1
|
Zito A, Martinelli A, Masiero M, Akhmedov M, Kwee I. NPM: latent batch effects correction of omics data by nearest-pair matching. BIOINFORMATICS (OXFORD, ENGLAND) 2025; 41:btaf084. [PMID: 39999010 PMCID: PMC11925496 DOI: 10.1093/bioinformatics/btaf084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 12/18/2024] [Accepted: 02/21/2025] [Indexed: 02/27/2025]
Abstract
MOTIVATION Batch effects (BEs) are a predominant source of noise in omics data and often mask real biological signals. BEs remain common in existing datasets. Current methods for BE correction mostly rely on specific assumptions or complex models, and may not detect and adjust BEs adequately, impacting downstream analysis and discovery power. To address these challenges we developed NPM, a nearest-neighbor matching-based method that adjusts BEs and may outperform other methods in a wide range of datasets. RESULTS We assessed distinct metrics and graphical readouts, and compared our method to commonly used BE correction methods. NPM demonstrates the ability in correcting for BEs, while preserving biological differences. It may outperform other methods based on multiple metrics. Altogether, NPM proves to be a valuable BE correction approach to maximize discovery in biomedical research, with applicability in clinical research where latent BEs are often dominant. AVAILABILITY AND IMPLEMENTATION NPM is freely available on GitHub (https://github.com/bigomics/NPM) and on Omics Playground (https://bigomics.ch/omics-playground). Computer codes for analyses are available at (https://github.com/bigomics/NPM). The datasets underlying this article are the following: GSE120099, GSE82177, GSE162760, GSE171343, GSE153380, GSE163214, GSE182440, GSE163857, GSE117970, GSE173078, and GSE10846. All these datasets are publicly available and can be freely accessed on the Gene Expression Omnibus repository.
Collapse
Affiliation(s)
- Antonino Zito
- BigOmics Analytics, Via Serafino Balestra 12, Lugano 6900, Switzerland
| | - Axel Martinelli
- BigOmics Analytics, Via Serafino Balestra 12, Lugano 6900, Switzerland
| | - Mauro Masiero
- BigOmics Analytics, Via Serafino Balestra 12, Lugano 6900, Switzerland
| | | | - Ivo Kwee
- BigOmics Analytics, Via Serafino Balestra 12, Lugano 6900, Switzerland
| |
Collapse
|
2
|
Zhu S, Wang P, Hu J. m6A-modified lncRNA GAS5 promotes M1-polarization of microglia in alcohol use disorder. Brain Res Bull 2025; 221:111215. [PMID: 39828042 DOI: 10.1016/j.brainresbull.2025.111215] [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: 11/01/2024] [Revised: 01/02/2025] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
Long noncoding RNA (lncRNA) are essential for modulating the onset and progression of alcohol use disorder (AUD). In this study, we investigated the molecular pathways through which lncRNA may contribute to AUD development. We assessed the expression levels of long noncoding RNA GAS5 (lncRNA GAS5) and microRNA-136-5p (miR-136-5p) in AUD tissue samples and cell lines using reverse transcription-quantitative polymerase chain reaction. Detection of GAS5 N6-methyladenosine (m6A) modifications, facilitated by alkylation repair homolog 5 (ALKBH5), was performed using RNA immunoprecipitation and RNA pull-down assays. The effect of GAS5 on the functionality of SH-SY5Y cells was evaluated using CCK-8 and Transwell assays. Our findings showed high levels of GAS5 expression in both AUD tissues and cell lines. Overexpression of GAS5 decreased the migratory capability of SH-SY5Y cells, whereas silencing GAS5 increased this ability. Bioinformatics analyses predicted a relationship between expression levels of miR-136-5p and GAS5, which was subsequently confirmed using dual-luciferase reporter assays. Additionally, we discovered that GAS5 acts as a sponge for miR-136-5p, leading to the upregulation of ATF2. Elevated levels of ATF2 are associated with M1 microglial polarization. In summary, m6A-modified GAS5 may influence the M1 polarization of microglia via the miR-136-5p/ATF2 pathway. Statistical evaluations were performed using GraphPad Prism V8.0, employing the student's t-test for comparisons between two groups, assuming a normal distribution and equal variances. When variances were unequal, but normality was maintained, the corrected Student's t-test was applied. The non-parametric Wilcoxon rank-sum test was used to analyze non-normally distributed data, and one-way ANOVA was used to compare three or more groups. Independent replication was ensured in the studies, with each experiment repeated at least three times and statistical significance was set at P < 0.05.
Collapse
Affiliation(s)
- Shuang Zhu
- Mental Health Center, the First Affiliated Hospital of Harbin Medical University, Heilongjiang, China
| | - Peng Wang
- Binzhou People's Hospital of Breast Surgery, Shandong, China
| | - Jian Hu
- Mental Health Center, the First Affiliated Hospital of Harbin Medical University, Heilongjiang, China.
| |
Collapse
|
3
|
Mohd Nazri AK, Yahya N, Khan DM, Mohd Radzi NZ, Badruddin N, Abdul Latiff AH, Abdulaal MJ. Partial directed coherence analysis of resting-state EEG signals for alcohol use disorder detection using machine learning. Front Neurosci 2025; 18:1524513. [PMID: 39867451 PMCID: PMC11757881 DOI: 10.3389/fnins.2024.1524513] [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: 11/08/2024] [Accepted: 12/23/2024] [Indexed: 01/28/2025] Open
Abstract
Introduction Excessive alcohol consumption negatively impacts physical and psychiatric health, lifestyle, and societal interactions. Chronic alcohol abuse alters brain structure, leading to alcohol use disorder (AUD), a condition requiring early diagnosis for effective management. Current diagnostic methods, primarily reliant on subjective questionnaires, could benefit from objective measures. Method The study proposes a novel EEG-based classification approach, focusing on effective connectivity (EC) derived from resting-state EEG signals in combination with support vector machine (SVM) algorithms. EC estimation is performed using the partial directed coherence (PDC) technique. The analysis is conducted on an EEG dataset comprising 35 individuals with AUD and 35 healthy controls (HCs). The methodology evaluates the efficacy of connectivity features in distinguishing between AUD and HC and subsequently develops and assesses an EEG classification technique using EC matrices and SVM. Result The proposed methodology demonstrated promising performance, achieving a peak accuracy of 94.5% and an area under the curve (AUC) of 0.988, specifically using frequency bands 29, 36, 45, 46, and 52. Additionally, feature reduction techniques applied to the PDC adjacency matrices in the gamma band further improved classification outcomes. The SVM-based classification achieved an accuracy of 96.37 ± 0.45%, showcasing enhanced performance through the utilization of reduced PDC adjacency matrices. Discussion These results highlight the potential of the developed algorithm as a robust diagnostic tool for AUD detection, enhancing precision beyond subjective methods. Incorporating EC features derived from EEG signals can inform tailored treatment strategies, contributing to improved management of AUD.
Collapse
Affiliation(s)
| | - Norashikin Yahya
- Centre for Intelligent Signal and Imaging Research (CISIR), Electrical and Electronic Engineering Department, Universiti Teknologi PETRONAS, Bandar Seri Iskandar, Perak, Malaysia
| | - Danish M. Khan
- Department of Data Science and Artificial Intelligence, School of Engineering and Technology, Sunway University, Petaling Jaya, Selangor, Malaysia
| | - Noor'Izni Zafirah Mohd Radzi
- Centre for Intelligent Signal and Imaging Research (CISIR), Electrical and Electronic Engineering Department, Universiti Teknologi PETRONAS, Bandar Seri Iskandar, Perak, Malaysia
| | - Nasreen Badruddin
- Centre for Intelligent Signal and Imaging Research (CISIR), Electrical and Electronic Engineering Department, Universiti Teknologi PETRONAS, Bandar Seri Iskandar, Perak, Malaysia
| | - Abdul Halim Abdul Latiff
- Centre for Subsurface Imaging, Department of Geosciences, Universiti Teknologi PETRONAS, Bandar Seri Iskandar, Perak, Malaysia
| | - Mohammed J. Abdulaal
- Center of Excellence in Intelligent Engineering Systems (CEIES), Department of Electrical and Computer Engineering, Faculty of Engineering, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
4
|
Ngo AL, Ahmad CM, Gharavi Alkhansari N, Nguyen L, Zhang H. Epigenetic Insights into Substance Use Disorder and Associated Psychiatric Conditions. Complex Psychiatry 2025; 11:12-36. [PMID: 40201238 PMCID: PMC11975344 DOI: 10.1159/000544912] [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: 09/11/2024] [Accepted: 02/19/2025] [Indexed: 04/10/2025] Open
Abstract
Background Substance use disorder (SUD) is closely associated with epigenetic modifications that significantly impact mental health outcomes. Alcohol and drug misuse induce widespread changes in the epigenome and transcriptome of the central nervous system, disrupting critical processes such as reward signaling and emotional regulation. These alterations in epigenetic regulation and gene expression often persist even after substance cessation, potentially contributing to the onset or worsening of psychiatric conditions, including schizophrenia, depression, stress, and anxiety. Summary This review delves into key epigenetic mechanisms underlying SUD and its comorbid psychiatric disorders, with a focus on DNA methylation, histone modifications, and noncoding RNA regulation. Additionally, it examines the influence of environmental and biological factors on the epigenome and evaluates emerging epigenetic-based therapeutic strategies aimed at treating SUD and related psychiatric conditions. Key Messages Gaining a deeper understanding of the epigenetic mechanisms driving SUD and its associated psychiatric disorders is crucial for the development of effective therapeutic interventions. This review highlights the potential of epigenetic-based pharmacological strategies to mitigate the societal and personal burdens linked to SUD and its mental health complications.
Collapse
Affiliation(s)
- Ambrose Loc Ngo
- College of Medicine, Kansas City University, Kansas City, MO, USA
| | | | | | - Linda Nguyen
- College of Pharmacy, Western University, Pomona, CA, USA
| | - Huiping Zhang
- Department of Psychiatry, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Section of Biomedical Genetics, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| |
Collapse
|
5
|
Pritha AN, Pasmay AA, Noor S. Recent Advances in the Role of Non-coding RNAs in Fetal Alcohol Spectrum Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1473:129-155. [PMID: 40128478 DOI: 10.1007/978-3-031-81908-7_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
Despite numerous preclinical studies modeling fetal alcohol spectrum disorder (FASD)-associated neurodevelopmental deficits to date, a comprehensive molecular landscape dictating these deficits remains poorly understood. Non-coding RNAs constitute a substantial layer of epigenetic regulation of gene expression at the transcriptional, post-transcriptional, translational, and post-translational levels. Yet, little is known about the differential expression of non-coding RNAs in the context of prenatal alcohol exposure (PAE) that are mechanistically linked with FASD-related neurobehavior deficits. This chapter reviews our current knowledge from preclinical studies in non-coding RNA-mediated molecular mechanisms that may underlie FASD pathophysiology. This chapter also summarizes relevant clinical evidence and current efforts in utilizing these non-coding RNA molecules as biomarkers of PAE-associated deficits impacting central nervous system (CNS) function. Unraveling the diverse roles of various species of non-coding RNAs is critical to enhancing our comprehension of these intricate molecular pathways. Understanding these pathways would likely contribute to identifying critical molecular target(s) for developing efficient treatment strategies and prognostic and diagnostic markers fostering advancements in treating and managing FASD-related CNS dysfunction.
