1
|
Zhang L, Zhang J, Wang N, Liu C, Wang S, Dong X, Yang L, Bao X, Nie X, Li J. Bioinformatics-based identification of CTSS, DOK2, and ENTPD1 as potential blood biomarkers of schizophrenia. BMC Psychiatry 2025; 25:157. [PMID: 39972407 PMCID: PMC11841330 DOI: 10.1186/s12888-025-06512-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 01/20/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Although schizophrenia is a severe mental disorder that significantly impacts patients and society, there are currently no reliable blood-based biomarkers to assist in its diagnosis. The diagnosis primarily relies on clinical assessment and patient history, a method that is inherently subjective and prone to errors, potentially leading to diagnostic delays. In this study, we aim to utilize bioinformatics approaches to explore potential blood-based biomarkers for the diagnosis of schizophrenia. By employing advanced bioinformatics techniques, we hope to identify key genes and construct an effective diagnostic model, providing the clinic with a more objective and accurate diagnostic tool. METHODS In this research, we employed bioinformatics techniques to identify potential blood-based biomarkers for the diagnosis of schizophrenia. Initially, we selected schizophrenia-associated differentially expressed genes (DEGs) from the Gene Expression Omnibus (GEO) database through the datasets GSE27383, GSE38484, and GSE38481. Subsequently, we performed Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses on these DEGs to elucidate their biological functions and related pathways. Furthermore, we constructed a protein-protein interaction (PPI) network of the differentially expressed genes to identify key genes and matched them with their target microRNAs (miRNAs). In addition, we assessed the diagnostic potential of these key genes through immune infiltration analysis. The aim of this study is to reveal the roles of these hub genes in the pathogenesis of schizophrenia. RESULTS Through bioinformatics analysis, we have identified three potential hub genes associated with the pathogenesis of schizophrenia: CTSS, DOK2, and ENTPD1. These genes are significantly correlated with the development of schizophrenia and may serve as promising diagnostic biomarkers for the condition. CONCLUSION In this study, we have identified three pivotal genes-CTSS, DOK2, and ENTPD1-that are intimately associated with the pathogenesis of schizophrenia. The discovery of these genes not only enhances the precision of diagnostic efforts for schizophrenia but also provides a robust scientific foundation for the development of innovative treatment approaches for schizophrenia and related disorders. The identification of these biomarkers offers a tangible basis for early, accurate diagnosis, treatment, prognostic assessment, and rehabilitation evaluation in schizophrenia, potentially improving patients' quality of life and supporting the development of personalized therapeutics and antipsychotic medications.
Collapse
Affiliation(s)
- Lei Zhang
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
| | - Jiale Zhang
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
| | - Na Wang
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
| | - Chenwei Liu
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
| | - Shuting Wang
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
| | - Xiaotao Dong
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
| | - Lu Yang
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
| | - Xiaohong Bao
- Department of Cardiothoracic Surgery, School of Medicine, Taizhou Central Hospital, Taizhou University, Taizhou, 318000, China
| | - Xiaobo Nie
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
| | - Jicheng Li
- School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China.
| |
Collapse
|
2
|
Neri M, Brovelli A, Castro S, Fraisopi F, Gatica M, Herzog R, Mediano PAM, Mindlin I, Petri G, Bor D, Rosas FE, Tramacere A, Estarellas M. A Taxonomy of Neuroscientific Strategies Based on Interaction Orders. Eur J Neurosci 2025; 61:e16676. [PMID: 39906974 DOI: 10.1111/ejn.16676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/15/2024] [Accepted: 12/29/2024] [Indexed: 02/06/2025]
Abstract
In recent decades, neuroscience has advanced with increasingly sophisticated strategies for recording and analysing brain activity, enabling detailed investigations into the roles of functional units, such as individual neurons, brain regions and their interactions. Recently, new strategies for the investigation of cognitive functions regard the study of higher order interactions-that is, the interactions involving more than two brain regions or neurons. Although methods focusing on individual units and their interactions at various levels offer valuable and often complementary insights, each approach comes with its own set of limitations. In this context, a conceptual map to categorize and locate diverse strategies could be crucial to orient researchers and guide future research directions. To this end, we define the spectrum of orders of interaction, namely, a framework that categorizes the interactions among neurons or brain regions based on the number of elements involved in these interactions. We use a simulation of a toy model and a few case studies to demonstrate the utility and the challenges of the exploration of the spectrum. We conclude by proposing future research directions aimed at enhancing our understanding of brain function and cognition through a more nuanced methodological framework.
Collapse
Affiliation(s)
- Matteo Neri
- Institut de Neurosciences de la Timone, Aix-Marseille Université, UMR 7289 CNRS, Marseille, France
| | - Andrea Brovelli
- Institut de Neurosciences de la Timone, Aix-Marseille Université, UMR 7289 CNRS, Marseille, France
| | - Samy Castro
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364, Strasbourg, France
- Institut de Neurosciences Des Systèmes (INS), Aix-Marseille Université, UMR 1106, Marseille, France
| | - Fausto Fraisopi
- Institute for Advanced Study, Aix-Marseille University, Marseille, France
| | - Marilyn Gatica
- NPLab, Network Science Institute, Northeastern University London, London, UK
| | - Ruben Herzog
- DreamTeam, Paris Brain Institute (ICM), Paris, France
| | - Pedro A M Mediano
- Department of Computing, Imperial College London, London, UK
- Division of Psychology and Language Sciences, University College London, London, UK
| | - Ivan Mindlin
- DreamTeam, Paris Brain Institute (ICM), Paris, France
- PICNIC lab, Paris Brain Institute (ICM), Paris, France
| | - Giovanni Petri
- NPLab, Network Science Institute, Northeastern University London, London, UK
- Department of Physics, Northeastern University, Boston, Massachusetts, USA
- NPLab, CENTAI Institute, Turin, Italy
| | - Daniel Bor
- Department of Psychology, School of Biological and Behavioural Sciences, Queen Mary University of London, London, UK
- Department of Psychology, University of Cambridge, Cambridge, UK
| | - Fernando E Rosas
- Sussex Centre for Consciousness Science and Sussex AI, Department of Informatics, University of Sussex, Brighton, UK
- Center for Psychedelic Research and Centre for Complexity Science, Department of Brain Science, Imperial College London, London, UK
- Centre for Eudaimonia and Human Flourishing, University of Oxford, Oxford, UK
- Principles of Intelligent Behavior in Biological and Social Systems (PIBBSS), Prague, Czechia
| | - Antonella Tramacere
- Department of Philosophy, Communication and Performing Arts, Roma Tre University, Rome, Italy
| | - Mar Estarellas
- Department of Psychology, School of Biological and Behavioural Sciences, Queen Mary University of London, London, UK
- Department of Psychology, University of Cambridge, Cambridge, UK
| |
Collapse
|
3
|
Dor H, Hertzberg L. Schizophrenia Biomarkers: Blood Transcriptome Suggests Two Molecular Subtypes. Neuromolecular Med 2024; 26:50. [PMID: 39609319 PMCID: PMC11604812 DOI: 10.1007/s12017-024-08817-x] [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: 09/22/2024] [Accepted: 11/12/2024] [Indexed: 11/30/2024]
Abstract
Schizophrenia is a chronic illness that imposes a significant burden on patients, their families, and the health care system. While it has a substantial genetic component, its heterogeneous nature-both genetic and clinical-limits the ability to identify causal genes and mechanisms. In this study, we analyzed the blood transcriptomes of 398 samples (212 patients with schizophrenia and 186 controls) obtained from five public datasets. We demonstrated this heterogeneity by clustering patients with schizophrenia into two molecular subtypes using an unsupervised machine-learning algorithm. We found that the genes most influential in clustering were enriched in pathways related to the ribosome and ubiquitin-proteasomes system, which are known to be associated with schizophrenia. Based on the expression levels of these genes, we developed a logistic regression model capable of predicting schizophrenia samples in unrelated datasets with a positive predictive value of 64% (p value = 0.039). In the future, integrating blood transcriptomics with clinical characteristics may enable the definition of distinct molecular subtypes, leading to a better understanding of schizophrenia pathophysiology and aiding in the development of personalized drugs and treatment options.
Collapse
Affiliation(s)
- Herut Dor
- The Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Libi Hertzberg
- The Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel.
- Department of Physics of Complex Systems, Weizmann Institute of Science, 76100, Rehovot, Israel.
- Shalvata Mental Health Center, Affiliated with the Faculty of Medicine, Tel-Aviv University, 13 Aliat Hanoar St., 45100, Hod Hasharon, Israel.
| |
Collapse
|
4
|
Burrack N, Yitzhaky A, Mizrahi L, Wang M, Stern S, Hertzberg L. Altered Expression of PDE4 Genes in Schizophrenia: Insights from a Brain and Blood Sample Meta-Analysis and iPSC-Derived Neurons. Genes (Basel) 2024; 15:609. [PMID: 38790238 PMCID: PMC11121586 DOI: 10.3390/genes15050609] [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: 06/04/2023] [Revised: 05/03/2024] [Accepted: 05/05/2024] [Indexed: 05/26/2024] Open
Abstract
Schizophrenia symptomatology includes negative symptoms and cognitive impairment. Several studies have linked schizophrenia with the PDE4 family of enzymes due to their genetic association and function in cognitive processes such as long-term potentiation. We conducted a systematic gene expression meta-analysis of four PDE4 genes (PDE4A-D) in 10 brain sample datasets (437 samples) and three blood sample datasets (300 samples). Subsequently, we measured mRNA levels in iPSC-derived hippocampal dentate gyrus neurons generated from fibroblasts of three groups: healthy controls, healthy monozygotic twins (MZ), and their MZ siblings with schizophrenia. We found downregulation of PDE4B in brain tissues, further validated by independent data of the CommonMind consortium (515 samples). Interestingly, the downregulation signal was present in a subgroup of the patients, while the others showed no differential expression or even upregulation. Notably, PDE4A, PDE4B, and PDE4D exhibited upregulation in iPSC-derived neurons compared to healthy controls, whereas in blood samples, PDE4B was found to be upregulated while PDE4A was downregulated. While the precise mechanism and direction of altered PDE4 expression necessitate further investigation, the observed multilevel differential expression across the brain, blood, and iPSC-derived neurons compellingly suggests the involvement of PDE4 genes in the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Nitzan Burrack
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84101, Israel;
- Clinical Research Center, Soroka University Medical Center, Beer-Sheva 84101, Israel
| | - Assif Yitzhaky
- Department of Physics of Complex Systems, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Liron Mizrahi
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3103301, Israel
| | - Meiyan Wang
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Shani Stern
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3103301, Israel
| | - Libi Hertzberg
- Department of Physics of Complex Systems, Weizmann Institute of Science, Rehovot 76100, Israel
- Shalvata Mental Health Center, Affiliated with the Faculty of Medicine, Tel-Aviv University, 13 Aliat Hanoar St., Hod Hasharon 45100, Israel
| |
Collapse
|
5
|
Fiorito AM, Fakra E, Sescousse G, Ibrahim EC, Rey R. Molecular mapping of a core transcriptional signature of microglia-specific genes in schizophrenia. Transl Psychiatry 2023; 13:386. [PMID: 38092734 PMCID: PMC10719376 DOI: 10.1038/s41398-023-02677-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 11/20/2023] [Accepted: 11/22/2023] [Indexed: 12/17/2023] Open
Abstract
Besides playing a central role in neuroinflammation, microglia regulate synaptic development and is involved in plasticity. Converging lines of evidence suggest that these different processes play a critical role in schizophrenia. Furthermore, previous studies reported altered transcription of microglia genes in schizophrenia, while microglia itself seems to be involved in the etiopathology of the disease. However, the regional specificity of these brain transcriptional abnormalities remains unclear. Moreover, it is unknown whether brain and peripheral expression of microglia genes are related. Thus, we investigated the expression of a pre-registered list of 10 genes from a core signature of human microglia both at brain and peripheral levels. We included 9 independent Gene Expression Omnibus datasets (764 samples obtained from 266 individuals with schizophrenia and 237 healthy controls) from 8 different brain regions and 3 peripheral tissues. We report evidence of a widespread transcriptional alteration of microglia genes both in brain tissues (we observed a decreased expression in the cerebellum, associative striatum, hippocampus, and parietal cortex of individuals with schizophrenia compared with healthy controls) and whole blood (characterized by a mixed altered expression pattern). Our results suggest that brain underexpression of microglia genes may represent a candidate transcriptional signature for schizophrenia. Moreover, the dual brain-whole blood transcriptional alterations of microglia/macrophage genes identified support the model of schizophrenia as a whole-body disorder and lend weight to the use of blood samples as a potential source of biological peripheral biomarkers.
