitively establish the role of nitric oxide in the induction of apoptosis.
Collapse
Affiliation(s)
- Vladimir I. Muronetz
- Belozersky Institute of Physico Chemical Biology, Lomonosov Moscow State University, 119234 Moscow, Russia; (I.A.S.); (E.V.S.)
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia;
| | - Maria V. Medvedeva
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia;
| | - Irina A. Sevostyanova
- Belozersky Institute of Physico Chemical Biology, Lomonosov Moscow State University, 119234 Moscow, Russia; (I.A.S.); (E.V.S.)
| | - Elena V. Schmalhausen
- Belozersky Institute of Physico Chemical Biology, Lomonosov Moscow State University, 119234 Moscow, Russia; (I.A.S.); (E.V.S.)
| |
Collapse
|