Collapse
Affiliation(s)
- Ariana N Pritha
- Department of Neurosciences and New Mexico Alcohol Research Center, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Andrea A Pasmay
- Department of Neurosciences and New Mexico Alcohol Research Center, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Shahani Noor
- Department of Neurosciences and New Mexico Alcohol Research Center, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA.
| |
Collapse
|
6
|
Rosoff DB, Wagner J, Bell AS, Mavromatis LA, Jung J, Lohoff FW. A multi-omics Mendelian randomization study identifies new therapeutic targets for alcohol use disorder and problem drinking. Nat Hum Behav 2025; 9:188-207. [PMID: 39528761 DOI: 10.1038/s41562-024-02040-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 10/01/2024] [Indexed: 11/16/2024]
Abstract
Integrating proteomic and transcriptomic data with genetic architectures of problematic alcohol use and alcohol consumption behaviours can advance our understanding and help identify therapeutic targets. We conducted systematic screens using genome-wise association study data from ~3,500 cortical proteins (N = 722) and ~6,100 genes in 8 canonical brain cell types (N = 192) with 4 alcohol-related outcomes (N ≤ 537,349), identifying 217 cortical proteins and 255 cell-type genes associated with these behaviours, with 36 proteins and 37 cell-type genes being new. Although there was limited overlap between proteome and transcriptome targets, downstream neuroimaging revealed shared neurophysiological pathways. Colocalization with independent genome-wise association study data further prioritized 16 proteins, including CAB39L and NRBP1, and 12 cell-type genes, implicating mechanisms such as mTOR signalling. In addition, genes such as SAMHD1, VIPAS39, NUP160 and INO80E were identified as having favourable neuropsychiatric profiles. These findings provide insights into the genetic landscapes governing problematic alcohol use and alcohol consumption behaviours, highlighting promising therapeutic targets for future research.
Collapse
Affiliation(s)
- Daniel B Rosoff
- Section on Clinical Genomics and Experimental Therapeutics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
- NIH Oxford-Cambridge Scholars Program, National Institutes of Health, Bethesda, MD, USA
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Josephin Wagner
- Section on Clinical Genomics and Experimental Therapeutics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Andrew S Bell
- Section on Clinical Genomics and Experimental Therapeutics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Lucas A Mavromatis
- Section on Clinical Genomics and Experimental Therapeutics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Jeesun Jung
- Section on Clinical Genomics and Experimental Therapeutics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Falk W Lohoff
- Section on Clinical Genomics and Experimental Therapeutics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
7
|
Zhang X. Highly effective batch effect correction method for RNA-seq count data. Comput Struct Biotechnol J 2024; 27:58-64. [PMID: 39802213 PMCID: PMC11718288 DOI: 10.1016/j.csbj.2024.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/13/2024] [Accepted: 12/14/2024] [Indexed: 01/16/2025] Open
Abstract
RNA sequencing (RNA-seq) has become a cornerstone of transcriptomics, providing detailed insights into gene expression across diverse biological conditions and sample types. However, RNA-seq data are often confounded by batch effects, systematic non-biological variations that compromise data reliability and obscure true biological differences. To address these challenges, we introduce ComBat-ref, a refined batch effect correction method designed to enhance the statistical power and reliability of differential expression analysis in RNA-seq data. Building on the principles of ComBat-seq, ComBat-ref employs a negative binomial model for count data adjustment but innovates by selecting a reference batch with the smallest dispersion, preserving count data for the reference batch, and adjusting other batches towards the reference batch. Our method demonstrated superior performance in both simulated environments and real-world datasets, including the growth factor receptor network (GFRN) data and NASA GeneLab transcriptomic datasets, significantly improving sensitivity and specificity compared to existing methods. By effectively mitigating batch effects while maintaining high detection power, ComBat-ref provides a robust solution for improving the accuracy and interpretability of RNA-seq data analyses.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- Department of Computer Science and Information Science, California State University San Marcos, 333 S. Twin Oaks Valley Rd, San Marcos, CA 92096, USA
| |
Collapse
|
8
|
Airapetov MI, Eresko SO, Shamaeva SA, Bychkov ER, Lebedev AA, Shabanov PD. Study of Neuroinflammation in the Rat Hippocampus during Ethanol Exposure and Pharmacological Correction with Azithromycin: New Data and Future Perspectives. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1911-1921. [PMID: 39647820 DOI: 10.1134/s0006297924110051] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 07/07/2024] [Accepted: 10/09/2024] [Indexed: 12/10/2024]
Abstract
With prolonged ethanol ingestion, disturbances in the emotional spectrum develop, and memory problems are noted. These symptoms could be mediated by the development of neurochemical changes in the hippocampus of the brain. Although there is evidence that hippocampus is vulnerable to chronic alcohol intoxication and that neuroinflammation and neurodegeneration develop in this brain region, the key molecular mechanisms have not been identified. The aim of the study was to investigate changes in the immune system in the periphery as well as in the hippocampus of rat brain during ethanol exposure and during pharmacological correction with azithromycin (AZM). Long-term ethanol exposure was modeled by injecting rats with a 20% ethanol solution (4 g/kg) for 4 weeks. General biochemical and clinical blood analysis was performed in animals. Expression levels of the cytokine genes (Il1β, Ccl2, Il6, Il11, Il13, Tnfα, Tgfβ), Toll-like receptor system genes (Tlr3, Tl4, Tlr7, Nfkb1, Hmgb1), and TLR system-related microRNA molecules (miR-182, miR-155-5p, miR-96-5p, miR-let-7b) were evaluated in the hippocampus. IL-1β protein content was also assessed in the hippocampus. Prolonged exposure to alcohol caused increase in the mRNA and protein levels of IL-1β, and decrease in the mRNA levels of Tnfα, Il11, Tlr3, and Tlr7. The contents of miRlet7b, miR96, and miR155 were downregulated in the hippocampus after long-term alcohol exposure. Elevated levels of THE Il1β mRNA and protein and Hmgb1 mRNA were maintained under conditions of ethanol abstinence. The Tlr3 mRNA levels were decreased after abstinence. Administration of AZM reduced the IL1β, TLR3, and HMGB1 mRNA levels under conditions of ethanol abstinence; and at higher doses of the drug decrease in the IL-1β protein levels in the hippocampus of rat brain was observed. Thus, the study provided new insights into the mechanisms of neuroinflammation in the hippocampus during prolonged exposure to ethanol and upon abstinence. The obtained results allowed us to suggest a number of tasks for further studies in this direction.
Collapse
Affiliation(s)
- Marat I Airapetov
- Institute of Experimental Medicine, St. Petersburg, 197376, Russia.
- Military medical academy of S. M. Kirov, St. Petersburg, 194044, Russia
| | - Sergei O Eresko
- Institute of Experimental Medicine, St. Petersburg, 197376, Russia
- Saint-Petersburg National Research University of Information Technologies, Mechanics and Optics, St. Petersburg, 197022, Russia
| | | | | | - Andrei A Lebedev
- Institute of Experimental Medicine, St. Petersburg, 197376, Russia
| | - Petr D Shabanov
- Institute of Experimental Medicine, St. Petersburg, 197376, Russia
- Military medical academy of S. M. Kirov, St. Petersburg, 194044, Russia
| |
Collapse
|
9
|
Besong OTO, Koo JS, Zhang H. Brain lncRNA-mRNA co-expression regulatory networks and alcohol use disorder. Genomics 2024; 116:110928. [PMID: 39197730 PMCID: PMC11531342 DOI: 10.1016/j.ygeno.2024.110928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 08/08/2024] [Accepted: 08/24/2024] [Indexed: 09/01/2024]
Abstract
Prolonged alcohol consumption can disturb the expression of both coding and noncoding genes in the brain. These dysregulated genes may co-express in modules and interact within networks, consequently influencing the susceptibility to developing alcohol use disorder (AUD). In the present study, we performed an RNA-seq analysis of the expression of both long noncoding RNAs (lncRNAs) and messenger RNAs (mRNAs) in 192 postmortem tissue samples collected from eight brain regions (amygdala, caudate nucleus, cerebellum, hippocampus, nucleus accumbens, prefrontal cortex, putamen, and ventral tegmental area) of 12 AUD and 12 control subjects of European ancestry. Applying the limma-voom method, we detected a total of 57 lncRNAs and 51 mRNAs exhibiting significant differential expression (Padj < 0.05 and fold-change ≥2) across at least one of the eight brain regions investigated. Machine learning analysis further confirmed the potential of these top genes in predicting AUD. Through Weighted Gene Co-expression Network Analysis (WGCNA), we identified distinct lncRNA-mRNA co-expression modules associated with AUD in each of the eight brain regions. Additionally, lncRNA-mRNA co-expression networks were constructed for each brain region using Cytoscape to reveal gene regulatory interactions implicated in AUD. Hub genes within these networks were found to be enriched in several key KEGG pathways, including Axon Guidance, MAPK Signaling, p53 Signaling, Adherens Junction, and Neurodegeneration. Our results underscore the significance of networks involving AUD-associated lncRNAs and mRNAs in modulating neuroplasticity in response to alcohol exposure. Further elucidating these molecular mechanisms holds promise for the development of targeted therapeutic interventions for AUD.
Collapse
Affiliation(s)
- Ojong Tabi Ojong Besong
- Department of Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Section of Biomedical Genetics, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Ji Sun Koo
- Department of Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Section of Biomedical Genetics, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Huiping Zhang
- Department of Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Section of Biomedical Genetics, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
| |
Collapse
|
10
|
Sprang M, Möllmann J, Andrade-Navarro MA, Fontaine JF. Overlooked poor-quality patient samples in sequencing data impair reproducibility of published clinically relevant datasets. Genome Biol 2024; 25:222. [PMID: 39152483 PMCID: PMC11328481 DOI: 10.1186/s13059-024-03331-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 07/08/2024] [Indexed: 08/19/2024] Open
Abstract
BACKGROUND Reproducibility is a major concern in biomedical studies, and existing publication guidelines do not solve the problem. Batch effects and quality imbalances between groups of biological samples are major factors hampering reproducibility. Yet, the latter is rarely considered in the scientific literature. RESULTS Our analysis uses 40 clinically relevant RNA-seq datasets to quantify the impact of quality imbalance between groups of samples on the reproducibility of gene expression studies. High-quality imbalance is frequent (14 datasets; 35%), and hundreds of quality markers are present in more than 50% of the datasets. Enrichment analysis suggests common stress-driven effects among the low-quality samples and highlights a complementary role of transcription factors and miRNAs to regulate stress response. Preliminary ChIP-seq results show similar trends. Quality imbalance has an impact on the number of differential genes derived by comparing control to disease samples (the higher the imbalance, the higher the number of genes), on the proportion of quality markers in top differential genes (the higher the imbalance, the higher the proportion; up to 22%) and on the proportion of known disease genes in top differential genes (the higher the imbalance, the lower the proportion). We show that removing outliers based on their quality score improves the resulting downstream analysis. CONCLUSIONS Thanks to a stringent selection of well-designed datasets, we demonstrate that quality imbalance between groups of samples can significantly reduce the relevance of differential genes, consequently reducing reproducibility between studies. Appropriate experimental design and analysis methods can substantially reduce the problem.