Collapse
Affiliation(s)
- Anna M Fiorito
- Lyon Neuroscience Research Center, INSERM U1028, CNRS UMR 5292, PSYR2 Team, University of Lyon, Lyon, France
- Centre Hospitalier Le Vinatier, Bron, France
| | - Eric Fakra
- Lyon Neuroscience Research Center, INSERM U1028, CNRS UMR 5292, PSYR2 Team, University of Lyon, Lyon, France
- Department of Psychiatry, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Guillaume Sescousse
- Lyon Neuroscience Research Center, INSERM U1028, CNRS UMR 5292, PSYR2 Team, University of Lyon, Lyon, France
- Centre Hospitalier Le Vinatier, Bron, France
| | - El Chérif Ibrahim
- Aix-Marseille Univ, CNRS, INT, Institut de Neurosciences de la Timone, Marseille, France
| | - Romain Rey
- Lyon Neuroscience Research Center, INSERM U1028, CNRS UMR 5292, PSYR2 Team, University of Lyon, Lyon, France.
- Centre Hospitalier Le Vinatier, Bron, France.
- Fondation FondaMental, Créteil, France.
| |
Collapse
|
6
|
Hess JL, Quinn TP, Zhang C, Hearn GC, Chen S, Kong SW, Cairns M, Tsuang MT, Faraone SV, Glatt SJ. BrainGENIE: The Brain Gene Expression and Network Imputation Engine. Transl Psychiatry 2023; 13:98. [PMID: 36949060 PMCID: PMC10033657 DOI: 10.1038/s41398-023-02390-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 02/23/2023] [Accepted: 02/28/2023] [Indexed: 03/24/2023] Open
Abstract
In vivo experimental analysis of human brain tissue poses substantial challenges and ethical concerns. To address this problem, we developed a computational method called the Brain Gene Expression and Network-Imputation Engine (BrainGENIE) that leverages peripheral-blood transcriptomes to predict brain tissue-specific gene-expression levels. Paired blood-brain transcriptomic data collected by the Genotype-Tissue Expression (GTEx) Project was used to train BrainGENIE models to predict gene-expression levels in ten distinct brain regions using whole-blood gene-expression profiles. The performance of BrainGENIE was compared to PrediXcan, a popular method for imputing gene expression levels from genotypes. BrainGENIE significantly predicted brain tissue-specific expression levels for 2947-11,816 genes (false-discovery rate-adjusted p < 0.05), including many transcripts that cannot be predicted significantly by a transcriptome-imputation method such as PrediXcan. BrainGENIE recapitulated measured diagnosis-related gene-expression changes in the brain for autism, bipolar disorder, and schizophrenia better than direct correlations from blood and predictions from PrediXcan. We developed a convenient software toolset for deploying BrainGENIE, and provide recommendations for how best to implement models. BrainGENIE complements and, in some ways, outperforms existing transcriptome-imputation tools, providing biologically meaningful predictions and opening new research avenues.
Collapse
Affiliation(s)
- Jonathan L Hess
- Department of Psychiatry & Behavioral Sciences, Norton College of Medicine at SUNY Upstate Medical University, Syracuse, NY, USA
| | - Thomas P Quinn
- Applied Artificial Intelligence Institute (A2I2), Deakin University, Geelong, Australia
| | - Chunling Zhang
- Department of Neuroscience & Physiology, Norton College of Medicine at SUNY Upstate Medical University, Syracuse, NY, USA
| | - Gentry C Hearn
- Department of Neuroscience & Physiology, Norton College of Medicine at SUNY Upstate Medical University, Syracuse, NY, USA
| | - Samuel Chen
- Department of Psychiatry & Behavioral Sciences, Norton College of Medicine at SUNY Upstate Medical University, Syracuse, NY, USA
| | - Sek Won Kong
- Computational Health Informatics Program, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Murray Cairns
- School of Biomedical Sciences & Pharmacy, Faculty of Health, The University of Newcastle, New South Wales, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, Newcastle, Australia
- Centre for Brain & Mental Health Research, The University of Newcastle, Callaghan, Australia
| | - Ming T Tsuang
- Center for Behavioral Genomics, Department of Psychiatry, Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA, USA
- Harvard Institute of Psychiatric Epidemiology and Genetics, Boston, MA, USA
| | - Stephen V Faraone
- Department of Psychiatry & Behavioral Sciences, Norton College of Medicine at SUNY Upstate Medical University, Syracuse, NY, USA
- Department of Neuroscience & Physiology, Norton College of Medicine at SUNY Upstate Medical University, Syracuse, NY, USA
| | - Stephen J Glatt
- Department of Psychiatry & Behavioral Sciences, Norton College of Medicine at SUNY Upstate Medical University, Syracuse, NY, USA.
- Department of Neuroscience & Physiology, Norton College of Medicine at SUNY Upstate Medical University, Syracuse, NY, USA.
| |
Collapse
|
7
|
Chen Z, Li X, Cui X, Zhang L, Liu Q, Lu Y, Wang X, Shi H, Ding M, Yang Y, Li W, Lv L. Association of CTNND2 gene polymorphism with schizophrenia: Two-sample case-control study in Chinese Han population. Int J Psychiatry Med 2023:912174231164669. [PMID: 36930964 DOI: 10.1177/00912174231164669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/19/2023]
Abstract
OBJECTIVES Genetic factors play an important role in the etiology of schizophrenia (SZ). Catenin Delta 2 (CTNND2) is one of the genes regulating neuronal development in the brain. It is unclear whether CTNND2 is involved in SZ. With the hypothesis that CTNND2 may be a risk gene for SZ, we performed a case-control association analysis to investigate if CTNND2 gene single nucleotide polymorphisms (SNPs) are implicated in SZ in a Han Chinese northern population. MATERIALS AND METHODS We recruited subjects from 2010 to 2022 from the Han population of northern Henan and divided them into two case-control samples, including a discovery sample (SZ = 528 and control = 528) and replication sample (SZ = 2458 and control = 6914). Twenty-one SNPs were genotyped on the Illumina BeadStation 500G platform using GoldenGate technology and analyzed by PLINK. Positive and Negative Syndrome Scale (PANSS) was used to assess clinical symptoms. RESULTS Rs16901943, rs7733427, and rs2168878 SNPs were associated with SZ (Chi2 = 7.484, 11.576, and 5.391, respectively, df = 1; p = 0.006, 0.00067, and 0.02, respectively) in two samples. Rs10058868 was associated with SZ in male patients in the discovery sample (Chi2 = 6.264, df = 1, p = .044). Only rs7733427 survived Bonferroni correction. Linkage disequilibrium block three haplotypes were associated with SZ in the discovery and total sample. PANSS analysis of the four SNPs implicated rs10058868 and rs2168878 with symptoms of depression and excitement, respectively, in the SZ patients. CONCLUSION Four SNPs were identified as being correlated with SZ. The CTNND2 gene may be involved in susceptibility to SZ.
Collapse
Affiliation(s)
- Zhaonian Chen
- 34727The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Xiaojing Li
- 34727The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Xiangzheng Cui
- 34727The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Luwen Zhang
- 34727The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Qing Liu
- 34727The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yanli Lu
- 34727The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Xiujuan Wang
- 34727The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Han Shi
- 34727The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Minli Ding
- 34727The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yongfeng Yang
- 34727The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Wenqiang Li
- 34727The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Luxian Lv
- 34727The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
8
|
Bharadhwaj VS, Mubeen S, Sargsyan A, Jose GM, Geissler S, Hofmann-Apitius M, Domingo-Fernández D, Kodamullil AT. Integrative analysis to identify shared mechanisms between schizophrenia and bipolar disorder and their comorbidities. Prog Neuropsychopharmacol Biol Psychiatry 2023; 122:110688. [PMID: 36462601 DOI: 10.1016/j.pnpbp.2022.110688] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 11/04/2022] [Accepted: 11/27/2022] [Indexed: 12/04/2022]
Abstract
Schizophrenia and bipolar disorder are characterized by highly similar neuropsychological signatures, implying shared neurobiological mechanisms between these two disorders. These disorders also have comorbidities, such as type 2 diabetes mellitus (T2DM). To date, an understanding of the mechanisms that mediate the link between these two disorders remains incomplete. In this work, we identify and investigate shared patterns across multiple schizophrenia, bipolar disorder and T2DM gene expression datasets through multiple strategies. Firstly, we investigate dysregulation patterns at the gene-level and compare our findings against disease-specific knowledge graphs (KGs). Secondly, we analyze the concordance of co-expression patterns across datasets to identify disease-specific as well as common pathways. Thirdly, we examine enriched pathways across datasets and disorders to identify common biological mechanisms between them. Lastly, we investigate the correspondence of shared genetic variants between these two disorders and T2DM as well as the disease-specific KGs. In conclusion, our work reveals several shared candidate genes and pathways, particularly those related to the immune system, such as TNF signaling pathway, IL-17 signaling pathway and NF-kappa B signaling pathway and nervous system, such as dopaminergic synapse and GABAergic synapse, which we propose mediate the link between schizophrenia and bipolar disorder and its shared comorbidity, T2DM.
Collapse
Affiliation(s)
- Vinay Srinivas Bharadhwaj
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing, Sankt Augustin 53757, Germany; Bonn-Aachen International Center for Information Technology (B-IT), University of Bonn, 53115 Bonn, Germany.
| | - Sarah Mubeen
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing, Sankt Augustin 53757, Germany; Bonn-Aachen International Center for Information Technology (B-IT), University of Bonn, 53115 Bonn, Germany; Fraunhofer Center for Machine Learning, Germany
| | - Astghik Sargsyan
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing, Sankt Augustin 53757, Germany
| | - Geena Mariya Jose
- Causality Biomodels, Kinfra Hi-Tech Park, Kalamassery, Cochin, Kerala 683503, India
| | | | - Martin Hofmann-Apitius
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing, Sankt Augustin 53757, Germany; Bonn-Aachen International Center for Information Technology (B-IT), University of Bonn, 53115 Bonn, Germany
| | - Daniel Domingo-Fernández
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing, Sankt Augustin 53757, Germany; Fraunhofer Center for Machine Learning, Germany; Enveda Biosciences, Boulder, CO, 80301, USA
| | - Alpha Tom Kodamullil
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing, Sankt Augustin 53757, Germany; Bonn-Aachen International Center for Information Technology (B-IT), University of Bonn, 53115 Bonn, Germany; Causality Biomodels, Kinfra Hi-Tech Park, Kalamassery, Cochin, Kerala 683503, India
| |
Collapse
|
9
|
Segev S, Yitzhaky A, Ben Shachar D, Hertzberg L. VDAC genes down-regulation in brain samples of individuals with schizophrenia is revealed by a systematic meta-analysis. Neurosci Res 2023:S0168-0102(23)00022-6. [PMID: 36717018 DOI: 10.1016/j.neures.2023.01.012] [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: 11/05/2022] [Revised: 01/23/2023] [Accepted: 01/26/2023] [Indexed: 01/29/2023]
Abstract
Mitochondrial dysfunction was shown to be involved in schizophrenia pathophysiology. Abnormal energy states can lead to alterations in neural function and thereby to the cognitive and behavioral aberrations characteristics of schizophrenia. Voltage-dependent anion-selective channels (VDAC) are located in the outer mitochondrial membrane and are involved in mitochondrial energy production. Only few studies explored VDAC genes' expression in schizophrenia, and their results were not consistent. We conducted a systematic meta-analysis of ten brain samples gene expression datasets (overall 368 samples, 179 schizophrenia, 189 controls). In addition, we conducted a meta-analysis of three blood samples datasets (overall 300 samples, 167 schizophrenia, 133 controls). Pairwise correlation analysis was conducted between the VDAC and proteasome subunit genes' expression patterns. VDAC1, VDAC2 and VDAC3 showed significant down-regulation in brain samples of patients with schizophrenia. They also showed significant positive correlations with the proteasome subunit genes' expression levels. Our findings suggest that VDAC genes might play a role in mitochondrial dysfunction in schizophrenia. VDAC1 was down-regulated also in blood samples, which suggests its potential role as a biomarker for schizophrenia. The correlation with proteasome subunits, which were previously shown to be down-regulated in a subgroup of the patients, suggests that our findings might characterize a subgroup of the patients. This direction has the potential to lead to patients' stratification and more precisely-targeted therapy and necessitates further study.