Collapse
Affiliation(s)
- Maximilian Sprang
- Faculty of Biology, Johannes Gutenberg-Universität Mainz, Biozentrum I, Hans-Dieter-Hüsch-Weg 15, Mainz, 55128, Germany
| | - Jannik Möllmann
- Faculty of Biology, Johannes Gutenberg-Universität Mainz, Biozentrum I, Hans-Dieter-Hüsch-Weg 15, Mainz, 55128, Germany
| | - Miguel A Andrade-Navarro
- Faculty of Biology, Johannes Gutenberg-Universität Mainz, Biozentrum I, Hans-Dieter-Hüsch-Weg 15, Mainz, 55128, Germany.
| | - Jean-Fred Fontaine
- Faculty of Biology, Johannes Gutenberg-Universität Mainz, Biozentrum I, Hans-Dieter-Hüsch-Weg 15, Mainz, 55128, Germany
- Central Institute for Decision Support Systems in Crop Protection (ZEPP), Rüdesheimer Str. 60-68, Bad Kreuznach, 55545, Germany
| |
Collapse
|
11
|
Wang JQ, Liang J, Wang JL, Shan F, Cao Y, Zhou X, Yan CY, Xia QR, Liu YR. Evaluation of plasma-derived extracellular vesicles miRNAs and their connection with hippocampal mRNAs in alcohol use disorder. Life Sci 2024; 351:122820. [PMID: 38857652 DOI: 10.1016/j.lfs.2024.122820] [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: 02/01/2024] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/12/2024]
Abstract
Alcohol use disorder (AUD) is a common mental illness with high morbidity and disability. The discovery of laboratory biomarkers has progressed slowly, resulting in suboptimal diagnosis and treatment of AUD. This study aimed to identify promising biomarkers, as well as the potential miRNA-mRNA networks associated with AUD pathogenesis. RNA sequencing was performed on plasma-derived small extracellular vesicles (sEVs) from AUD patients and healthy controls (HCs) to harvest miRNAs expression profiles. Machine learning (ML) models were built to screen characteristic miRNAs, whose target mRNAs were analyzed using TargetScan, miRanda and miRDB databases. Gene Expression Omnibus (GEO) datasets (GSE181804 and GSE180722) providing postmortem hippocampal gene expression profiles of AUD subjects were mined. A total of 247 differentially expressed (DE) plasma-derived sEVs miRNAs and 122 DE hippocampal mRNAs were obtained. Then, 22 overlapping sEVs miRNAs with high importance scores were gained by intersecting 5 ML models. As a result, we established a putative sEVs miRNA-hippocampal mRNA network that can effectively distinguish AUD patients from HCs. In conclusion, we proposed 5 AUD-representative sEVs miRNAs (hsa-miR-144-5p, hsa-miR-182-5p, hsa-miR-142-5p, hsa-miR-7-5p, and hsa-miR-15b-5p) that may participate in the pathogenesis of AUD by modulating downstream target hippocampal genes. These findings may provide novel insights into the diagnosis and treatment of AUD.
Collapse
Affiliation(s)
- Jie-Quan Wang
- Department of Pharmacy, Affiliated Psychological Hospital of Anhui Medical University, Hefei 230000, China; Psychopharmacology Research Laboratory, Anhui Mental Health Center, Hefei 230000, China; Department of Pharmacy, Hefei Fourth People's Hospital, Hefei 230000, China; Anhui Clinical Research Center for Mental Disorders, Hefei 230000, China
| | - Jun Liang
- Department of Pharmacy, Affiliated Psychological Hospital of Anhui Medical University, Hefei 230000, China; Psychopharmacology Research Laboratory, Anhui Mental Health Center, Hefei 230000, China; Department of Pharmacy, Hefei Fourth People's Hospital, Hefei 230000, China; Anhui Clinical Research Center for Mental Disorders, Hefei 230000, China
| | - Jin-Liang Wang
- Department of Pharmacy, Affiliated Psychological Hospital of Anhui Medical University, Hefei 230000, China; Psychopharmacology Research Laboratory, Anhui Mental Health Center, Hefei 230000, China; Department of Pharmacy, Hefei Fourth People's Hospital, Hefei 230000, China; Anhui Clinical Research Center for Mental Disorders, Hefei 230000, China
| | - Feng Shan
- Department of Pharmacy, Affiliated Psychological Hospital of Anhui Medical University, Hefei 230000, China; Psychopharmacology Research Laboratory, Anhui Mental Health Center, Hefei 230000, China; Department of Pharmacy, Hefei Fourth People's Hospital, Hefei 230000, China; Anhui Clinical Research Center for Mental Disorders, Hefei 230000, China
| | - Yin Cao
- Department of Pharmacy, Affiliated Psychological Hospital of Anhui Medical University, Hefei 230000, China; Psychopharmacology Research Laboratory, Anhui Mental Health Center, Hefei 230000, China; Department of Pharmacy, Hefei Fourth People's Hospital, Hefei 230000, China; Anhui Clinical Research Center for Mental Disorders, Hefei 230000, China
| | - Xuan Zhou
- Department of Pharmacy, Affiliated Psychological Hospital of Anhui Medical University, Hefei 230000, China; Psychopharmacology Research Laboratory, Anhui Mental Health Center, Hefei 230000, China; Department of Pharmacy, Hefei Fourth People's Hospital, Hefei 230000, China; Anhui Clinical Research Center for Mental Disorders, Hefei 230000, China
| | - Chun-Yu Yan
- Department of Pharmacy, Affiliated Psychological Hospital of Anhui Medical University, Hefei 230000, China; Psychopharmacology Research Laboratory, Anhui Mental Health Center, Hefei 230000, China; Department of Pharmacy, Hefei Fourth People's Hospital, Hefei 230000, China; Anhui Clinical Research Center for Mental Disorders, Hefei 230000, China
| | - Qing-Rong Xia
- Department of Pharmacy, Affiliated Psychological Hospital of Anhui Medical University, Hefei 230000, China; Psychopharmacology Research Laboratory, Anhui Mental Health Center, Hefei 230000, China; Department of Pharmacy, Hefei Fourth People's Hospital, Hefei 230000, China; Anhui Clinical Research Center for Mental Disorders, Hefei 230000, China.
| | - Ya-Ru Liu
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei 230001, China; The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei 230001, China.
| |
Collapse
|
12
|
Nohesara S, Mostafavi Abdolmaleky H, Thiagalingam S. Substance-Induced Psychiatric Disorders, Epigenetic and Microbiome Alterations, and Potential for Therapeutic Interventions. Brain Sci 2024; 14:769. [PMID: 39199463 PMCID: PMC11352452 DOI: 10.3390/brainsci14080769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 07/18/2024] [Accepted: 07/25/2024] [Indexed: 09/01/2024] Open
Abstract
Substance use disorders (SUDs) are complex biopsychosocial diseases that cause neurocognitive deficits and neurological impairments by altering the gene expression in reward-related brain areas. Repeated drug use gives rise to alterations in DNA methylation, histone modifications, and the expression of microRNAs in several brain areas that may be associated with the development of psychotic symptoms. The first section of this review discusses how substance use contributes to the development of psychotic symptoms via epigenetic alterations. Then, we present more evidence about the link between SUDs and brain epigenetic alterations. The next section presents associations between paternal and maternal exposure to substances and epigenetic alterations in the brains of offspring and the role of maternal diet in preventing substance-induced neurological impairments. Then, we introduce potential therapeutic agents/approaches such as methyl-rich diets to modify epigenetic alterations for alleviating psychotic symptoms or depression in SUDs. Next, we discuss how substance use-gut microbiome interactions contribute to the development of neurological impairments through epigenetic alterations and how gut microbiome-derived metabolites may become new therapeutics for normalizing epigenetic aberrations. Finally, we address possible challenges and future perspectives for alleviating psychotic symptoms and depression in patients with SUDs by modulating diets, the epigenome, and gut microbiome.
Collapse
Affiliation(s)
- Shabnam Nohesara
- Department of Medicine (Biomedical Genetics), Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA;
- Mental Health Research Center, Psychosocial Health Research Institute, Department of Psychiatry, School of Medicine, Iran University of Medical Sciences, Tehran 14535, Iran
| | - Hamid Mostafavi Abdolmaleky
- Department of Medicine (Biomedical Genetics), Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA;
- Nutrition/Metabolism Laboratory, Department of Surgery, BIDMC, Harvard Medical School, Boston, MA 02215, USA
| | - Sam Thiagalingam
- Department of Medicine (Biomedical Genetics), Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA;
- Department of Pathology & Laboratory Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
13
|
Du Y, Li R, Fu D, Zhang B, Cui A, Shao Y, Lai Z, Chen R, Chen B, Wang Z, Zhang W, Chu L. Multi-omics technologies and molecular biomarkers in brain tumor-related epilepsy. CNS Neurosci Ther 2024; 30:e14717. [PMID: 38641945 PMCID: PMC11031674 DOI: 10.1111/cns.14717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 03/04/2024] [Accepted: 03/29/2024] [Indexed: 04/21/2024] Open
Abstract
BACKGROUND Brain tumors are one of the leading causes of epilepsy, and brain tumor-related epilepsy (BTRE) is recognized as the major cause of intractable epilepsy, resulting in huge treatment cost and burden to patients, their families, and society. Although optimal treatment regimens are available, the majority of patients with BTRE show poor resolution of symptoms. BTRE has a very complex and multifactorial etiology, which includes several influencing factors such as genetic and molecular biomarkers. Advances in multi-omics technologies have enabled to elucidate the pathophysiological mechanisms and related biomarkers of BTRE. Here, we reviewed multi-omics technology-based research studies on BTRE published in the last few decades and discussed the present status, development, opportunities, challenges, and prospects in treating BTRE. METHODS First, we provided a general review of epilepsy, BTRE, and multi-omics techniques. Next, we described the specific multi-omics (including genomics, transcriptomics, epigenomics, proteomics, and metabolomics) techniques and related molecular biomarkers for BTRE. We then presented the associated pathogenetic mechanisms of BTRE. Finally, we discussed the development and application of novel omics techniques for diagnosing and treating BTRE. RESULTS Genomics studies have shown that the BRAF gene plays a role in BTRE development. Furthermore, the BRAF V600E variant was found to induce epileptogenesis in the neuronal cell lineage and tumorigenesis in the glial cell lineage. Several genomics studies have linked IDH variants with glioma-related epilepsy, and the overproduction of D2HG is considered to play a role in neuronal excitation that leads to seizure occurrence. The high expression level of Forkhead Box O4 (FOXO4) was associated with a reduced risk of epilepsy occurrence. In transcriptomics studies, VLGR1 was noted as a biomarker of epileptic onset in patients. Several miRNAs such as miR-128 and miRNA-196b participate in BTRE development. miR-128 might be negatively associated with the possibility of tumor-related epilepsy development. The lncRNA UBE2R2-AS1 inhibits the growth and invasion of glioma cells and promotes apoptosis. Quantitative proteomics has been used to determine dynamic changes of protein acetylation in epileptic and non-epileptic gliomas. In another proteomics study, a high expression of AQP-4 was detected in the brain of GBM patients with seizures. By using quantitative RT-PCR and immunohistochemistry assay, a study revealed that patients with astrocytomas and oligoastrocytomas showed high BCL2A1 expression and poor seizure control. By performing immunohistochemistry, several studies have reported the relationship between D2HG overproduction and seizure occurrence. Ki-67 overexpression in WHO grade II gliomas was found to be associated with poor postoperative seizure control. According to metabolomics research, the PI3K/AKT/mTOR pathway is associated with the development of glioma-related epileptogenesis. Another metabolomics study found that SV2A, P-gb, and CAD65/67 have the potential to function as biomarkers for BTRE. CONCLUSIONS Based on the synthesized information, this review provided new research perspectives and insights into the early diagnosis, etiological factors, and personalized treatment of BTRE.
Collapse
Affiliation(s)
- Yaoqiang Du
- Laboratory Medicine Center, Department of Transfusion MedicineZhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical CollegeHangzhouChina
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouChina
| | - Rusong Li
- The Second School of Clinical MedicineZhejiang Chinese Medical UniversityHangzhouChina
| | - Danqing Fu
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouChina
| | - Biqin Zhang
- Cancer Center, Department of HematologyZhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical CollegeHangzhouChina
| | - Ailin Cui
- Cancer Center, Department of Ultrasound MedicineZhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical CollegeHangzhouChina
| | - Yutian Shao
- Zhejiang BioAsia Life Science InstitutePinghuChina
| | - Zeyu Lai
- The Second School of Clinical MedicineZhejiang Chinese Medical UniversityHangzhouChina
| | - Rongrong Chen
- School of Clinical MedicineHangzhou Normal UniversityHangzhouChina
| | - Bingyu Chen
- Laboratory Medicine Center, Department of Transfusion MedicineZhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical CollegeHangzhouChina
| | - Zhen Wang
- Laboratory Medicine Center, Department of Transfusion MedicineZhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical CollegeHangzhouChina
| | - Wei Zhang
- The Second School of Clinical MedicineZhejiang Chinese Medical UniversityHangzhouChina
| | - Lisheng Chu
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouChina
- Department of PhysiologyZhejiang Chinese Medical UniversityHangzhouChina
| |
Collapse
|
14
|
Jiang S, Liu B, Lin K, Li L, Li R, Tan S, Zhang X, Jiang L, Ni H, Wang Y, Ding H, Hu J, Qian H, Ge R. Impacted spike frequency adaptation associated with reduction of KCNQ2/3 exacerbates seizure activity in temporal lobe epilepsy. Hippocampus 2024; 34:58-72. [PMID: 38049972 DOI: 10.1002/hipo.23587] [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: 01/09/2022] [Revised: 09/21/2023] [Accepted: 10/28/2023] [Indexed: 12/06/2023]
Abstract
Numerous epilepsy-related genes have been identified in recent decades by unbiased genome-wide screens. However, the available druggable targets for temporal lobe epilepsy (TLE) remain limited. Furthermore, a substantial pool of candidate genes potentially applicable to TLE therapy awaits further validation. In this study, we reveal the significant role of KCNQ2 and KCNQ3, two M-type potassium channel genes, in the onset of seizures in TLE. Our investigation began with a quantitative analysis of two publicly available TLE patient databases to establish a correlation between seizure onset and the downregulated expression of KCNQ2/3. We then replicated these pathological changes in a pilocarpine seizure mouse model and observed a decrease in spike frequency adaptation due to the affected M-currents in dentate gyrus granule neurons. In addition, we performed a small-scale simulation of the dentate gyrus network and confirmed that the impaired spike frequency adaptation of granule cells facilitated epileptiform activity throughout the network. This, in turn, resulted in prolonged seizure duration and reduced interictal intervals. Our findings shed light on an underlying mechanism contributing to ictogenesis in the TLE hippocampus and suggest a promising target for the development of antiepileptic drugs.
Collapse
Affiliation(s)
- Shicheng Jiang
- Department of Pathophysiology, Bengbu Medical College, Bengbu, Anhui, China
- Laboratory of Brain and Psychiatric Disease, Bengbu Medical College, Bengbu, Anhui, China
| | - Bei Liu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Kaiwen Lin
- Department of Pathophysiology, Bengbu Medical College, Bengbu, Anhui, China
- Laboratory of Brain and Psychiatric Disease, Bengbu Medical College, Bengbu, Anhui, China
| | - Lianjun Li
- Department of Pathophysiology, Bengbu Medical College, Bengbu, Anhui, China
- Laboratory of Brain and Psychiatric Disease, Bengbu Medical College, Bengbu, Anhui, China
| | - Rongrong Li
- Department of Pathophysiology, Bengbu Medical College, Bengbu, Anhui, China
- Laboratory of Brain and Psychiatric Disease, Bengbu Medical College, Bengbu, Anhui, China
| | - Shuo Tan
- Department of Pathophysiology, Bengbu Medical College, Bengbu, Anhui, China
- Laboratory of Brain and Psychiatric Disease, Bengbu Medical College, Bengbu, Anhui, China
| | - Xinyu Zhang
- Department of Pathophysiology, Bengbu Medical College, Bengbu, Anhui, China
- Laboratory of Brain and Psychiatric Disease, Bengbu Medical College, Bengbu, Anhui, China
| | - Lei Jiang
- Department of General Surgery, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Hong Ni
- Department of Pathophysiology, Bengbu Medical College, Bengbu, Anhui, China
- Laboratory of Brain and Psychiatric Disease, Bengbu Medical College, Bengbu, Anhui, China
| | - Yuanyuan Wang
- Department of Pathophysiology, Bengbu Medical College, Bengbu, Anhui, China
- Laboratory of Brain and Psychiatric Disease, Bengbu Medical College, Bengbu, Anhui, China
| | - Haihu Ding
- Department of Pathophysiology, Bengbu Medical College, Bengbu, Anhui, China
- Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Bengbu Medical College, Bengbu, Anhui, China
| | - Jing Hu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Hao Qian
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Rongjing Ge
- Department of Pathophysiology, Bengbu Medical College, Bengbu, Anhui, China
- Laboratory of Brain and Psychiatric Disease, Bengbu Medical College, Bengbu, Anhui, China
- Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Bengbu Medical College, Bengbu, Anhui, China
| |
Collapse
|
15
|
Occhipinti C, La Russa R, Iacoponi N, Lazzari J, Costantino A, Di Fazio N, Del Duca F, Maiese A, Fineschi V. miRNAs and Substances Abuse: Clinical and Forensic Pathological Implications: A Systematic Review. Int J Mol Sci 2023; 24:17122. [PMID: 38069445 PMCID: PMC10707252 DOI: 10.3390/ijms242317122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/17/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
Substance addiction is a chronic and relapsing brain disorder characterized by compulsive seeking and continued substance use, despite adverse consequences. The high prevalence and social burden of addiction are indisputable; however, the available intervention is insufficient. The modulation of gene expression and aberrant adaptation of neural networks are attributed to the changes in brain functions under repeated exposure to addictive substances. Considerable studies have demonstrated that miRNAs are strong modulators of post-transcriptional gene expression in substance addiction. The emerging role of microRNA (miRNA) provides new insights into many biological and pathological processes in the central nervous system: their variable expression in different regions of the brain and tissues may play a key role in regulating the pathophysiological events of addiction. This work provides an overview of the current literature on miRNAs involved in addiction, evaluating their impaired expression and regulatory role in neuroadaptation and synaptic plasticity. Clinical implications of such modulatory capacities will be estimated. Specifically, it will evaluate the potential diagnostic role of miRNAs in the various stages of drug and substance addiction. Future perspectives about miRNAs as potential novel therapeutic targets for substance addiction and abuse will also be provided.
Collapse
Affiliation(s)
- Carla Occhipinti
- Department of Surgical Pathology, Medical, Molecular and Critical Area, Institute of Legal Medicine, University of Pisa, 56126 Pisa, Italy; (C.O.); (N.I.); (J.L.); (A.C.)
| | - Raffaele La Russa
- Department of Clinical Medicine, Public Health, Life Sciences, and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
| | - Naomi Iacoponi
- Department of Surgical Pathology, Medical, Molecular and Critical Area, Institute of Legal Medicine, University of Pisa, 56126 Pisa, Italy; (C.O.); (N.I.); (J.L.); (A.C.)
| | - Julia Lazzari
- Department of Surgical Pathology, Medical, Molecular and Critical Area, Institute of Legal Medicine, University of Pisa, 56126 Pisa, Italy; (C.O.); (N.I.); (J.L.); (A.C.)
| | - Andrea Costantino
- Department of Surgical Pathology, Medical, Molecular and Critical Area, Institute of Legal Medicine, University of Pisa, 56126 Pisa, Italy; (C.O.); (N.I.); (J.L.); (A.C.)
| | - Nicola Di Fazio
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00161 Rome, Italy; (N.D.F.); (F.D.D.); (V.F.)
| | - Fabio Del Duca
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00161 Rome, Italy; (N.D.F.); (F.D.D.); (V.F.)
| | - Aniello Maiese
- Department of Surgical Pathology, Medical, Molecular and Critical Area, Institute of Legal Medicine, University of Pisa, 56126 Pisa, Italy; (C.O.); (N.I.); (J.L.); (A.C.)
| | - Vittorio Fineschi
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00161 Rome, Italy; (N.D.F.); (F.D.D.); (V.F.)
| |
Collapse
|
16
|
Airapetov MI, Eresko SO, Shamaeva SA, Matveev NM, Bychkov ER, Lebedev AA, Shabanov PD. Prolonged alcohol consumption influences microRNA expression in the nucleus accumbens of the rat brain. BIOMEDITSINSKAIA KHIMIIA 2023; 69:235-239. [PMID: 37705484 DOI: 10.18097/pbmc20236904235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
The microRNA (miR) species analyzed in this study are involved in molecular mechanisms of TLR4 and TLR7 signaling, mediating the development of neuroinflammation and neurodegeneration. We have investigated the expression levels of miR-let7b, miR-96, miR-182, miR-155, and the mRNA content of HMGB1, TLR3, TLR4 in the nucleus accumbens (NAc) of the brain of rats exposed to long-term alcoholization. The long-term alcoholization caused a decrease in miR-let7b, miR-96, miR-182, and TLR7 mRNA levels; this was accompanied by an increase in miR-155, TLR4, and Hmgb1 mRNA levels in the NAc of rat brain. TLR7 is functionally linked to miR-let7b. The data of a simultaneous decrease in miR-let7b and TLR7 mRNA are of interest for further studies; they may indicate on the lack of functionally significant links between Hmgb1 and the miR-let7b-TLR7 system in NAc. The existing evidence of a functional relationship between TLR4 with miR-155 and miR-182 and our observations on their expression changes during chronic alcoholization are very interesting and require further investigation. The suggestion about the development of neuroinflammatory process in NAc under prolonged alcohol exposure are relevant for studying the level of TLR gene expression in NAc, as well as the expression of miR species, which may have a functional relationship with the TLR system.