Collapse
Affiliation(s)
- Shaked Segev
- Sackler School of Medicine, Tel-Aviv University, Israel
| | - Assif Yitzhaky
- Department of Physics of Complex Systems, Weizmann Institute of Science, Rehovot, Israel
| | - Dorit Ben Shachar
- Psychobiology Research Lab, Department of Neuroscience, The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Israel
| | - Libi Hertzberg
- Sackler School of Medicine, Tel-Aviv University, Israel; Shalvata Mental Health Center, Israel; Department of Physics of Complex Systems, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
10
|
The Variation of Transcriptomic Perturbations is Associated with the Development and Progression of Various Diseases. DISEASE MARKERS 2022; 2022:2148627. [PMID: 36204511 PMCID: PMC9530920 DOI: 10.1155/2022/2148627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 08/31/2022] [Accepted: 09/07/2022] [Indexed: 11/28/2022]
Abstract
Background Although transcriptomic data have been widely applied to explore various diseases, few studies have investigated the association between transcriptomic perturbations and disease development in a wide variety of diseases. Methods Based on a previously developed algorithm for quantifying intratumor heterogeneity at the transcriptomic level, we defined the variation of transcriptomic perturbations (VTP) of a disease relative to the health status. Based on publicly available transcriptome datasets, we compared VTP values between the disease and health status and analyzed correlations between VTP values and disease progression or severity in various diseases, including neurological disorders, infectious diseases, cardiovascular diseases, respiratory diseases, liver diseases, kidney diseases, digestive diseases, and endocrine diseases. We also identified the genes and pathways whose expression perturbations correlated positively with VTP across diverse diseases. Results VTP values were upregulated in various diseases relative to their normal controls. VTP values were significantly greater in define than in possible or probable Alzheimer's disease. VTP values were significantly larger in intensive care unit (ICU) COVID-19 patients than in non-ICU patients, and in COVID-19 patients requiring mechanical ventilatory support (MVS) than in those not requiring MVS. VTP correlated positively with viral loads in acquired immune deficiency syndrome (AIDS) patients. Moreover, the AIDS patients treated with abacavir or zidovudine had lower VTP values than those without such therapies. In pulmonary tuberculosis (TB) patients, VTP values followed the pattern: active TB > latent TB > normal controls. VTP values were greater in clinically apparent than in presymptomatic malaria. VTP correlated negatively with the cardiac index of left ventricular ejection fraction (LVEF). In chronic obstructive pulmonary disease (COPD), VTP showed a negative correlation with forced expiratory volume in the first second (FEV1). VTP values increased with H. pylori infection and were upregulated in atrophic gastritis caused by H. pylori infection. The genes and pathways whose expression perturbations correlated positively with VTP scores across diseases were mainly involved in the regulation of immune, metabolic, and cellular activities. Conclusions VTP is upregulated in the disease versus health status, and its upregulation is associated with disease progression and severity in various diseases. Thus, VTP has potential clinical implications for disease diagnosis and prognosis.
Collapse
|
11
|
Ferguson LB, Roberts AJ, Mayfield RD, Messing RO. Blood and brain gene expression signatures of chronic intermittent ethanol consumption in mice. PLoS Comput Biol 2022; 18:e1009800. [PMID: 35176017 PMCID: PMC8853518 DOI: 10.1371/journal.pcbi.1009800] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 01/03/2022] [Indexed: 02/03/2023] Open
Abstract
Alcohol Use Disorder (AUD) is a chronic, relapsing syndrome diagnosed by a heterogeneous set of behavioral signs and symptoms. There are no laboratory tests that provide direct objective evidence for diagnosis. Microarray and RNA-Seq technologies enable genome-wide transcriptome profiling at low costs and provide an opportunity to identify biomarkers to facilitate diagnosis, prognosis, and treatment of patients. However, access to brain tissue in living patients is not possible. Blood contains cellular and extracellular RNAs that provide disease-relevant information for some brain diseases. We hypothesized that blood gene expression profiles can be used to diagnose AUD. We profiled brain (prefrontal cortex, amygdala, and hypothalamus) and blood gene expression levels in C57BL/6J mice using RNA-seq one week after chronic intermittent ethanol (CIE) exposure, a mouse model of alcohol dependence. We found a high degree of preservation (rho range: [0.50, 0.67]) between blood and brain transcript levels. There was small overlap between blood and brain DEGs, and considerable overlap of gene networks perturbed after CIE related to cell-cell signaling (e.g., GABA and glutamate receptor signaling), immune responses (e.g., antigen presentation), and protein processing / mitochondrial functioning (e.g., ubiquitination, oxidative phosphorylation). Blood gene expression data were used to train classifiers (logistic regression, random forest, and partial least squares discriminant analysis), which were highly accurate at predicting alcohol dependence status (maximum AUC: 90.1%). These results suggest that gene expression profiles from peripheral blood samples contain a biological signature of alcohol dependence that can discriminate between CIE and Air subjects.
Collapse
Affiliation(s)
- Laura B. Ferguson
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, Texas, United States of America
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, Texas, United States of America
- Department of Neuroscience, University of Texas at Austin, Austin, Texas, United States of America
| | - Amanda J. Roberts
- Animal Models Core Facility, The Scripps Research Institute, San Diego, California, United States of America
| | - R. Dayne Mayfield
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, Texas, United States of America
- Department of Neuroscience, University of Texas at Austin, Austin, Texas, United States of America
| | - Robert O. Messing
- Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, Texas, United States of America
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, Texas, United States of America
- Department of Neuroscience, University of Texas at Austin, Austin, Texas, United States of America
| |
Collapse
|
12
|
Chen J, Han X, Ye S, Liu L, Yang B, Cao Y, Zhuo R, Yao X. Integration of small RNA, degradome, and transcriptome sequencing data illustrates the mechanism of low phosphorus adaptation in Camellia oleifera. FRONTIERS IN PLANT SCIENCE 2022; 13:932926. [PMID: 35979079 PMCID: PMC9377520 DOI: 10.3389/fpls.2022.932926] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 07/11/2022] [Indexed: 05/02/2023]
Abstract
Phosphorus (P) is an indispensable macronutrient for plant growth and development, and it is involved in various cellular biological activities in plants. Camellia oleifera is a unique high-quality woody oil plant that grows in the hills and mountains of southern China. However, the available P content is deficient in southern woodland soil. Until now, few studies focused on the regulatory functions of microRNAs (miRNAs) and their target genes under low inorganic phosphate (Pi) stress. In this study, we integrated small RNA, degradome, and transcriptome sequencing data to investigate the mechanism of low Pi adaptation in C. oleifera. We identified 40,689 unigenes and 386 miRNAs by the deep sequencing technology and divided the miRNAs into four different groups. We found 32 miRNAs which were differentially expressed under low Pi treatment. A total of 414 target genes of 108 miRNAs were verified by degradome sequencing. Gene ontology (GO) functional analysis of target genes found that they were related to the signal response to the stimulus and transporter activity, indicating that they may respond to low Pi stress. The integrated analysis revealed that 31 miRNA-target pairs had negatively correlated expression patterns. A co-expression regulatory network was established based on the profiles of differentially expressed genes. In total, three hub genes (ARF22, WRKY53, and SCL6), which were the targets of differentially expressed miRNAs, were discovered. Our results showed that integrated analyses of the small RNA, degradome, and transcriptome sequencing data provided a valuable basis for investigating low Pi in C. oleifera and offer new perspectives on the mechanism of low Pi tolerance in woody oil plants.
Collapse
Affiliation(s)
- Juanjuan Chen
- State Key Laboratory of Tree Genetics and Breeding, Chinese Academy of Forestry, Hangzhou, China
- Key Laboratory of Tree Breeding of Zhejiang Province, Research Institute of Subtropical Forestry, Chinese Academy of Forestry, Hangzhou, China
- Forestry Faculty, Nanjing Forestry University, Nanjing, China
| | - Xiaojiao Han
- State Key Laboratory of Tree Genetics and Breeding, Chinese Academy of Forestry, Hangzhou, China
- Key Laboratory of Tree Breeding of Zhejiang Province, Research Institute of Subtropical Forestry, Chinese Academy of Forestry, Hangzhou, China
| | - Sicheng Ye
- State Key Laboratory of Tree Genetics and Breeding, Chinese Academy of Forestry, Hangzhou, China
- Key Laboratory of Tree Breeding of Zhejiang Province, Research Institute of Subtropical Forestry, Chinese Academy of Forestry, Hangzhou, China
| | - Linxiu Liu
- State Key Laboratory of Tree Genetics and Breeding, Chinese Academy of Forestry, Hangzhou, China
- Key Laboratory of Tree Breeding of Zhejiang Province, Research Institute of Subtropical Forestry, Chinese Academy of Forestry, Hangzhou, China
| | - Bingbing Yang
- State Key Laboratory of Tree Genetics and Breeding, Chinese Academy of Forestry, Hangzhou, China
- Key Laboratory of Tree Breeding of Zhejiang Province, Research Institute of Subtropical Forestry, Chinese Academy of Forestry, Hangzhou, China
| | - Yongqing Cao
- State Key Laboratory of Tree Genetics and Breeding, Chinese Academy of Forestry, Hangzhou, China
- Key Laboratory of Tree Breeding of Zhejiang Province, Research Institute of Subtropical Forestry, Chinese Academy of Forestry, Hangzhou, China
| | - Renying Zhuo
- State Key Laboratory of Tree Genetics and Breeding, Chinese Academy of Forestry, Hangzhou, China
- Key Laboratory of Tree Breeding of Zhejiang Province, Research Institute of Subtropical Forestry, Chinese Academy of Forestry, Hangzhou, China
- *Correspondence: Renying Zhuo,
| | - Xiaohua Yao
- State Key Laboratory of Tree Genetics and Breeding, Chinese Academy of Forestry, Hangzhou, China
- Key Laboratory of Tree Breeding of Zhejiang Province, Research Institute of Subtropical Forestry, Chinese Academy of Forestry, Hangzhou, China
- Xiaohua Yao,
| |
Collapse
|
13
|
Wang H, Liu W, Yu B, Yu X, Chen B. Identification of Key Modules and Hub Genes of Annulus Fibrosus in Intervertebral Disc Degeneration. Front Genet 2021; 11:596174. [PMID: 33584795 PMCID: PMC7875098 DOI: 10.3389/fgene.2020.596174] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 12/17/2020] [Indexed: 11/24/2022] Open
Abstract
Background: Intervertebral disc degeneration impairs the quality of patients lives. Even though there has been development of many therapeutic strategies, most of them remain unsatisfactory due to the limited understanding of the mechanisms that underlie the intervertebral disc degeneration. Questions/purposes: This study is meant to identify the key modules and hub genes related to the annulus fibrosus in intervertebral disc degeneration (IDD) through: (1) constructing a weighted gene co-expression network; (2) identifying key modules and hub genes; (3) verifying the relationships of key modules and hub genes with IDD; and (4) confirming the expression pattern of hub genes in clinical samples. Methods: The Gene Expression Omnibus provided 24 sets of annulus fibrosus microarray data. Differentially expressed genes between the annulus fibrosus of degenerative and non-degenerative intervertebral disc samples have gone through the Gene Ontology (GO) and pathway analysis. The construction of a gene network and classification of genes into different modules were conducted through performing Weighted Gene Co-expression Network Analysis. The identification of modules and hub genes that were most related to intervertebral disc degeneration was proceeded. In order to verify the relationships of the module and hub genes with intervertebral disc degeneration, Ingenuity Pathway Analysis was operated. Clinical samples were adopted to help verify the hub gene expression profile. Results: One thousand one hundred ninety differentially expressed genes were identified. Terms and pathways associated with intervertebral disc degeneration were presented by GO and pathway analysis. The construction of a Weighted Gene Coexpression Network was completed and clustering differentially expressed genes into four modules was also achieved. The module with the lowest P-value and the highest absolute correlation coefficient was selected and its relationship with intervertebral disc degeneration was confirmed by Ingenuity Pathway Analysis. The identification of hub genes and the confirmation of their expression profile were also realized. Conclusions: This study generated a comprehensive overview of the gene networks underlying annulus fibrosus in intervertebral disc degeneration. Clinical Relevance: Modules and hub genes identified in this study are highly associated with intervertebral disc degeneration, and may serve as potential therapeutic targets for intervertebral disc degeneration.