Collapse
Affiliation(s)
- M I Airapetov
- Institute of Experimental Medicine, St. Petersburg, Russia; Military Medical Academy of S.M. Kirov, St. Petersburg, Russia
| | - S O Eresko
- Institute of Experimental Medicine, St. Petersburg, Russia; North-Western State Medical University named after I.I. Mechnikov, St. Petersburg, Russia
| | - S A Shamaeva
- Institute of Experimental Medicine, St. Petersburg, Russia
| | - N M Matveev
- Institute of Experimental Medicine, St. Petersburg, Russia
| | - E R Bychkov
- Institute of Experimental Medicine, St. Petersburg, Russia
| | - A A Lebedev
- Institute of Experimental Medicine, St. Petersburg, Russia
| | - P D Shabanov
- Institute of Experimental Medicine, St. Petersburg, Russia
| |
Collapse
|
17
|
Cheng J, Ho WK, Wu BT, Liu HP, Lin WY. miRNA profiling as a complementary diagnostic tool for amyotrophic lateral sclerosis. Sci Rep 2023; 13:13805. [PMID: 37612427 PMCID: PMC10447559 DOI: 10.1038/s41598-023-40879-y] [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: 05/25/2023] [Accepted: 08/17/2023] [Indexed: 08/25/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS), the most prevalent motor neuron disease characterized by its complex genetic structure, lacks a single diagnostic test capable of providing a conclusive diagnosis. In order to demonstrate the potential for genetic diagnosis and shed light on the pathogenic role of miRNAs in ALS, we developed an ALS diagnostic rule by training the model using 80% of a miRNA profiling dataset consisting of 253 ALS samples and 103 control samples. Subsequently, we validated the diagnostic rule using the remaining 20% of unseen samples. The diagnostic rule we developed includes miR-205-5p, miR-206, miR-376a-5p, miR-412-5p, miR-3927-3p, miR-4701-3p, miR-6763-5p, and miR-6801-3p. Remarkably, the rule achieved an 82% true positive rate and a 73% true negative rate when predicting the unseen samples. Furthermore, the identified miRNAs target 21 genes in the PI3K-Akt pathway and 27 genes in the ALS pathway, including notable genes such as BCL2, NEFH, and OPTN. We propose that miRNA profiling may serve as a complementary diagnostic tool to supplement the clinical presentation and aid in the early recognition of ALS.
Collapse
Affiliation(s)
- Jack Cheng
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan
- Department of Medical Research, China Medical University Hospital, Taichung, 40447, Taiwan
| | - Wen-Kuang Ho
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan
| | - Bor-Tsang Wu
- Department of Senior Citizen Service Management, National Taichung University of Science and Technology, Taichung City, 40343, Taiwan
| | - Hsin-Ping Liu
- Graduate Institute of Acupuncture Science, College of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan.
| | - Wei-Yong Lin
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan.
- Department of Medical Research, China Medical University Hospital, Taichung, 40447, Taiwan.
| |
Collapse
|
18
|
Kumaresan V, Lim Y, Juneja P, Tipton AE, de Guglielmo G, Carrette LLG, Kallupi M, Maturin L, Liu Y, George O, Zhang H. Abstinence from Escalation of Cocaine Intake Changes the microRNA Landscape in the Cortico-Accumbal Pathway. Biomedicines 2023; 11:1368. [PMID: 37239038 PMCID: PMC10216163 DOI: 10.3390/biomedicines11051368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/25/2023] [Accepted: 04/30/2023] [Indexed: 05/28/2023] Open
Abstract
Cocaine administration alters the microRNA (miRNA) landscape in the cortico-accumbal pathway. These changes in miRNA can play a major role in the posttranscriptional regulation of gene expression during withdrawal. This study aimed to investigate the changes in microRNA expression in the cortico-accumbal pathway during acute withdrawal and protracted abstinence following escalated cocaine intake. Small RNA sequencing (sRNA-seq) was used to profile miRNA transcriptomic changes in the cortico-accumbal pathway [infralimbic- and prelimbic-prefrontal cortex (IL and PL) and nucleus accumbens (NAc)] of rats with extended access to cocaine self-administration followed by an 18-h withdrawal or a 4-week abstinence. An 18-h withdrawal led to differential expression (fold-change > 1.5 and p < 0.05) of 21 miRNAs in the IL, 18 miRNAs in the PL, and two miRNAs in the NAc. The mRNAs potentially targeted by these miRNAs were enriched in the following pathways: gap junctions, neurotrophin signaling, MAPK signaling, and cocaine addiction. Moreover, a 4-week abstinence led to differential expression (fold-change > 1.5 and p < 0.05) of 23 miRNAs in the IL, seven in the PL, and five miRNAs in the NAc. The mRNAs potentially targeted by these miRNAs were enriched in pathways including gap junctions, cocaine addiction, MAPK signaling, glutamatergic synapse, morphine addiction, and amphetamine addiction. Additionally, the expression levels of several miRNAs differentially expressed in either the IL or the NAc were significantly correlated with addiction behaviors. Our findings highlight the impact of acute and protracted abstinence from escalated cocaine intake on miRNA expression in the cortico-accumbal pathway, a key circuit in addiction, and suggest developing novel biomarkers and therapeutic approaches to prevent relapse by targeting abstinence-associated miRNAs and their regulated mRNAs.
Collapse
Affiliation(s)
- Vidhya Kumaresan
- Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA;
| | - Yolpanhchana Lim
- Department of Psychiatry, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA; (Y.L.); (P.J.); (Y.L.)
- Department of Medicine (Biomedical Genetics), Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA
| | - Poorva Juneja
- Department of Psychiatry, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA; (Y.L.); (P.J.); (Y.L.)
- Department of Medicine (Biomedical Genetics), Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA
| | - Allison E. Tipton
- Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA;
| | - Giordano de Guglielmo
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA; (G.d.G.); (L.L.G.C.); (M.K.); (L.M.); (O.G.)
| | - Lieselot L. G. Carrette
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA; (G.d.G.); (L.L.G.C.); (M.K.); (L.M.); (O.G.)
| | - Marsida Kallupi
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA; (G.d.G.); (L.L.G.C.); (M.K.); (L.M.); (O.G.)
| | - Lisa Maturin
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA; (G.d.G.); (L.L.G.C.); (M.K.); (L.M.); (O.G.)
| | - Ying Liu
- Department of Psychiatry, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA; (Y.L.); (P.J.); (Y.L.)
- Department of Medicine (Biomedical Genetics), Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA
| | - Olivier George
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA; (G.d.G.); (L.L.G.C.); (M.K.); (L.M.); (O.G.)
| | - Huiping Zhang
- Department of Psychiatry, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA; (Y.L.); (P.J.); (Y.L.)
- Department of Medicine (Biomedical Genetics), Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
19
|
Kołosowska KA, Schratt G, Winterer J. microRNA-dependent regulation of gene expression in GABAergic interneurons. Front Cell Neurosci 2023; 17:1188574. [PMID: 37213213 PMCID: PMC10196030 DOI: 10.3389/fncel.2023.1188574] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 04/20/2023] [Indexed: 05/23/2023] Open
Abstract
Information processing within neuronal circuits relies on their proper development and a balanced interplay between principal and local inhibitory interneurons within those circuits. Gamma-aminobutyric acid (GABA)ergic inhibitory interneurons are a remarkably heterogeneous population, comprising subclasses based on their morphological, electrophysiological, and molecular features, with differential connectivity and activity patterns. microRNA (miRNA)-dependent post-transcriptional control of gene expression represents an important regulatory mechanism for neuronal development and plasticity. miRNAs are a large group of small non-coding RNAs (21-24 nucleotides) acting as negative regulators of mRNA translation and stability. However, while miRNA-dependent gene regulation in principal neurons has been described heretofore in several studies, an understanding of the role of miRNAs in inhibitory interneurons is only beginning to emerge. Recent research demonstrated that miRNAs are differentially expressed in interneuron subclasses, are vitally important for migration, maturation, and survival of interneurons during embryonic development and are crucial for cognitive function and memory formation. In this review, we discuss recent progress in understanding miRNA-dependent regulation of gene expression in interneuron development and function. We aim to shed light onto mechanisms by which miRNAs in GABAergic interneurons contribute to sculpting neuronal circuits, and how their dysregulation may underlie the emergence of numerous neurodevelopmental and neuropsychiatric disorders.
Collapse
Affiliation(s)
| | - Gerhard Schratt
- Lab of Systems Neuroscience, Department of Health Science and Technology, Institute for Neuroscience, Swiss Federal Institute of Technology ETH, Zurich, Switzerland
| | - Jochen Winterer
- Lab of Systems Neuroscience, Department of Health Science and Technology, Institute for Neuroscience, Swiss Federal Institute of Technology ETH, Zurich, Switzerland
| |
Collapse
|
20
|
The Effects of Transcranial Focused Ultrasound Stimulation of Nucleus Accumbens on Neuronal Gene Expression and Brain Tissue in High Alcohol-Preferring Rats. Mol Neurobiol 2023; 60:1099-1116. [PMID: 36417101 DOI: 10.1007/s12035-022-03130-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 11/09/2022] [Indexed: 11/24/2022]
Abstract
We investigated the effect of low-intensity focused ultrasound (LIFU) on gene expression related to alcohol dependence and histological effects on brain tissue. We also aimed at determining the miRNA-mRNA relationship and their pathways in alcohol dependence-induced expression changes after focused ultrasound therapy. We designed a case-control study for 100 days of observation to investigate differences in gene expression in the short-term stimulation group (STS) and long-term stimulation group (LTS) compared with the control sham group (SG). The study was performed in our Experimental Research Laboratory. 24 male high alcohol-preferring rats 63 to 79 days old, weighing 270 to 300 g, were included in the experiment. LTS received 50-day LIFU and STS received 10-day LIFU and 40-day sham stimulation, while the SG received 50-day sham stimulation. In miRNA expression analysis, it was found that LIFU caused gene expression differences in NAc. Significant differences were found between the groups for gene expression. Compared to the SG, the expression of 454 genes in the NAc region was changed in the STS while the expression of 382 genes was changed in the LTS. In the LTS, the expression of 32 genes was changed in total compared to STS. Our data suggest that LIFU targeted on NAc may assist in the treatment of alcohol dependence, especially in the long term possibly through altering gene expression. Our immunohistochemical studies verified that LIFU does not cause any tissue damage. These findings may lead to new studies in investigating the efficacy of LIFU for the treatment of alcohol dependence and also for other psychiatric disorders.
Collapse
|
21
|
Popova D, Gameiro-Ros I, Youssef MM, Zalamea P, Morris AD, Prytkova I, Jadali A, Kwan KY, Kamarajan C, Salvatore JE, Xuei X, Chorlian DB, Porjesz B, Kuperman S, Dick DM, Goate A, Edenberg HJ, Tischfield JA, Pang ZP, Slesinger PA, Hart RP. Alcohol reverses the effects of KCNJ6 (GIRK2) noncoding variants on excitability of human glutamatergic neurons. Mol Psychiatry 2023; 28:746-758. [PMID: 36207584 PMCID: PMC9542475 DOI: 10.1038/s41380-022-01818-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 09/20/2022] [Accepted: 09/23/2022] [Indexed: 01/19/2023]
Abstract
Synonymous and noncoding single nucleotide polymorphisms (SNPs) in the KCNJ6 gene, encoding G protein-gated inwardly rectifying potassium channel subunit 2 (GIRK2), have been linked with increased electroencephalographic frontal theta event-related oscillations (ERO) in subjects diagnosed with alcohol use disorder (AUD). To identify molecular and cellular mechanisms while retaining the appropriate genetic background, we generated induced excitatory glutamatergic neurons (iN) from iPSCs derived from four AUD-diagnosed subjects with KCNJ6 variants ("Affected: AF") and four control subjects without variants ("Unaffected: UN"). Neurons were analyzed for changes in gene expression, morphology, excitability and physiological properties. Single-cell RNA sequencing suggests that KCNJ6 AF variant neurons have altered patterns of synaptic transmission and cell projection morphogenesis. Results confirm that AF neurons express lower levels of GIRK2, have greater neurite area, and elevated excitability. Interestingly, exposure to intoxicating concentrations of ethanol induces GIRK2 expression and reverses functional effects in AF neurons. Ectopic overexpression of GIRK2 alone mimics the effect of ethanol to normalize induced excitability. We conclude that KCNJ6 variants decrease GIRK2 expression and increase excitability and that this effect can be minimized or reduced with ethanol.
Collapse
Affiliation(s)
- Dina Popova
- Human Genetics Institute, Rutgers University, Piscataway, NJ, USA
| | - Isabel Gameiro-Ros
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mark M Youssef
- Department of Cell Biology & Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Petronio Zalamea
- Human Genetics Institute, Rutgers University, Piscataway, NJ, USA
| | - Ayeshia D Morris
- Joint Program in Toxicology, Rutgers University, Piscataway, NJ, USA
| | - Iya Prytkova
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Azadeh Jadali
- Department of Cell Biology & Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Kelvin Y Kwan
- Department of Cell Biology & Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Chella Kamarajan
- Dept. of Psychiatry & Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Jessica E Salvatore
- Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
| | - Xiaoling Xuei
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - David B Chorlian
- Dept. of Psychiatry & Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Bernice Porjesz
- Dept. of Psychiatry & Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Samuel Kuperman
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Danielle M Dick
- Rutgers Addiction Research Center, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
| | - Alison Goate
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Howard J Edenberg
- Department of Biochemistry and Molecular Biology, Indiana Univ School of Medicine, Indianapolis, IN, USA
| | - Jay A Tischfield
- Human Genetics Institute, Rutgers University, Piscataway, NJ, USA
| | - Zhiping P Pang
- Human Genetics Institute, Rutgers University, Piscataway, NJ, USA
- Child Health Institute, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Paul A Slesinger
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ronald P Hart
- Human Genetics Institute, Rutgers University, Piscataway, NJ, USA.
- Department of Cell Biology & Neuroscience, Rutgers University, Piscataway, NJ, USA.
| |
Collapse
|
22
|
Domi E, Barchiesi R, Barbier E. Epigenetic Dysregulation in Alcohol-Associated Behaviors: Preclinical and Clinical Evidence. Curr Top Behav Neurosci 2023. [PMID: 36717533 DOI: 10.1007/7854_2022_410] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Alcohol use disorder (AUD) is characterized by loss of control over intake and drinking despite harmful consequences. At a molecular level, AUD is associated with long-term neuroadaptations in key brain regions that are involved in reward processing and decision-making. Over the last decades, a great effort has been made to understand the neurobiological basis underlying AUD. Epigenetic mechanisms have emerged as an important mechanism in the regulation of long-term alcohol-induced gene expression changes. Here, we review the literature supporting a role for epigenetic processes in AUD. We particularly focused on the three most studied epigenetic mechanisms: DNA methylation, Histone modification and non-coding RNAs. Clinical studies indicate an association between AUD and DNA methylation both at the gene and global levels. Using behavioral paradigms that mimic some of the characteristics of AUD, preclinical studies demonstrate that changes in epigenetic mechanisms can functionally impact alcohol-associated behaviors. While many studies support a therapeutic potential for targeting epigenetic enzymes, more research is needed to fully understand their role in AUD. Identification of brain circuits underlying alcohol-associated behaviors has made major advances in recent years. However, there are very few studies that investigate how epigenetic mechanisms can affect these circuits or impact the neuronal ensembles that promote alcohol-associated behaviors. Studies that focus on the role of circuit-specific and cell-specific epigenetic changes for clinically relevant alcohol behaviors may provide new insights on the functional role of epigenetic processes in AUD.
Collapse
Affiliation(s)
- Esi Domi
- Department of Biomedical and Clinical Sciences, Center for Social and Affective Neuroscience, Linköping University, Linköping, Sweden
- School of Pharmacy, Pharmacology Unit, Center for Neuroscience, University of Camerino, Camerino, Italy
| | - Riccardo Barchiesi
- Department of Neuroscience, Waggoner Center for Alcohol and Alcohol Addiction Research, University of Texas at Austin, Austin, TX, USA
| | - Estelle Barbier
- Department of Biomedical and Clinical Sciences, Center for Social and Affective Neuroscience, Linköping University, Linköping, Sweden.
| |
Collapse
|
23
|
Thomas KT, Vermare A, Egleston SO, Wang YD, Mishra A, Lin T, Peng J, Zakharenko SS. MicroRNA 3' ends shorten during adolescent brain maturation. Front Mol Neurosci 2023; 16:1168695. [PMID: 37122627 PMCID: PMC10140418 DOI: 10.3389/fnmol.2023.1168695] [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: 02/17/2023] [Accepted: 03/21/2023] [Indexed: 05/02/2023] Open
Abstract
MicroRNA (miRNA) dysregulation is well-documented in psychiatric disease, but miRNA dynamics remain poorly understood during adolescent and early adult brain maturation, when symptoms often first appear. Here, we use RNA sequencing to examine miRNAs and their mRNA targets in cortex and hippocampus from early-, mid-, and late-adolescent and adult mice. Furthermore, we use quantitative proteomics by tandem mass tag mass spectrometry (TMT-MS) to examine protein dynamics in cortex from the same subjects. We found that ~25% of miRNAs' 3' ends shorten with age due to increased 3' trimming and decreased U tailing. Particularly, shorter but functionally competent isoforms (isomiRs) of miR-338-3p increase up to 10-fold during adolescence and only in brain. MiRNAs that undergo 3' shortening exhibit stronger negative correlations with targets that decrease with age and stronger positive correlations with targets that increase with age, than miRNAs with stable 3' ends. Increased 3' shortening with age was also observed in available mouse and human miRNA-seq data sets, and stronger correlations between miRNAs that undergo shortening and their mRNA targets were observed in two of the three available data sets. We conclude that age-associated miRNA 3' shortening is a well-conserved feature of postnatal brain maturation.
Collapse
Affiliation(s)
- Kristen T. Thomas
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Anaïs Vermare
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Suzannah O. Egleston
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Yong-Dong Wang
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Ashutosh Mishra
- Center for Proteomics and Metabolomics, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Tong Lin
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Junmin Peng
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, United States
- Center for Proteomics and Metabolomics, St. Jude Children’s Research Hospital, Memphis, TN, United States
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Stanislav S. Zakharenko
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, United States
- *Correspondence: Stanislav S. Zakharenko,
| |
Collapse
|
24
|
Wang JQ, Liu YR, Xia QR, Liang J, Wang JL, Li J. Functional roles, regulatory mechanisms and theranostics applications of ncRNAs in alcohol use disorder. Int J Biol Sci 2023; 19:1316-1335. [PMID: 36923934 PMCID: PMC10008696 DOI: 10.7150/ijbs.81518] [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: 12/04/2022] [Accepted: 02/02/2023] [Indexed: 03/14/2023] Open
Abstract
Alcohol use disorder (AUD) is one of the most prevalent neuropsychological disorders worldwide, and its pathogenesis is convoluted and poorly understood. There is considerable evidence demonstrating significant associations between multiple heritable factors and the onset and progression of AUD. In recent years, a substantial body of research conducted by emerging biotechnologies has increasingly highlighted the crucial roles of noncoding RNAs (ncRNAs) in the pathophysiology of mental diseases. As in-depth understanding of ncRNAs and their mechanisms of action, they have emerged as prospective diagnostic indicators and preclinical therapeutic targets for a variety of psychiatric illness, including AUD. Of note, dysregulated expression of ncRNAs such as circRNAs, lncRNAs and miRNAs was routinely found in AUD individuals, and besides, exogenous regulation of partial ncRNAs has also been shown to be effective in ameliorating alcohol preference and excessive alcohol consumption. However, the exact molecular mechanism still remains elusive. Herein, we systematically summarized current knowledge regarding alterations in the expression of certain ncRNAs as well as their-mediated regulatory mechanisms in individuals with AUD. And finally, we detailedly reviewed the potential theranostics applications of gene therapy agents targeting ncRNAs in AUD mice. Overall, a deeper comprehension of functional roles and biological mechanisms of ncRNAs may make significant contributions to the accurate diagnosis and effective treatment of AUD.