Collapse
Affiliation(s)
- Hantao Wang
- Department of Spine Surgery, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
- Department of Orthopedics, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Wenhui Liu
- Plastic & Reconstructive Surgery of the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bo Yu
- Department of Medicine, Lincoln Medical Center, Bronx, NY, United States
| | - Xiaosheng Yu
- Department of Spine Surgery, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Bin Chen
- Department of Spine Surgery, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
14
|
Li HJ, Goff A, Rudzinskas SA, Jung Y, Dubey N, Hoffman J, Hipolito D, Mazzu M, Rubinow DR, Schmidt PJ, Goldman D. Altered estradiol-dependent cellular Ca 2+ homeostasis and endoplasmic reticulum stress response in Premenstrual Dysphoric Disorder. Mol Psychiatry 2021; 26:6963-6974. [PMID: 34035477 PMCID: PMC8613306 DOI: 10.1038/s41380-021-01144-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 04/11/2021] [Accepted: 04/21/2021] [Indexed: 02/04/2023]
Abstract
Premenstrual Dysphoric Disorder (PMDD) is characterized by debilitating mood symptoms in the luteal phase of the menstrual cycle. Prior studies of affected women have implicated a differential response to ovarian steroids. However, the molecular basis of these patients' differential response to hormone remains poorly understood. We performed transcriptomic analyses of lymphoblastoid cell lines (LCLs) derived from women with PMDD and asymptomatic controls cultured under untreated (steroid-free), estradiol-treated (E2), and progesterone-treated (P4) conditions. Weighted gene correlation network analysis (WGCNA) of transcriptomes identified four gene modules with significant diagnosis x hormone interactions, including one enriched for neuronal functions. Next, in a gene-level analysis comparing transcriptional response to hormone across diagnoses, a generalized linear model identified 1522 genes differentially responsive to E2 (E2-DRGs). Among the top 10 E2-DRGs was a physically interacting network (NUCB1, DST, GCC2, GOLGB1) involved in endoplasmic reticulum (ER)-Golgi function. qRT-PCR validation reproduced a diagnosis x E2 interaction (F(1,24)=7.01, p = 0.014) for NUCB1, a regulator of cellular Ca2+ and ER stress. Finally, we used a thapsigargin (Tg) challenge assay to test whether E2 induces differences in Ca2+ homeostasis and ER stress response in PMDD. PMDD LCLs had a 1.36-fold decrease in Tg-induced XBP1 splicing response compared to controls, and a 1.62-fold decreased response (p = 0.005), with a diagnosis x treatment interaction (F(3,33)=3.51, p = 0.026) in the E2-exposed condition. Altered hormone-dependent in cellular Ca2+ dynamics and ER stress may contribute to the pathophysiology of PMDD.
Collapse
Affiliation(s)
- Howard J. Li
- grid.47100.320000000419368710Dept. of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, CT USA ,grid.416868.50000 0004 0464 0574Section on Behavioral Endocrinology, National Institute of Mental Health, NIH, Bethesda, MD USA
| | - Allison Goff
- grid.420085.b0000 0004 0481 4802Laboratory of Neurogenetics, National Institute of Alcohol Abuse and Alcoholism, NIH, Bethesda, MD USA
| | - Sarah A. Rudzinskas
- grid.416868.50000 0004 0464 0574Section on Behavioral Endocrinology, National Institute of Mental Health, NIH, Bethesda, MD USA
| | - Yonwoo Jung
- grid.420085.b0000 0004 0481 4802Laboratory of Neurogenetics, National Institute of Alcohol Abuse and Alcoholism, NIH, Bethesda, MD USA
| | - Neelima Dubey
- grid.416868.50000 0004 0464 0574Section on Behavioral Endocrinology, National Institute of Mental Health, NIH, Bethesda, MD USA
| | - Jessica Hoffman
- grid.416868.50000 0004 0464 0574Section on Behavioral Endocrinology, National Institute of Mental Health, NIH, Bethesda, MD USA
| | - Dion Hipolito
- grid.420085.b0000 0004 0481 4802Laboratory of Neurogenetics, National Institute of Alcohol Abuse and Alcoholism, NIH, Bethesda, MD USA
| | - Maria Mazzu
- grid.416868.50000 0004 0464 0574Section on Behavioral Endocrinology, National Institute of Mental Health, NIH, Bethesda, MD USA
| | - David R. Rubinow
- grid.410711.20000 0001 1034 1720Dept. of Psychiatry, University of North Carolina, Chapel Hill, NC USA
| | - Peter J. Schmidt
- grid.416868.50000 0004 0464 0574Section on Behavioral Endocrinology, National Institute of Mental Health, NIH, Bethesda, MD USA
| | - David Goldman
- grid.420085.b0000 0004 0481 4802Laboratory of Neurogenetics, National Institute of Alcohol Abuse and Alcoholism, NIH, Bethesda, MD USA
| |
Collapse
|
15
|
Zhang ZQ, Wu WW, Chen JD, Zhang GY, Lin JY, Wu YK, Zhang Y, Su YA, Li JT, Si TM. Weighted Gene Coexpression Network Analysis Reveals Essential Genes and Pathways in Bipolar Disorder. Front Psychiatry 2021; 12:553305. [PMID: 33815158 PMCID: PMC8010671 DOI: 10.3389/fpsyt.2021.553305] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 02/24/2021] [Indexed: 11/13/2022] Open
Abstract
Bipolar disorder (BD) is a major and highly heritable mental illness with severe psychosocial impairment, but its etiology and pathogenesis remains unclear. This study aimed to identify the essential pathways and genes involved in BD using weighted gene coexpression network analysis (WGCNA), a bioinformatic method studying the relationships between genes and phenotypes. Using two available BD gene expression datasets (GSE5388, GSE5389), we constructed a gene coexpression network and identified modules related to BD. The analyses of Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathways were performed to explore functional enrichment of the candidate modules. A protein-protein interaction (PPI) network was further constructed to identify the potential hub genes. Ten coexpression modules were identified from the top 5,000 genes in 77 samples and three modules were significantly associated with BD, which were involved in several biological processes (e.g., the actin filament-based process) and pathways (e.g., MAPK signaling). Four genes (NOTCH1, POMC, NGF, and DRD2) were identified as candidate hub genes by PPI analysis and CytoHubba. Finally, we carried out validation analyses in a separate dataset, GSE12649, and verified NOTCH1 as a hub gene and the involvement of several biological processes such as actin filament-based process and axon development. Taken together, our findings revealed several candidate pathways and genes (NOTCH1) in the pathogenesis of BD and call for further investigation for their potential research values in BD diagnosis and treatment.
Collapse
Affiliation(s)
- Zhen-Qing Zhang
- Xiamen Xianyue Hospital, Xiamen, China.,Peking University Sixth Hospital, Peking University Institute of Mental Health, Peking University, Beijing, China
| | | | | | - Guang-Yin Zhang
- Department of Psychosomatic Medicine, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jing-Yu Lin
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Peking University, Beijing, China
| | - Yan-Kun Wu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Peking University, Beijing, China
| | - Yu Zhang
- Institute of Mental Health, Hebei North University, Hebei, China
| | - Yun-Ai Su
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Peking University, Beijing, China
| | - Ji-Tao Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Peking University, Beijing, China
| | - Tian-Mei Si
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Peking University, Beijing, China
| |
Collapse
|
16
|
Li R, Wang Q, Qiu Y, Meng Y, Wei L, Wang H, Mo R, Zou D, Liu C. A Potential Autophagy-Related Competing Endogenous RNA Network and Corresponding Diagnostic Efficacy in Schizophrenia. Front Psychiatry 2021; 12:628361. [PMID: 33708146 PMCID: PMC7940829 DOI: 10.3389/fpsyt.2021.628361] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 02/02/2021] [Indexed: 12/25/2022] Open
Abstract
Competing endogenous RNA (ceRNA) and autophagy were related to neurological diseases. But the relationship among ceRNA, autophagy and Schizophrenia (SZ) was not clear. In this study, we obtained gene expression profile of SZ patients (GSE38484, GSE54578, and GSE16930) from Gene Expression Omnibus (GEO) database. Then we screened the autophagy-related differentially expressed lncRNA, miRNA, and mRNA (DElncRNA, DEmiRNA, and DEmRNA) combined with Gene database from The National Center for Biotechnology Information (NCBI). In addition, we performed enrichment analysis. The result showed that biological processes (BPs) mainly were associated with cellular responses to oxygen concentration. The enriched pathways mainly included ErbB, AMPK, mTOR signaling pathway and cell cycle. Furthermore, we constructed autophagy-related ceRNA network based on the TargetScan database. Moreover, we explored the diagnostic efficiency of lncRNA, miRNA and mRNA in ceRNA, through gene set variation analysis (GSVA). The result showed that the diagnostic efficiency was robust, especially miRNA (AUC = 0.884). The miRNA included hsa-miR-423-5p, hsa-miR-4532, hsa-miR-593-3p, hsa-miR-618, hsa-miR-4723-3p, hsa-miR-4640-3p, hsa-miR-296-5p, and hsa-miR-3943. The result of this study may be helpful for deepening the pathophysiology of SZ. In addition, our finding may provide a guideline for the clinical diagnosis of SZ.
Collapse
Affiliation(s)
- Rongjie Li
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, China.,Department of Internal Medicine, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Qiaoye Wang
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, China.,Department of Internal Medicine, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yufen Qiu
- Maternal and Child Health Hospital and Obstetrics and Gynecology Hospital of Guangxi Zhuang Autonomous Region, Guangxi, China
| | - Youshi Meng
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, China.,Department of Internal Medicine, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Lei Wei
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, China.,Department of Internal Medicine, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Hao Wang
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, China.,Department of Internal Medicine, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ruikang Mo
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, China.,Department of Internal Medicine, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Donghua Zou
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, China.,Department of Internal Medicine, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Chunbin Liu
- Department of Internal Medicine, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
17
|
Hathy E, Szabó E, Varga N, Erdei Z, Tordai C, Czehlár B, Baradits M, Jezsó B, Koller J, Nagy L, Molnár MJ, Homolya L, Nemoda Z, Apáti Á, Réthelyi JM. Investigation of de novo mutations in a schizophrenia case-parent trio by induced pluripotent stem cell-based in vitro disease modeling: convergence of schizophrenia- and autism-related cellular phenotypes. Stem Cell Res Ther 2020; 11:504. [PMID: 33246498 PMCID: PMC7694414 DOI: 10.1186/s13287-020-01980-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 10/18/2020] [Indexed: 12/30/2022] Open
Abstract
Background De novo mutations (DNMs) have been implicated in the etiology of schizophrenia (SZ), a chronic debilitating psychiatric disorder characterized by hallucinations, delusions, cognitive dysfunction, and decreased community functioning. Several DNMs have been identified by examining SZ cases and their unaffected parents; however, in most cases, the biological significance of these mutations remains elusive. To overcome this limitation, we have developed an approach of using induced pluripotent stem cell (iPSC) lines from each member of a SZ case-parent trio, in order to investigate the effects of DNMs in cellular progenies of interest, particularly in dentate gyrus neuronal progenitors. Methods We identified a male SZ patient characterized by early disease onset and negative symptoms, who is a carrier of 3 non-synonymous DNMs in genes LRRC7, KHSRP, and KIR2DL1. iPSC lines were generated from his and his parents’ peripheral blood mononuclear cells using Sendai virus-based reprogramming and differentiated into neuronal progenitor cells (NPCs) and hippocampal dentate gyrus granule cells. We used RNASeq to explore transcriptomic differences and calcium (Ca2+) imaging, cell proliferation, migration, oxidative stress, and mitochondrial assays to characterize the investigated NPC lines. Results NPCs derived from the SZ patient exhibited transcriptomic differences related to Wnt signaling, neuronal differentiation, axonal guidance and synaptic function, and decreased Ca2+ reactivity to glutamate. Moreover, we could observe increased cellular proliferation and alterations in mitochondrial quantity and morphology. Conclusions The approach of reprograming case-parent trios represents an opportunity for investigating the molecular effects of disease-causing mutations and comparing these in cell lines with reduced variation in genetic background. Our results are indicative of a partial overlap between schizophrenia and autism-related phenotypes in the investigated family. Limitations Our study investigated only one family; therefore, the generalizability of findings is limited. We could not derive iPSCs from two other siblings to test for possible genetic effects in the family that are not driven by DNMs. The transcriptomic and functional assays were limited to the NPC stage, although these variables should also be investigated at the mature neuronal stage.