Collapse
Affiliation(s)
- Jie-Quan Wang
- Department of Pharmacy, Affiliated Psychological Hospital of Anhui Medical University, Hefei, 230000, China.,Department of Pharmacy, Hefei Fourth People's Hospital, Hefei, 230000, China.,Psychopharmacology Research Laboratory, Anhui Mental Health Center, Hefei, 230000, China.,Anhui Clinical Research Center for Mental Disorders, Hefei,230000, China
| | - Ya-Ru Liu
- Department of Pharmacy, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.,The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, 230022, China
| | - Qing-Rong Xia
- Department of Pharmacy, Affiliated Psychological Hospital of Anhui Medical University, Hefei, 230000, China.,Department of Pharmacy, Hefei Fourth People's Hospital, Hefei, 230000, China.,Psychopharmacology Research Laboratory, Anhui Mental Health Center, Hefei, 230000, China.,Anhui Clinical Research Center for Mental Disorders, Hefei,230000, China
| | - Jun Liang
- Department of Pharmacy, Affiliated Psychological Hospital of Anhui Medical University, Hefei, 230000, China.,Department of Pharmacy, Hefei Fourth People's Hospital, Hefei, 230000, China.,Psychopharmacology Research Laboratory, Anhui Mental Health Center, Hefei, 230000, China.,Anhui Clinical Research Center for Mental Disorders, Hefei,230000, China
| | - Jin-Liang Wang
- Department of Pharmacy, Affiliated Psychological Hospital of Anhui Medical University, Hefei, 230000, China.,Department of Pharmacy, Hefei Fourth People's Hospital, Hefei, 230000, China.,Psychopharmacology Research Laboratory, Anhui Mental Health Center, Hefei, 230000, China.,Anhui Clinical Research Center for Mental Disorders, Hefei,230000, China
| | - Jun Li
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| |
Collapse
|
25
|
Seyednejad SA, Sartor GC. Noncoding RNA therapeutics for substance use disorder. ADVANCES IN DRUG AND ALCOHOL RESEARCH 2022; 2:10807. [PMID: 36601439 PMCID: PMC9808746 DOI: 10.3389/adar.2022.10807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Although noncoding RNAs (ncRNAs) have been shown to regulate maladaptive neuroadaptations that drive compulsive drug use, ncRNA-targeting therapeutics for substance use disorder (SUD) have yet to be clinically tested. Recent advances in RNA-based drugs have improved many therapeutic issues related to immune response, specificity, and delivery, leading to multiple successful clinical trials for other diseases. As the need for safe and effective treatments for SUD continues to grow, novel nucleic acid-based therapeutics represent an appealing approach to target ncRNA mechanisms in SUD. Here, we review ncRNA processes implicated in SUD, discuss recent therapeutic approaches for targeting ncRNAs, and highlight potential opportunities and challenges of ncRNA-targeting therapeutics for SUD.
Collapse
Affiliation(s)
- Seyed Afshin Seyednejad
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, United States
- Connecticut Institute for the Brain and Cognitive Sciences (CT IBACS), Storrs, CT, United States
| | - Gregory C. Sartor
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, United States
- Connecticut Institute for the Brain and Cognitive Sciences (CT IBACS), Storrs, CT, United States
| |
Collapse
|
26
|
Plasil SL, Collins VJ, Baratta AM, Farris SP, Homanics GE. Hippocampal ceRNA networks from chronic intermittent ethanol vapor-exposed male mice and functional analysis of top-ranked lncRNA genes for ethanol drinking phenotypes. ADVANCES IN DRUG AND ALCOHOL RESEARCH 2022; 2:10831. [PMID: 36908580 PMCID: PMC10004261 DOI: 10.3389/adar.2022.10831] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The molecular mechanisms regulating the development and progression of alcohol use disorder (AUD) are largely unknown. While noncoding RNAs have previously been implicated as playing key roles in AUD, long-noncoding RNA (lncRNA) remains understudied in relation to AUD. In this study, we first identified ethanol-responsive lncRNAs in the mouse hippocampus that are transcriptional network hub genes. Microarray analysis of lncRNA, miRNA, circular RNA, and protein coding gene expression in the hippocampus from chronic intermittent ethanol vapor- or air- (control) exposed mice was used to identify ethanol-responsive competing endogenous RNA (ceRNA) networks. Highly interconnected lncRNAs (genes that had the strongest overall correlation to all other dysregulated genes identified) were ranked. The top four lncRNAs were novel, previously uncharacterized genes named Gm42575, 4930413E15Rik, Gm15767, and Gm33447, hereafter referred to as Pitt1, Pitt2, Pitt3, and Pitt4, respectively. We subsequently tested the hypothesis that CRISPR/Cas9 mutagenesis of the putative promoter and first exon of these lncRNAs in C57BL/6J mice would alter ethanol drinking behavior. The Drinking in the Dark (DID) assay was used to examine binge-like drinking behavior, and the Every-Other-Day Two-Bottle Choice (EOD-2BC) assay was used to examine intermittent ethanol consumption and preference. No significant differences between control and mutant mice were observed in the DID assay. Female-specific reductions in ethanol consumption were observed in the EOD-2BC assay for Pitt1, Pitt3, and Pitt4 mutant mice compared to controls. Male-specific alterations in ethanol preference were observed for Pitt1 and Pitt2. Female-specific increases in ethanol preference were observed for Pitt3 and Pitt4. Total fluid consumption was reduced in Pitt1 and Pitt2 mutants at 15% v/v ethanol and in Pitt3 and Pitt4 at 20% v/v ethanol in females only. We conclude that all lncRNAs targeted altered ethanol drinking behavior, and that lncRNAs Pitt1, Pitt3, and Pitt4 influenced ethanol consumption in a sex-specific manner. Further research is necessary to elucidate the biological mechanisms for these effects. These findings add to the literature implicating noncoding RNAs in AUD and suggest lncRNAs also play an important regulatory role in the disease.
Collapse
Affiliation(s)
- SL Plasil
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - VJ Collins
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - AM Baratta
- Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - SP Farris
- Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - GE Homanics
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
27
|
Ramos A, Joshi RS, Szabo G. Innate immune activation: Parallels in alcohol use disorder and Alzheimer’s disease. Front Mol Neurosci 2022; 15:910298. [PMID: 36157070 PMCID: PMC9505690 DOI: 10.3389/fnmol.2022.910298] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 07/18/2022] [Indexed: 11/16/2022] Open
Abstract
Alcohol use disorder is associated with systemic inflammation and organ dysfunction especially in the liver and the brain. For more than a decade, studies have highlighted alcohol abuse-mediated impairment of brain function and acceleration of neurodegeneration through inflammatory mechanisms that directly involve innate immune cells. Furthermore, recent studies indicate overlapping genetic risk factors between alcohol use and neurodegenerative disorders, specifically regarding the role of innate immunity in the pathomechanisms of both areas. Considering the pressing need for a better understanding of the relevance of alcohol abuse in dementia progression, here we summarize the molecular mechanisms of neuroinflammation observed in alcohol abuse and Alzheimer’s disease, the most common cause of dementia. In addition, we highlight mechanisms that are already established in the field of Alzheimer’s disease that may be relevant to explore in alcoholism to better understand alcohol mediated neurodegeneration and dementia, including the relevance of the liver-brain axis.
Collapse
Affiliation(s)
- Adriana Ramos
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Radhika S. Joshi
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Gyongyi Szabo
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- Broad Institute of MIT and Harvard, Cambridge, MA, United States
- *Correspondence: Gyongyi Szabo,
| |
Collapse
|
28
|
Zhao Y, Qin F, Han S, Li S, Zhao Y, Wang H, Tian J, Cen X. MicroRNAs in drug addiction: Current status and future perspectives. Pharmacol Ther 2022; 236:108215. [DOI: 10.1016/j.pharmthera.2022.108215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/11/2022] [Accepted: 05/16/2022] [Indexed: 12/21/2022]
|
29
|
Sprang M, Andrade-Navarro MA, Fontaine JF. Batch effect detection and correction in RNA-seq data using machine-learning-based automated assessment of quality. BMC Bioinformatics 2022; 23:279. [PMID: 35836114 PMCID: PMC9284682 DOI: 10.1186/s12859-022-04775-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 06/08/2022] [Indexed: 11/26/2022] Open
Abstract
Background The constant evolving and development of next-generation sequencing techniques lead to high throughput data composed of datasets that include a large number of biological samples. Although a large number of samples are usually experimentally processed by batches, scientific publications are often elusive about this information, which can greatly impact the quality of the samples and confound further statistical analyzes. Because dedicated bioinformatics methods developed to detect unwanted sources of variance in the data can wrongly detect real biological signals, such methods could benefit from using a quality-aware approach. Results We recently developed statistical guidelines and a machine learning tool to automatically evaluate the quality of a next-generation-sequencing sample. We leveraged this quality assessment to detect and correct batch effects in 12 publicly available RNA-seq datasets with available batch information. We were able to distinguish batches by our quality score and used it to correct for some batch effects in sample clustering. Overall, the correction was evaluated as comparable to or better than the reference method that uses a priori knowledge of the batches (in 10 and 1 datasets of 12, respectively; total = 92%). When coupled to outlier removal, the correction was more often evaluated as better than the reference (comparable or better in 5 and 6 datasets of 12, respectively; total = 92%). Conclusions In this work, we show the capabilities of our software to detect batches in public RNA-seq datasets from differences in the predicted quality of their samples. We also use these insights to correct the batch effect and observe the relation of sample quality and batch effect. These observations reinforce our expectation that while batch effects do correlate with differences in quality, batch effects also arise from other artifacts and are more suitably corrected statistically in well-designed experiments. Supplementary Information The online version contains supplementary material available at 10.1186/s12859-022-04775-y.
Collapse
Affiliation(s)
- Maximilian Sprang
- Faculty of Biology, Johannes Gutenberg-Universität Mainz, Biozentrum I, Hans-Dieter-Hüsch-Weg 15, 55128, Mainz, Germany.
| | - Miguel A Andrade-Navarro
- Faculty of Biology, Johannes Gutenberg-Universität Mainz, Biozentrum I, Hans-Dieter-Hüsch-Weg 15, 55128, Mainz, Germany
| | - Jean-Fred Fontaine
- Faculty of Biology, Johannes Gutenberg-Universität Mainz, Biozentrum I, Hans-Dieter-Hüsch-Weg 15, 55128, Mainz, Germany
| |
Collapse
|
30
|
Integrative analysis of expression profile indicates the ECM receptor and LTP dysfunction in the glioma-related epilepsy. BMC Genomics 2022; 23:430. [PMID: 35676651 PMCID: PMC9175475 DOI: 10.1186/s12864-022-08665-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 06/01/2022] [Indexed: 11/23/2022] Open
Abstract
Background Seizures are a common symptom in glioma patients, and they can cause brain dysfunction. However, the mechanism by which glioma-related epilepsy (GRE) causes alterations in brain networks remains elusive. Objective To investigate the potential pathogenic mechanism of GRE by analyzing the dynamic expression profiles of microRNA/ mRNA/ lncRNA in brain tissues of glioma patients. Methods Brain tissues of 16 patients with GRE and 9 patients with glioma without epilepsy (GNE) were collected. The total RNA was dephosphorylated, labeled, and hybridized to the Agilent Human miRNA Microarray, Release 19.0, 8 × 60 K. The cDNA was labeled and hybridized to the Agilent LncRNA + mRNA Human Gene Expression Microarray V3.0, 4 × 180 K. The raw data was extracted from hybridized images using Agilent Feature Extraction, and quantile normalization was performed using the Agilent GeneSpring. P-value < 0.05 and absolute fold change > 2 were considered the threshold of differential expression data. Data analyses were performed using R and Bioconductor. Results We found that 3 differentially expressed miRNAs (miR-10a-5p, miR-10b-5p, miR-629-3p), 6 differentially expressed lncRNAs (TTN-AS1, LINC00641, SNHG14, LINC00894, SNHG1, OIP5-AS1), and 49 differentially expressed mRNAs play a vitally critical role in developing GRE. The expression of GABARAPL1, GRAMD1B, and IQSEC3 were validated more than twofold higher in the GRE group than in the GNE group in the validation cohort. Pathways including ECM receptor interaction and long-term potentiation (LTP) may contribute to the disease’s progression. Meanwhile, We built a lncRNA-microRNA-Gene regulatory network with structural and functional significance. Conclusion These findings can offer a fresh perspective on GRE-induced brain network changes. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-022-08665-8.
Collapse
|
31
|
Liu Y, Zhang H. RNA m6A Modification Changes in Postmortem Nucleus Accumbens of Subjects with Alcohol Use Disorder: A Pilot Study. Genes (Basel) 2022; 13:958. [PMID: 35741720 PMCID: PMC9222907 DOI: 10.3390/genes13060958] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND The nucleus accumbens (NAc) is a key brain structure mediating the rewarding effect of alcohol and drug abuse. Chronic alcohol consumption may alter RNA methylome (or epitranscriptome) in the NAc, leading to altered gene expression and thus behavioral neuroadaptation to alcohol. METHODS This pilot study profiled the epitranscriptomes of mRNAs, long noncoding RNAs (lncRNAs), and microRNAs (miRNAs) in postmortem NAc of three male Caucasian subjects with alcohol use disorder (AUD) and three matched male Caucasian control subjects using Arraystar's m6A-mRNA&lncRNA Epitranscriptomic Microarray assay. Differentially methylated (DM) RNAs and the function of DM RNAs were analyzed by biostatistics and bioinformatics programs. RESULTS 26 mRNAs were hypermethylated and three mRNAs were hypomethylated in the NAc of AUD subjects (≥2-fold changes and p ≤ 0.05). Most of these 29 DM mRNAs are involved in immune-related pathways (e.g., IL-17 signaling). Moreover, four lncRNAs were hypermethylated and one lncRNA was hypomethylated in the NAc of AUD subjects (≥2-fold changes and p ≤ 0.05). Additionally, three miRNAs were hypermethylated in the NAc of AUD subjects (≥2-fold changes and p ≤ 0.05). CONCLUSIONS This study revealed RNA methylomic changes in the NAc of AUD subjects, suggesting that chronic alcohol consumption may lead to AUD through epitranscriptomic RNA modifications. Our findings need to be replicated in a larger sample.
Collapse
Affiliation(s)
- Ying Liu
- Department of Psychiatry, Boston University School of Medicine, Boston, MA 02118, USA;
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA 02118, USA
| | - Huiping Zhang
- Department of Psychiatry, Boston University School of Medicine, Boston, MA 02118, USA;
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
32
|
Mohamed DI, Alaa El-Din Aly El-Waseef D, Nabih ES, El-Kharashi OA, Abd El-Kareem HF, Abo Nahas HH, Abdel-Wahab BA, Helmy YA, Alshawwa SZ, Saied EM. Acetylsalicylic Acid Suppresses Alcoholism-Induced Cognitive Impairment Associated with Atorvastatin Intake by Targeting Cerebral miRNA155 and NLRP3: In Vivo, and In Silico Study. Pharmaceutics 2022; 14:529. [PMID: 35335908 PMCID: PMC8948796 DOI: 10.3390/pharmaceutics14030529] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 12/13/2022] Open
Abstract
Alcoholism is one of the most common diseases that can lead to the development of several chronic diseases including steatosis, and cognitive dysfunction. Statins are lipid-lowering drugs that are commonly prescribed for patients with fatty liver diseases; however, the exact effect of statins on cognitive function is still not fully understood. In the present study, we have investigated the molecular and microscopic basis of cognitive impairment induced by alcohol and/or Atorvastatin (ATOR) administration to male Wistar albino rats and explored the possible protective effect of acetylsalicylic acid (ASA). The biochemical analysis indicated that either alcohol or ATOR or together in combination produced a significant increase in the nucleotide-binding domain-like receptor 3 (NLRP3), interleukin-1β (IL-1β) miRNA155 expression levels in the frontal cortex of the brain tissue. The histological and morphometric analysis showed signs of degeneration in the neurons and the glial cells with aggregations of inflammatory cells and a decrease in the mean thickness of the frontal cortex. Immunohistochemical analysis showed a significant increase in the caspase-8 immunoreaction in the neurons and glial cells of the frontal cortex. Interestingly, administration of ASA reversed the deleterious effect of the alcohol and ATOR intake and improved the cognitive function as indicated by biochemical and histological analysis. ASA significantly decreased the expression levels of miRNA155, NLRP3, and IL1B, and produced a significant decrease in caspase-8 immunoreaction in the neurons and glial cells of the frontal cortex with a reduction in the process of neuroinflammation and neuronal damage. To further investigate these findings, we have performed an extensive molecular docking study to investigate the binding affinity of ASA to the binding pockets of the NLRP3 protein. Our results indicated that ASA has high binding scores toward the active sites of the NLRP3 NACHT domain with the ability to bind to the NLRP3 pockets by a set of hydrophilic and hydrophobic interactions. Taken together, the present study highlights the protective pharmacological effect of ASA to attenuate the deleterious effect of alcohol intake and long term ATOR therapy on the cognitive function via targeting miRNA155 and NLRP3 proteins.
Collapse
Affiliation(s)
- Doaa I. Mohamed
- Department of Clinical Pharmacology and Therapeutics, Faculty of Medicine, Ain Shams University, Cairo 11566, Egypt;
| | | | - Enas S. Nabih
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Ain Shams University, Cairo 11566, Egypt;
| | - Omnyah A. El-Kharashi
- Department of Clinical Pharmacology and Therapeutics, Faculty of Medicine, Ain Shams University, Cairo 11566, Egypt;
| | - Hanaa F. Abd El-Kareem
- Zoology Department, Faculty of Science, Ain Shams University, Abbasseya, Cairo 11566, Egypt;
| | | | - Basel A. Abdel-Wahab
- Department of Medical Pharmacology, College of Medicine, Assiut University, Assiut 71111, Egypt;
- Department of Pharmacology, College of Pharmacy, Najran University, Najran 1988, Saudi Arabia
| | - Yosra A. Helmy
- Department of Veterinary Science, College of Agriculture, Food, and Environment, University of Kentucky, Lexington, KY 40503, USA;
- Department of Animal Hygiene, Zoonoses and Animal Ethology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Samar Zuhair Alshawwa
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia;
| | - Essa M. Saied
- Chemistry Department, Faculty of Science, Suez Canal University, Ismailia 41522, Egypt
- Institute for Chemistry, Humboldt Universität zu Berlin, Brook-Taylor-Str. 2, 12489 Berlin, Germany
| |
Collapse
|
33
|
Mead EA, Boulghassoul-Pietrzykowska N, Wang Y, Anees O, Kinstlinger NS, Lee M, Hamza S, Feng Y, Pietrzykowski AZ. Non-Invasive microRNA Profiling in Saliva can Serve as a Biomarker of Alcohol Exposure and Its Effects in Humans. Front Genet 2022; 12:804222. [PMID: 35126468 PMCID: PMC8812725 DOI: 10.3389/fgene.2021.804222] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/13/2021] [Indexed: 12/14/2022] Open
Abstract
Alcohol Use Disorder (AUD) is one of the most prevalent mental disorders worldwide. Considering the widespread occurrence of AUD, a reliable, cheap, non-invasive biomarker of alcohol consumption is desired by healthcare providers, clinicians, researchers, public health and criminal justice officials. microRNAs could serve as such biomarkers. They are easily detectable in saliva, which can be sampled from individuals in a non-invasive manner. Moreover, microRNAs expression is dynamically regulated by environmental factors, including alcohol. Since excessive alcohol consumption is a hallmark of alcohol abuse, we have profiled microRNA expression in the saliva of chronic, heavy alcohol abusers using microRNA microarrays. We observed significant changes in salivary microRNA expression caused by excessive alcohol consumption. These changes fell into three categories: downregulated microRNAs, upregulated microRNAs, and microRNAs upregulated de novo. Analysis of these combinatorial changes in microRNA expression suggests dysregulation of specific biological pathways leading to impairment of the immune system and development of several types of epithelial cancer. Moreover, some of the altered microRNAs are also modulators of inflammation, suggesting their contribution to pro-inflammatory mechanisms of alcohol actions. Establishment of the cellular source of microRNAs in saliva corroborated these results. We determined that most of the microRNAs in saliva come from two types of cells: leukocytes involved in immune responses and inflammation, and buccal cells, involved in development of epithelial, oral cancers. In summary, we propose that microRNA profiling in saliva can be a useful, non-invasive biomarker allowing the monitoring of alcohol abuse, as well as alcohol-related inflammation and early detection of cancer.
Collapse
Affiliation(s)
- Edward A. Mead
- Laboratory of Adaptation, Reward and Addiction, Department of Animal Sciences, Rutgers University, New Brunswick, NJ, United States
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Nadia Boulghassoul-Pietrzykowska
- Laboratory of Adaptation, Reward and Addiction, Department of Animal Sciences, Rutgers University, New Brunswick, NJ, United States
- Mayo Clinic Health System, NWWI, Barron, WI, United States
- Department of Medicine, Capital Health, Trenton, NJ, United States
- Weight and Life MD, Hamilton, NJ, United States
| | - Yongping Wang
- Laboratory of Adaptation, Reward and Addiction, Department of Animal Sciences, Rutgers University, New Brunswick, NJ, United States
- Holmdel Township School, Holmdel, NJ, United States
| | - Onaiza Anees
- Laboratory of Adaptation, Reward and Addiction, Department of Animal Sciences, Rutgers University, New Brunswick, NJ, United States
- Virginia Commonwealth University Health, CMH Behavioral Health, South Hill, VA, United States
| | - Noah S. Kinstlinger
- Laboratory of Adaptation, Reward and Addiction, Department of Animal Sciences, Rutgers University, New Brunswick, NJ, United States
- Albert Einstein College of Medicine, Bronx, NY, United States
| | - Maximillian Lee
- Laboratory of Adaptation, Reward and Addiction, Department of Animal Sciences, Rutgers University, New Brunswick, NJ, United States
- George Washington University, School of Medicine and Health Sciences, Washington DC, MA, United States
| | - Shireen Hamza
- Laboratory of Adaptation, Reward and Addiction, Department of Animal Sciences, Rutgers University, New Brunswick, NJ, United States
- Department of the History of Science, Harvard University, Cambridge, MA, United States
| | - Yaping Feng
- Waksman Genomics Core Facility, Rutgers University, Piscataway, NJ, United States
- Bioinformatics Department, Admera Health, South Plainfield, NJ, United States
| | - Andrzej Z. Pietrzykowski
- Laboratory of Adaptation, Reward and Addiction, Department of Animal Sciences, Rutgers University, New Brunswick, NJ, United States
- Weight and Life MD, Hamilton, NJ, United States
| |
Collapse
|