Collapse
Affiliation(s)
- Edit Hathy
- National Brain Research Project (NAP) Molecular Psychiatry Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
| | - Eszter Szabó
- Molecular Cell Biology Research Group, Institute of Enzymology, Research Center for Natural Sciences, 1117 Magyar tudósok körútja 2, Budapest, Hungary
| | - Nóra Varga
- Molecular Cell Biology Research Group, Institute of Enzymology, Research Center for Natural Sciences, 1117 Magyar tudósok körútja 2, Budapest, Hungary
| | - Zsuzsa Erdei
- Molecular Cell Biology Research Group, Institute of Enzymology, Research Center for Natural Sciences, 1117 Magyar tudósok körútja 2, Budapest, Hungary
| | - Csongor Tordai
- National Brain Research Project (NAP) Molecular Psychiatry Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
| | - Boróka Czehlár
- National Brain Research Project (NAP) Molecular Psychiatry Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
| | - Máté Baradits
- National Brain Research Project (NAP) Molecular Psychiatry Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
| | - Bálint Jezsó
- Molecular Cell Biology Research Group, Institute of Enzymology, Research Center for Natural Sciences, 1117 Magyar tudósok körútja 2, Budapest, Hungary
| | - Júlia Koller
- Institute of Rare Disorders and Genomic Medicine, Semmelweis University, Budapest, Hungary
| | - László Nagy
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Mária Judit Molnár
- Institute of Rare Disorders and Genomic Medicine, Semmelweis University, Budapest, Hungary
| | - László Homolya
- Molecular Cell Biology Research Group, Institute of Enzymology, Research Center for Natural Sciences, 1117 Magyar tudósok körútja 2, Budapest, Hungary
| | - Zsófia Nemoda
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Ágota Apáti
- Molecular Cell Biology Research Group, Institute of Enzymology, Research Center for Natural Sciences, 1117 Magyar tudósok körútja 2, Budapest, Hungary.
| | - János M Réthelyi
- National Brain Research Project (NAP) Molecular Psychiatry Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary. .,Department of Psychiatry and Psychotherapy, Semmelweis University, Balassa utca 6, Budapest, 1083, Hungary.
| |
Collapse
|
18
|
Widespread transcriptional disruption of the microRNA biogenesis machinery in brain and peripheral tissues of individuals with schizophrenia. Transl Psychiatry 2020; 10:376. [PMID: 33149139 PMCID: PMC7642431 DOI: 10.1038/s41398-020-01052-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 09/16/2020] [Accepted: 10/01/2020] [Indexed: 12/17/2022] Open
Abstract
In schizophrenia, altered transcription in brain and peripheral tissues may be due to altered expression of the microRNA biogenesis machinery genes. In this study, we explore the expression of these genes both at the cerebral and peripheral levels. We used shinyGEO application to analyze gene expression from ten Gene Expression Omnibus datasets, in order to perform differential expression analyses for eight genes encoding the microRNA biogenesis machinery. First, we compared expression of the candidate genes between control subjects and individuals with schizophrenia in postmortem cerebral samples from seven different brain regions. Then, we compared the expression of the candidate genes between control subjects and individuals with schizophrenia in three peripheral tissues. In brain and peripheral tissues of individuals with schizophrenia, we report distinct altered expression patterns of the microRNA biogenesis machinery genes. In the dorsolateral prefrontal cortex, associative striatum and cerebellum of individuals with schizophrenia, we observed an overexpression pattern of some candidate genes suggesting a heightened miRNA production in these brain regions. Additionally, mixed transcriptional abnormalities were identified in the hippocampus. Moreover, in the blood and olfactory epithelium of individuals with schizophrenia, we observed distinct aberrant transcription patterns of the candidate genes. Remarkably, in individuals with schizophrenia, we report DICER1 overexpression in the dorsolateral prefrontal cortex, hippocampus and cerebellum as well as a congruent DICER1 upregulation in the blood compartment suggesting that it may represent a peripheral marker. Transcriptional disruption of the miRNA biogenesis machinery may contribute to schizophrenia pathogenesis both in brain and peripheral tissues.
Collapse
|
19
|
Li X, Wang C, Zhang X, Liu J, Wang Y, Li C, Guo D. Weighted gene co-expression network analysis revealed key biomarkers associated with the diagnosis of hypertrophic cardiomyopathy. Hereditas 2020; 157:42. [PMID: 33099311 PMCID: PMC7585681 DOI: 10.1186/s41065-020-00155-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 10/16/2020] [Indexed: 12/12/2022] Open
Abstract
Objective To reveal the molecular mechanism underlying the pathogenesis of HCM and find new effective therapeutic strategies using a systematic biological approach. Methods The WGCNA algorithm was applied to building the co-expression network of HCM samples. A sample cluster analysis was performed using the hclust tool and a co-expression module was constructed. The WGCNA algorithm was used to study the interactive connection between co-expression modules and draw a heat map to show the strength of interactions between modules. The genetic information of the respective modules was mapped to the associated GO terms and KEGG pathways, and the Hub Genes with the highest connectivity in each module were identified. The Wilcoxon test was used to verify the expression level of hub genes between HCM and normal samples, and the “pROC” R package was used to verify the possibility of hub genes as biomarkers. Finally, the potential functions of hub genes were analyzed by GSEA software. Results Seven co-expression modules were constructed using sample clustering analysis. GO and KEGG enrichment analysis judged that the turquoise module is an important module. The hub genes of each module are RPL35A for module Black, FH for module Blue, PREI3 for module Brown, CREB1 for module Green, LOC641848 for module Pink, MYH7 for module Turquoise and MYL6 for module Yellow. The results of the differential expression analysis indicate that MYH7 and FH are considered true hub genes. In addition, the ROC curves revealed their high diagnostic value as biomarkers for HCM. Finally, in the results of the GSEA analysis, MYH7 and FH highly expressed genes were enriched with the “proteasome” and a “PPAR signaling pathway,” respectively. Conclusions The MYH7 and FH genes may be the true hub genes of HCM. Their respective enriched pathways, namely the “proteasome” and the “PPAR signaling pathway,” may play an important role in the development of HCM.
Collapse
Affiliation(s)
- Xin Li
- Department of Cardiovascular, The Third Central Hospital of Tianjin, Tianjin, China
| | - Chenxin Wang
- Department of Respiratory medicine, The Third Central Hospital of Tianjin, Tianjin, China
| | - Xiaoqing Zhang
- Department of internal medicine, Affiliated Hospital of Nankai University, Tianjin, China
| | - Jiali Liu
- Department of Hematology, Taian City Central Hospital, 29 Longtan Road, Taian, 271000, Shandong, China
| | - Yu Wang
- Department of Cardiovascular, The Third Central Hospital of Tianjin, Tianjin, China
| | - Chunpu Li
- Department of Orthopedics, Taian City Central Hospital, 29 Longtan Road, Taian, 271000, Shandong, China.
| | - Dongmei Guo
- Department of Hematology, Taian City Central Hospital, 29 Longtan Road, Taian, 271000, Shandong, China.
| |
Collapse
|
20
|
Chen J, Cao H, Kaufmann T, Westlye LT, Tost H, Meyer-Lindenberg A, Schwarz E. Identification of Reproducible BCL11A Alterations in Schizophrenia Through Individual-Level Prediction of Coexpression. Schizophr Bull 2020; 46:1165-1171. [PMID: 32232389 PMCID: PMC7505190 DOI: 10.1093/schbul/sbaa047] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Previous studies have provided evidence for an alteration of genetic coexpression in schizophrenia (SCZ). However, such analyses have thus far lacked biological specificity for individual genes, which may be critical for identifying illness-relevant effects. Therefore, we applied machine learning to identify gene-specific coexpression differences at the individual subject level and compared these between individuals with SCZ, bipolar disorder, major depressive disorder (MDD), autism spectrum disorder (ASD), and healthy controls. Utilizing transcriptome-wide gene expression data from 21 independent datasets, comprising a total of 9509 participants, we identified a reproducible decrease of BCL11A coexpression across 4 SCZ datasets that showed diagnostic specificity for SCZ when compared with ASD and MDD. We further demonstrate that individual-level coexpression differences can be combined in multivariate coexpression scores that show reproducible illness classification across independent datasets in SCZ and ASD. This study demonstrates that machine learning can capture gene-specific coexpression differences at the individual subject level for SCZ and identify novel biomarker candidates.
Collapse
Affiliation(s)
- Junfang Chen
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Han Cao
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Tobias Kaufmann
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Lars T Westlye
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Heike Tost
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Andreas Meyer-Lindenberg
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Emanuel Schwarz
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
21
|
Duke CG, Bach SV, Revanna JS, Sultan FA, Southern NT, Davis MN, Carullo NVN, Bauman AJ, Phillips RA, Day JJ. An Improved CRISPR/dCas9 Interference Tool for Neuronal Gene Suppression. Front Genome Ed 2020; 2:9. [PMID: 34713218 PMCID: PMC8525373 DOI: 10.3389/fgeed.2020.00009] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 08/03/2020] [Indexed: 01/01/2023] Open
Abstract
The expression of genetic material governs brain development, differentiation, and function, and targeted manipulation of gene expression is required to understand contributions of gene function to health and disease states. Although recent improvements in CRISPR/dCas9 interference (CRISPRi) technology have enabled targeted transcriptional repression at selected genomic sites, integrating these techniques for use in non-dividing neuronal systems remains challenging. Previously, we optimized a dual lentivirus expression system to express CRISPR-based activation machinery in post-mitotic neurons. Here we used a similar strategy to adapt an improved dCas9-KRAB-MeCP2 repression system for robust transcriptional inhibition in neurons. We find that lentiviral delivery of a dCas9-KRAB-MeCP2 construct driven by the neuron-selective human synapsin promoter enabled transgene expression in primary rat neurons. Next, we demonstrate transcriptional repression using CRISPR sgRNAs targeting diverse gene promoters, and show superiority of this system in neurons compared to existing RNA interference methods for robust transcript specific manipulation at the complex Brain-derived neurotrophic factor (Bdnf) gene. Our findings advance this improved CRISPRi technology for use in neuronal systems for the first time, potentially enabling improved ability to manipulate gene expression states in the nervous system.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Jeremy J. Day
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
22
|
Integrated Systems Analysis Explores Dysfunctional Molecular Modules and Regulatory Factors in Children with Autism Spectrum Disorder. J Mol Neurosci 2020; 71:358-368. [PMID: 32653993 DOI: 10.1007/s12031-020-01658-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 07/03/2020] [Indexed: 12/22/2022]
Abstract
Autism spectrum disorder (ASD) is a genetic neurodevelopmental disorder involving multiple genes that occurs in early childhood, and a number of risk genes have been reported in previous studies. However, the molecular mechanism of the polygenic regulation leading to pathological changes in ASD remains unclear. First, we identified 8 dysregulated gene coexpression modules by analyzing blood transcriptome data from 96 children with ASD and 42 controls. These modules are rich in ASD risk genes and function related to metabolism, immunity, neurodevelopment, and signaling. The regulatory factors of each module including microRNA (miRNA) and transcription factors (TFs) were subsequently predicted based on transcriptional and posttranscriptional regulation. We identified a set of miRNAs that regulate metabolic and immune modules, as well as transcription factors that cause dysregulation of the modules, and we constructed a coregulatory network between the regulatory factors and modules. Our work reveals dysfunctional modules in children with ASD, elucidates the role of miRNA and transcription factor dysregulation in the pathophysiology of ASD, and helps us to further understand the underlying molecular mechanism of ASD.
Collapse
|
23
|
Jiao M, Li J, Zhang Q, Xu X, Li R, Dong P, Meng C, Li Y, Wang L, Qi W, Kang K, Wang H, Wang T. Identification of Four Potential Biomarkers Associated With Coronary Artery Disease in Non-diabetic Patients by Gene Co-expression Network Analysis. Front Genet 2020; 11:542. [PMID: 32714363 PMCID: PMC7344232 DOI: 10.3389/fgene.2020.00542] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 05/05/2020] [Indexed: 12/17/2022] Open
Abstract
Background Coronary artery disease (CAD) is a type of cardiovascular disease that greatly hurts the health of human beings. Diabetic status is one of the largest clinical factors affecting CAD-associated gene expression changes. Most of the studies focus on diabetic patients, whereas few have been done for non-diabetic patients. Since the pathophysiological processes may vary among these patients, we cannot simply follow the standard based on the data from diabetic patients. Therefore, the prognostic and predictive diagnostic biomarkers for CAD in non-diabetic patient need to be fully recognized. Materials and Methods To screen out candidate genes associated with CAD in non-diabetic patients, weighted gene co-expression network analysis (WGCNA) was constructed to conduct an analysis of microarray expression profiling in patients with CAD. First, the microarray data GSE20680 and GSE20681 were downloaded from NCBI. We constructed co-expression modules via WGCNA after excluding the diabetic patients. As a result, 18 co-expression modules were screened out, including 1,225 differentially expressed genes (DEGs) that were obtained from 152 patients (luminal stenosis ≥50% in at least one major vessel) and 170 patients (stenosis of <50%). Subsequently, a Pearson's correlation analysis was conducted between the modules and clinical traits. Then, a functional enrichment analysis was conducted, and we used gene network analysis to reveal hub genes. Last, we validated the hub genes with peripheral blood samples in an independent patient cohort using RT-qPCR. Results The results showed that the midnight blue module and the yellow module played vital roles in the pathogenesis of CAD in non-diabetic patients. Additionally, CD40, F11R, TNRC18, and calcium/calmodulin-dependent protein kinase type II gamma (CAMK2G) were screened out and validated using enzyme-linked immunosorbent assay (ELISA) in an independent patient cohort and immunohistochemical (IHC) staining in an atherosclerosis mouse model. Conclusion Our findings demonstrate that hub genes, CD40, F11R, TNRC18, and CAMK2G, are surrogate diagnostic biomarkers and/or therapeutic targets for CAD in non-diabetic patients and require deeper validation.
Collapse
Affiliation(s)
- Min Jiao
- Department of Cardiology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Jingtian Li
- Department of Cardiology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Quan Zhang
- Department of Cardiology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Xiufeng Xu
- Department of Neurology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Ruidong Li
- Graduate Program in Genetics, Genomics, and Bioinformatics, University of California, Riverside, Riverside, CA, United States
| | - Peikang Dong
- Department of Cardiology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Chun Meng
- Department of Cardiology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Yi Li
- Department of Cardiology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Lijuan Wang
- Department of Cardiology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Wanpeng Qi
- Department of Cardiology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Kai Kang
- Department of Cardiology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Hongjie Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Wang
- Department of Cardiology, Affiliated Hospital of Weifang Medical University, Weifang, China
| |
Collapse
|
24
|
Gysi DM, Nowick K. Construction, comparison and evolution of networks in life sciences and other disciplines. J R Soc Interface 2020; 17:20190610. [PMID: 32370689 PMCID: PMC7276545 DOI: 10.1098/rsif.2019.0610] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 04/09/2020] [Indexed: 12/12/2022] Open
Abstract
Network approaches have become pervasive in many research fields. They allow for a more comprehensive understanding of complex relationships between entities as well as their group-level properties and dynamics. Many networks change over time, be it within seconds or millions of years, depending on the nature of the network. Our focus will be on comparative network analyses in life sciences, where deciphering temporal network changes is a core interest of molecular, ecological, neuropsychological and evolutionary biologists. Further, we will take a journey through different disciplines, such as social sciences, finance and computational gastronomy, to present commonalities and differences in how networks change and can be analysed. Finally, we envision how borrowing ideas from these disciplines could enrich the future of life science research.
Collapse
Affiliation(s)
- Deisy Morselli Gysi
- Department of Computer Science, Interdisciplinary Center of Bioinformatics, University of Leipzig, 04109 Leipzig, Germany
- Swarm Intelligence and Complex Systems Group, Faculty of Mathematics and Computer Science, University of Leipzig, 04109 Leipzig, Germany
- Center for Complex Networks Research, Northeastern University, 177 Huntington Avenue, Boston, MA 02115, USA
| | - Katja Nowick
- Human Biology Group, Institute for Biology, Faculty of Biology, Chemistry, Pharmacy, Freie Universität Berlin, Königin-Luise-Straβe 1-3, 14195 Berlin, Germany
| |
Collapse
|
25
|
Grant P, Gabriel F, Kuepper Y, Wielpuetz C, Hennig J. Psychosis-proneness correlates with expression levels of dopaminergic genes. Eur Psychiatry 2020; 29:304-6. [DOI: 10.1016/j.eurpsy.2013.12.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 12/04/2013] [Accepted: 12/17/2013] [Indexed: 02/07/2023] Open
Abstract
AbstractPsychosis-proneness or schizotypy is a personality organisation mirroring individual risk for schizophrenia-development. Believed to be a fully dimensional construct sharing considerable geno- and phenotypal variance with clinical schizophrenia, it has become an increasingly promising tool for basic psychosis-research. Although many studies show genetic commonalities between schizotypy and schizophrenia, changes in regulation of gene expression have never been examined in schizotypy before. We therefore extracted RNA from the blood, a valid surrogate for brain tissue, of a large sample of 67 healthy male volunteers and correlated the activities of all genes relevant for dopaminergic neurotransmission with the positive schizotypy-scale of the O-LIFE. We found significant negative correlations regarding the expression of the genes COMT, MAOB, DRD4, DRD5 and FOS, indicating that increased schizotypy coincides with higher levels of dopaminergic dysregulation on the mRNA-level. Considering the advantages of this method, we suggest that it be applied more often in fundamental psychosis-research.
Collapse
|
26
|
Patel H, Iniesta R, Stahl D, Dobson RJ, Newhouse SJ. Working Towards a Blood-Derived Gene Expression Biomarker Specific for Alzheimer's Disease. J Alzheimers Dis 2020; 74:545-561. [PMID: 32065794 PMCID: PMC7175937 DOI: 10.3233/jad-191163] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2020] [Indexed: 11/15/2022]
Abstract
BACKGROUND The typical approach to identify blood-derived gene expression signatures as a biomarker for Alzheimer's disease (AD) have relied on training classification models using AD and healthy controls only. This may inadvertently result in the identification of markers for general illness rather than being disease-specific. OBJECTIVE Investigate whether incorporating additional related disorders in the classification model development process can lead to the discovery of an AD-specific gene expression signature. METHODS Two types of XGBoost classification models were developed. The first used 160 AD and 127 healthy controls and the second used the same 160 AD with 6,318 upsampled mixed controls consisting of Parkinson's disease, multiple sclerosis, amyotrophic lateral sclerosis, bipolar disorder, schizophrenia, coronary artery disease, rheumatoid arthritis, chronic obstructive pulmonary disease, and cognitively healthy subjects. Both classification models were evaluated in an independent cohort consisting of 127 AD and 687 mixed controls. RESULTS The AD versus healthy control models resulted in an average 48.7% sensitivity (95% CI = 34.7-64.6), 41.9% specificity (95% CI = 26.8-54.3), 13.6% PPV (95% CI = 9.9-18.5), and 81.1% NPV (95% CI = 73.3-87.7). In contrast, the mixed control models resulted in an average of 40.8% sensitivity (95% CI = 27.5-52.0), 95.3% specificity (95% CI = 93.3-97.1), 61.4% PPV (95% CI = 53.8-69.6), and 89.7% NPV (95% CI = 87.8-91.4). CONCLUSIONS This early work demonstrates the value of incorporating additional related disorders into the classification model developmental process, which can result in models with improved ability to distinguish AD from a heterogeneous aging population. However, further improvement to the sensitivity of the test is still required.
Collapse
Affiliation(s)
- Hamel Patel
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
- NIHR BioResource Centre Maudsley, NIHR Maudsley Biomedical Research Centre (BRC) at South London and Maudsley NHS Foundation Trust (SLaM) & Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, UK
| | - Raquel Iniesta
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Daniel Stahl
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Richard J.B. Dobson
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
- NIHR BioResource Centre Maudsley, NIHR Maudsley Biomedical Research Centre (BRC) at South London and Maudsley NHS Foundation Trust (SLaM) & Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, UK
- Health Data Research UK London, University College London, London, UK
- Institute of Health Informatics, University College London, London, UK
- The National Institute for Health Research University College London Hospitals Biomedical Research Centre, University College London, London, UK
| | - Stephen J. Newhouse
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
- NIHR BioResource Centre Maudsley, NIHR Maudsley Biomedical Research Centre (BRC) at South London and Maudsley NHS Foundation Trust (SLaM) & Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, UK
- Health Data Research UK London, University College London, London, UK
- Institute of Health Informatics, University College London, London, UK
- The National Institute for Health Research University College London Hospitals Biomedical Research Centre, University College London, London, UK
| |
Collapse
|
27
|
Bongen E, Lucian H, Khatri A, Fragiadakis GK, Bjornson ZB, Nolan GP, Utz PJ, Khatri P. Sex Differences in the Blood Transcriptome Identify Robust Changes in Immune Cell Proportions with Aging and Influenza Infection. Cell Rep 2019; 29:1961-1973.e4. [PMID: 31722210 PMCID: PMC6856718 DOI: 10.1016/j.celrep.2019.10.019] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 07/12/2019] [Accepted: 10/03/2019] [Indexed: 02/09/2023] Open
Abstract
Sex differences in autoimmunity and infection suggest that a better understanding of molecular sex differences will improve the diagnosis and treatment of immune-related disease. We identified 144 differentially expressed genes, referred to as immune sex expression signature (iSEXS), between human males and females using an integrated multi-cohort analysis of blood transcriptome profiles from six discovery cohorts from five continents with 458 healthy individuals. We validated iSEXS in 11 additional cohorts of 524 peripheral blood samples. When we separated iSEXS into genes located on sex chromosomes (XY-iSEXS) or autosomes (autosomal-iSEXS), both modules distinguished males and females. iSEXS reflects sex differences in immune cell proportions, with female-associated genes showing higher expression by CD4+ T cells and male-associated genes showing higher expression by myeloid cells. Autosomal-iSEXS detected an increase in monocytes with age in females, reflected sex-differential immune cell dynamics during influenza infection, and predicted antibody response in males, but not females.
Collapse
Affiliation(s)
- Erika Bongen
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Program in Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Haley Lucian
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Avani Khatri
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Gabriela K Fragiadakis
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Baxter Laboratory for Stem Cell Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Zachary B Bjornson
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Baxter Laboratory for Stem Cell Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Garry P Nolan
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Baxter Laboratory for Stem Cell Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Paul J Utz
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Purvesh Khatri
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Biomedical Informatics Research, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
28
|
Abstract
Until recently, advances in understanding the genetic architecture of psychiatric disorders have been impeded by a historic, and often mandated, commitment to the use of traditional, and unvalidated, categorical diagnoses in isolation as the relevant phenotype. Such studies typically required lengthy structured interviews to delineate differences in the character and duration of behavioral symptomatology amongst disorders that were thought to be etiologic, and they were often underpowered as a result. Increasing acceptance of the fact that co-morbidity in psychiatric disorders is the rule rather than the exception has led to alternative designs in which shared dimensional symptomatology is analyzed as a quantitative trait and to association analyses in which combined polygenic risk scores are computationally compared across multiple traditional categorical diagnoses to identify both distinct and unique genetic and environmental elements. Increasing evidence that most mental disorders share many common genetic risk variants and environmental risk modifiers suggests that the broad spectrum of psychiatric pathology represents the pleiotropic display of a more limited series of pathologic events in neuronal development than was originally believed, regulated by many common risk variants and a smaller number of rare ones.
Collapse
Affiliation(s)
- Tova Fuller
- Deptartment of Psychiatry, UCSF Weill Institute for Neurosciences, University of California, San Francisco School of Medicine, San Francisco, CA, USA
| | - Victor Reus
- Deptartment of Psychiatry, UCSF Weill Institute for Neurosciences, University of California, San Francisco School of Medicine, San Francisco, CA, USA
| |
Collapse
|
29
|
Karunakaran KB, Chaparala S, Ganapathiraju MK. Potentially repurposable drugs for schizophrenia identified from its interactome. Sci Rep 2019; 9:12682. [PMID: 31481665 PMCID: PMC6722087 DOI: 10.1038/s41598-019-48307-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 07/11/2019] [Indexed: 12/13/2022] Open
Abstract
We previously presented the protein-protein interaction network of schizophrenia associated genes, and from it, the drug-protein interactome which showed the drugs that target any of the proteins in the interactome. Here, we studied these drugs further to identify whether any of them may potentially be repurposable for schizophrenia. In schizophrenia, gene expression has been described as a measurable aspect of the disease reflecting the action of risk genes. We studied each of the drugs from the interactome using the BaseSpace Correlation Engine, and shortlisted those that had a negative correlation with differential gene expression of schizophrenia. This analysis resulted in 12 drugs whose differential gene expression (drug versus normal) had an anti-correlation with differential expression for schizophrenia (disorder versus normal). Some of these drugs were already being tested for their clinical activity in schizophrenia and other neuropsychiatric disorders. Several proteins in the protein interactome of the targets of several of these drugs were associated with various neuropsychiatric disorders. The network of genes with opposite drug-induced versus schizophrenia-associated expression profiles were significantly enriched in pathways relevant to schizophrenia etiology and GWAS genes associated with traits or diseases that had a pathophysiological overlap with schizophrenia. Drugs that targeted the same genes as the shortlisted drugs, have also demonstrated clinical activity in schizophrenia and other related disorders. This integrated computational analysis will help translate insights from the schizophrenia drug-protein interactome to clinical research - an important step, especially in the field of psychiatric drug development which faces a high failure rate.
Collapse
Affiliation(s)
- Kalyani B Karunakaran
- Supercomputer Education and Research Centre, Indian Institute of Science, Indian Institute of Science, Bengaluru, India
| | | | - Madhavi K Ganapathiraju
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, USA.
- Intelligent Systems Program, University of Pittsburgh, Pittsburgh, USA.
| |
Collapse
|
30
|
Niu HM, Yang P, Chen HH, Hao RH, Dong SS, Yao S, Chen XF, Yan H, Zhang YJ, Chen YX, Jiang F, Yang TL, Guo Y. Comprehensive functional annotation of susceptibility SNPs prioritized 10 genes for schizophrenia. Transl Psychiatry 2019; 9:56. [PMID: 30705251 PMCID: PMC6355777 DOI: 10.1038/s41398-019-0398-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 12/27/2018] [Accepted: 01/10/2019] [Indexed: 12/18/2022] Open
Abstract
Nearly 95% of susceptibility SNPs identified by genome-wide association studies (GWASs) are located in non-coding regions, which causes a lot of difficulty in deciphering their biological functions on disease pathogenesis. Here, we aimed to conduct a comprehensive functional annotation for all the schizophrenia susceptibility loci obtained from GWASs. Considering varieties of epigenomic regulatory elements, we annotated all 22,688 acquired susceptibility SNPs according to their genomic positions to obtain functional SNPs. The comprehensive annotation indicated that these functional SNPs are broadly involved in diverse biological processes. Histone modification enrichment showed that H3K27ac, H3K36me3, H3K4me1, and H3K4me3 were related to the development of schizophrenia. Transcription factors (TFs) prediction, methylation quantitative trait loci (meQTL) analyses, expression quantitative trait loci (eQTL) analyses, and proteomic quantitative trait loci analyses (pQTL) identified 447 target protein-coding genes. Subsequently, differential expression analyses between schizophrenia cases and controls, nervous system phenotypes from mouse models, and protein-protein interaction with known schizophrenia-related pathways and genes were carried out with our target genes. We finaly prioritized 10 target genes for schizophrenia (CACNA1C, CLU, CSNK2B, GABBR1, GRIN2A, MAPK3, NOTCH4, SRR, TNF, and SYNGAP1). Our results may serve as an encyclopedia of schizophrenia susceptibility SNPs and offer holistic guides for post-GWAS functional experiments.
Collapse
Affiliation(s)
- Hui-Min Niu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, 710049, Xi'an, China
| | - Ping Yang
- Department of Psychiatry, Hunan Brain Hospital, Changsha, Hunan Province, China
| | - Huan-Huan Chen
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, 710049, Xi'an, China
| | - Ruo-Han Hao
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, 710049, Xi'an, China
| | - Shan-Shan Dong
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, 710049, Xi'an, China
| | - Shi Yao
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, 710049, Xi'an, China
| | - Xiao-Feng Chen
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, 710049, Xi'an, China
| | - Han Yan
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, 710049, Xi'an, China
| | - Yu-Jie Zhang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, 710049, Xi'an, China
| | - Yi-Xiao Chen
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, 710049, Xi'an, China
| | - Feng Jiang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, 710049, Xi'an, China
| | - Tie-Lin Yang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, 710049, Xi'an, China
| | - Yan Guo
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, 710049, Xi'an, China.
| |
Collapse
|
31
|
Huang P, Li F, Li L, You Y, Luo S, Dong Z, Gao Q, Wu S, Brünner N, Stenvang J. lncRNA profile study reveals the mRNAs and lncRNAs associated with docetaxel resistance in breast cancer cells. Sci Rep 2018; 8:17970. [PMID: 30568280 PMCID: PMC6299474 DOI: 10.1038/s41598-018-36231-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 11/13/2018] [Indexed: 02/07/2023] Open
Abstract
Resistance to adjuvant systemic treatment, including taxanes (docetaxel and paclitaxel) is a major clinical problem for breast cancer patients. lncRNAs (long non-coding RNAs) are non-coding transcripts, which have recently emerged as important players in a variety of biological processes, including cancer development and chemotherapy resistance. However, the contribution of lncRNAs to docetaxel resistance in breast cancer and the relationship between lncRNAs and taxane-resistance genes are still unclear. Here, we performed comprehensive RNA sequencing and analyses on two docetaxel-resistant breast cancer cell lines (MCF7-RES and MDA-RES) and their docetaxel-sensitive parental cell lines. We identified protein coding genes and pathways that may contribute to docetaxel resistance. More importantly, we identified lncRNAs that were consistently up-regulated or down-regulated in both the MCF7-RES and MDA-RES cells. The co-expression network and location analyses pinpointed four overexpressed lncRNAs located within or near the ABCB1 (ATP-binding cassette subfamily B member 1) locus, which might up-regulate the expression of ABCB1. We also identified the lncRNA EPB41L4A-AS2 (EPB41L4A Antisense RNA 2) as a potential biomarker for docetaxel sensitivity. These findings have improved our understanding of the mechanisms underlying docetaxel resistance in breast cancer and have provided potential biomarkers to predict the response to docetaxel in breast cancer patients.
Collapse
Affiliation(s)
- Peide Huang
- Section of Pharmacotherapy, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen N, Denmark
- BGI Genomics, BGI-Shenzhen, Shenzhen, 518083, China
- The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Fengyu Li
- BGI Genomics, BGI-Shenzhen, Shenzhen, 518083, China
| | - Lin Li
- BGI Genomics, BGI-Shenzhen, Shenzhen, 518083, China
| | - Yuling You
- BGI Genomics, BGI-Shenzhen, Shenzhen, 518083, China
| | - Shizhi Luo
- BGI Genomics, BGI-Shenzhen, Shenzhen, 518083, China
| | | | - Qiang Gao
- BGI Genomics, BGI-Shenzhen, Shenzhen, 518083, China
| | - Song Wu
- The Affiliated Luohu Hospital of Shenzhen University, Shenzhen Luohu Hospital Group, Shenzhen, China.
| | - Nils Brünner
- Section of Pharmacotherapy, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen N, Denmark.
| | - Jan Stenvang
- Section of Pharmacotherapy, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen N, Denmark.
| |
Collapse
|
32
|
Dopamine perturbation of gene co-expression networks reveals differential response in schizophrenia for translational machinery. Transl Psychiatry 2018; 8:278. [PMID: 30546022 PMCID: PMC6293320 DOI: 10.1038/s41398-018-0325-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 11/13/2018] [Indexed: 12/02/2022] Open
Abstract
The dopaminergic hypothesis of schizophrenia (SZ) postulates that positive symptoms of SZ, in particular psychosis, are due to disturbed neurotransmission via the dopamine (DA) receptor D2 (DRD2). However, DA is a reactive molecule that yields various oxidative species, and thus has important non-receptor-mediated effects, with empirical evidence of cellular toxicity and neurodegeneration. Here we examine non-receptor-mediated effects of DA on gene co-expression networks and its potential role in SZ pathology. Transcriptomic profiles were measured by RNA-seq in B-cell transformed lymphoblastoid cell lines from 514 SZ cases and 690 controls, both before and after exposure to DA ex vivo (100 μM). Gene co-expression modules were identified using Weighted Gene Co-expression Network Analysis for both baseline and DA-stimulated conditions, with each module characterized for biological function and tested for association with SZ status and SNPs from a genome-wide panel. We identified seven co-expression modules under baseline, of which six were preserved in DA-stimulated data. One module shows significantly increased association with SZ after DA perturbation (baseline: P = 0.023; DA-stimulated: P = 7.8 × 10-5; ΔAIC = -10.5) and is highly enriched for genes related to ribosomal proteins and translation (FDR = 4 × 10-141), mitochondrial oxidative phosphorylation, and neurodegeneration. SNP association testing revealed tentative QTLs underlying module co-expression, notably at FASTKD2 (top P = 2.8 × 10-6), a gene involved in mitochondrial translation. These results substantiate the role of translational machinery in SZ pathogenesis, providing insights into a possible dopaminergic mechanism disrupting mitochondrial function, and demonstrates the utility of disease-relevant functional perturbation in the study of complex genetic etiologies.
Collapse
|
33
|
Breen MS, Wingo AP, Koen N, Donald KA, Nicol M, Zar HJ, Ressler KJ, Buxbaum JD, Stein DJ. Gene expression in cord blood links genetic risk for neurodevelopmental disorders with maternal psychological distress and adverse childhood outcomes. Brain Behav Immun 2018; 73:320-330. [PMID: 29791872 PMCID: PMC6191930 DOI: 10.1016/j.bbi.2018.05.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 02/11/2018] [Accepted: 05/18/2018] [Indexed: 11/29/2022] Open
Abstract
Prenatal exposure to maternal stress and depression has been identified as a risk factor for adverse behavioral and neurodevelopmental outcomes in early childhood. However, the molecular mechanisms through which maternal psychopathology shapes offspring development remain poorly understood. We applied transcriptome-wide screens to 149 umbilical cord blood samples from neonates born to mothers with posttraumatic stress disorder (PTSD; n = 20), depression (n = 31) and PTSD with comorbid depression (n = 13), compared to carefully matched trauma exposed controls (n = 23) and healthy mothers (n = 62). Analyses by maternal diagnoses revealed a clear pattern of gene expression signatures distinguishing neonates born to mothers with a history of psychopathology from those without. Co-expression network analysis identified distinct gene expression perturbations across maternal diagnoses, including two depression-related modules implicated in axon-guidance and mRNA stability, as well as two PTSD-related modules implicated in TNF signaling and cellular response to stress. Notably, these disease-related modules were enriched with brain-expressed genes and genetic risk loci for autism spectrum disorder and schizophrenia, which may imply a causal role for impaired developmental outcomes. These molecular alterations preceded changes in clinical measures at twenty-four months, including reductions in cognitive and socio-emotional outcomes in affected infants. Collectively, these findings indicate that prenatal exposure to maternal psychological distress induces neuronal, immunological and behavioral abnormalities in affected offspring and support the search for early biomarkers of exposures to adverse in utero environments and the classification of children at risk for impaired development.
Collapse
Affiliation(s)
- Michael S Breen
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Genetic and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Aliza P Wingo
- Atlanta Veterans Affairs Medical Center, Atlanta, GA, USA; Department of Psychiatry, School of Medicine, Emory University, Atlanta, GA, USA
| | - Nastassja Koen
- Department of Psychiatry and Mental Health, University of Cape Town, South Africa; South African Medical Research Council (SAMRC) Unit on Risk & Resilience in Mental Disorders, University of Cape Town, Cape Town, South Africa
| | - Kirsten A Donald
- Department of Psychiatry and Mental Health, University of Cape Town, South Africa; South African Medical Research Council (SAMRC) Unit on Risk & Resilience in Mental Disorders, University of Cape Town, Cape Town, South Africa; Department of Paediatrics and Child Health and MRC Unit on Child & Adolescent Health, University of Cape Town, Cape Town, South Africa
| | - Mark Nicol
- Division of Medical Microbiology, Department of Pathology, University of Cape Town and National Health Laboratory Service, South Africa
| | - Heather J Zar
- Department of Paediatrics and Child Health and MRC Unit on Child & Adolescent Health, University of Cape Town, Cape Town, South Africa
| | - Kerry J Ressler
- Department of Psychiatry, School of Medicine, Emory University, Atlanta, GA, USA; McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - Joseph D Buxbaum
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Genetic and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dan J Stein
- Department of Psychiatry and Mental Health, University of Cape Town, South Africa; South African Medical Research Council (SAMRC) Unit on Risk & Resilience in Mental Disorders, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
34
|
Le TT, Savitz J, Suzuki H, Misaki M, Teague TK, White BC, Marino JH, Wiley G, Gaffney PM, Drevets WC, McKinney BA, Bodurka J. Identification and replication of RNA-Seq gene network modules associated with depression severity. Transl Psychiatry 2018; 8:180. [PMID: 30185774 PMCID: PMC6125582 DOI: 10.1038/s41398-018-0234-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 06/21/2018] [Accepted: 07/14/2018] [Indexed: 01/08/2023] Open
Abstract
Genomic variation underlying major depressive disorder (MDD) likely involves the interaction and regulation of multiple genes in a network. Data-driven co-expression network module inference has the potential to account for variation within regulatory networks, reduce the dimensionality of RNA-Seq data, and detect significant gene-expression modules associated with depression severity. We performed an RNA-Seq gene co-expression network analysis of mRNA data obtained from the peripheral blood mononuclear cells of unmedicated MDD (n = 78) and healthy control (n = 79) subjects. Across the combined MDD and HC groups, we assigned genes into modules using hierarchical clustering with a dynamic tree cut method and projected the expression data onto a lower-dimensional module space by computing the single-sample gene set enrichment score of each module. We tested the single-sample scores of each module for association with levels of depression severity measured by the Montgomery-Åsberg Depression Scale (MADRS). Independent of MDD status, we identified 23 gene modules from the co-expression network. Two modules were significantly associated with the MADRS score after multiple comparison adjustment (adjusted p = 0.009, 0.028 at 0.05 FDR threshold), and one of these modules replicated in a previous RNA-Seq study of MDD (p = 0.03). The two MADRS-associated modules contain genes previously implicated in mood disorders and show enrichment of apoptosis and B cell receptor signaling. The genes in these modules show a correlation between network centrality and univariate association with depression, suggesting that intramodular hub genes are more likely to be related to MDD compared to other genes in a module.
Collapse
Affiliation(s)
- Trang T Le
- Department of Mathematics, The University of Tulsa, Tulsa, OK, USA
- Laureate Institute for Brain Research, Tulsa, OK, USA
| | - Jonathan Savitz
- Laureate Institute for Brain Research, Tulsa, OK, USA
- School of Community Medicine, University of Tulsa, Tulsa, OK, USA
| | - Hideo Suzuki
- Laureate Institute for Brain Research, Tulsa, OK, USA
- Department of Educational Psychology, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Masaya Misaki
- Laureate Institute for Brain Research, Tulsa, OK, USA
| | - T Kent Teague
- Departments of Surgery and Psychiatry, University of Oklahoma School of Community Medicine, Tulsa, OK, USA
- Department of Pharmaceutical Sciences, University of Oklahoma College of Pharmacy, Tulsa, OK, USA
- Department of Biochemistry and Microbiology, Oklahoma State University Center for the Health Sciences, Tulsa, OK, USA
| | - Bill C White
- Tandy School of Computer Sciences, The University of Tulsa, Tulsa, OK, USA
| | - Julie H Marino
- Department of Surgery, Integrative Immunology Center, University of Oklahoma School of Community Medicine, Tulsa, OK, USA
| | - Graham Wiley
- Arthritis and Clinical Immunology Research Program, Division of Genomics and Data Sciences, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Patrick M Gaffney
- Arthritis and Clinical Immunology Research Program, Division of Genomics and Data Sciences, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Wayne C Drevets
- Janssen Research & Development, LLC, Johnson & Johnson, Inc, Titusville, NJ, USA
| | - Brett A McKinney
- Department of Mathematics, The University of Tulsa, Tulsa, OK, USA.
- Tandy School of Computer Sciences, The University of Tulsa, Tulsa, OK, USA.
| | - Jerzy Bodurka
- Laureate Institute for Brain Research, Tulsa, OK, USA.
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA.
| |
Collapse
|
35
|
Weighted Gene Co-Expression Network Analysis Reveals Dysregulation of Mitochondrial Oxidative Phosphorylation in Eating Disorders. Genes (Basel) 2018; 9:genes9070325. [PMID: 29958387 PMCID: PMC6070803 DOI: 10.3390/genes9070325] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Revised: 06/16/2018] [Accepted: 06/25/2018] [Indexed: 01/22/2023] Open
Abstract
The underlying mechanisms of eating disorders (EDs) are very complicated and still poorly understood. The pathogenesis of EDs may involve the interplay of multiple genes. To investigate the dysregulated gene pathways in EDs we analyzed gene expression profiling in dorsolateral prefrontal cortex (DLPFC) tissues from 15 EDs cases, including 3 with anorexia nervosa (AN), 7 with bulimia nervosa (BN), 2 AN-BN cases, 3 cases of EDs not otherwise specified, and 102 controls. We further used a weighted gene co-expression network analysis to construct a gene co-expression network and to detect functional modules of highly correlated genes. The functional enrichment analysis of genes in co-expression modules indicated that an altered mitochondrial oxidative phosphorylation process may be involved in the pathogenesis of EDs.
Collapse
|
36
|
Identification of key gene modules for human osteosarcoma by co-expression analysis. World J Surg Oncol 2018; 16:89. [PMID: 29720180 PMCID: PMC5932805 DOI: 10.1186/s12957-018-1381-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Accepted: 04/03/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Osteosarcoma is a type of bone cancer casting huge threat to the human health worldwide. Previously, gene expression analyses were performed to identify biomarkers for cancer; however, systemic co-expression analysis for osteosarcoma is still in need. The aim of this study was to construct a gene co-expression network that predicts clusters of candidate genes associated with the pathogenesis of osteosarcoma. METHODS Here, we extracted the large scale of datasets from the GEO database. With systematical approaches, we identified the co-expression modules by using weighted gene co-expression network analysis (WGCNA) and investigated the functional enrichments of important modules at GO and KEGG terms. RESULTS First, seven co-expression modules, which contain different genes, were conducted for 2228 genes in the 22 human osteosarcoma samples. Then, correlation study showed that the hub genes between pairwise modules displayed great differences. Lastly, functional enrichments of the co-expression modules showed that the module 5 enriched in immune response, antigen processing, and presentation, which is in consistence with GO result. Therefore, we speculated that the module 5 may play a key role in the pathogenesis of osteosarcoma. CONCLUSIONS Here, we speculated that genes of the module 5 were the essential genes that were associated to human osteosarcoma. Together, our findings not only provided outline of co-expression gene modules for human osteosarcoma, but also promoted the understanding of these modules at functional aspects.
Collapse
|
37
|
Szekely E, Schwantes-An THL, Justice CM, Sabourin JA, Jansen PR, Muetzel RL, Sharp W, Tiemeier H, Sung H, White TJ, Wilson AF, Shaw P. Genetic associations with childhood brain growth, defined in two longitudinal cohorts. Genet Epidemiol 2018; 42:405-414. [PMID: 29682794 DOI: 10.1002/gepi.22122] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 03/05/2018] [Accepted: 03/19/2018] [Indexed: 01/29/2023]
Abstract
Genome-wide association studies (GWASs) are unraveling the genetics of adult brain neuroanatomy as measured by cross-sectional anatomic magnetic resonance imaging (aMRI). However, the genetic mechanisms that shape childhood brain development are, as yet, largely unexplored. In this study we identify common genetic variants associated with childhood brain development as defined by longitudinal aMRI. Genome-wide single nucleotide polymorphism (SNP) data were determined in two cohorts: one enriched for attention-deficit/hyperactivity disorder (ADHD) (LONG cohort: 458 participants; 119 with ADHD) and the other from a population-based cohort (Generation R: 257 participants). The growth of the brain's major regions (cerebral cortex, white matter, basal ganglia, and cerebellum) and one region of interest (the right lateral prefrontal cortex) were defined on all individuals from two aMRIs, and a GWAS and a pathway analysis were performed. In addition, association between polygenic risk for ADHD and brain growth was determined for the LONG cohort. For white matter growth, GWAS meta-analysis identified a genome-wide significant intergenic SNP (rs12386571, P = 9.09 × 10-9 ), near AKR1B10. This gene is part of the aldo-keto reductase superfamily and shows neural expression. No enrichment of neural pathways was detected and polygenic risk for ADHD was not associated with the brain growth phenotypes in the LONG cohort that was enriched for the diagnosis of ADHD. The study illustrates the use of a novel brain growth phenotype defined in vivo for further study.
Collapse
Affiliation(s)
- Eszter Szekely
- Section on Neurobehavioral Clinical Research, Social and Behavioral Research Branch, National Human Genome Research Institute, Bethesda, Maryland, United States of America.,Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Canada
| | - Tae-Hwi Linus Schwantes-An
- Genometrics Section, Computational and Statistical Genomics Branch, National Human Genome Research Institute, Baltimore, Maryland, United States of America.,Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Cristina M Justice
- Genometrics Section, Computational and Statistical Genomics Branch, National Human Genome Research Institute, Baltimore, Maryland, United States of America
| | - Jeremy A Sabourin
- Genometrics Section, Computational and Statistical Genomics Branch, National Human Genome Research Institute, Baltimore, Maryland, United States of America
| | - Philip R Jansen
- The Generation R Study Group, Erasmus Medical Center, Rotterdam, The Netherlands.,Department of Child and Adolescent Psychiatry/Psychology, Sophia Children's Hospital-Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ryan L Muetzel
- The Generation R Study Group, Erasmus Medical Center, Rotterdam, The Netherlands.,Department of Child and Adolescent Psychiatry/Psychology, Sophia Children's Hospital-Erasmus Medical Center, Rotterdam, The Netherlands
| | - Wendy Sharp
- Section on Neurobehavioral Clinical Research, Social and Behavioral Research Branch, National Human Genome Research Institute, Bethesda, Maryland, United States of America
| | - Henning Tiemeier
- The Generation R Study Group, Erasmus Medical Center, Rotterdam, The Netherlands.,Department of Child and Adolescent Psychiatry/Psychology, Sophia Children's Hospital-Erasmus Medical Center, Rotterdam, The Netherlands.,Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Heejong Sung
- Genometrics Section, Computational and Statistical Genomics Branch, National Human Genome Research Institute, Baltimore, Maryland, United States of America
| | - Tonya J White
- The Generation R Study Group, Erasmus Medical Center, Rotterdam, The Netherlands.,Department of Child and Adolescent Psychiatry/Psychology, Sophia Children's Hospital-Erasmus Medical Center, Rotterdam, The Netherlands
| | - Alexander F Wilson
- Genometrics Section, Computational and Statistical Genomics Branch, National Human Genome Research Institute, Baltimore, Maryland, United States of America
| | - Philip Shaw
- Section on Neurobehavioral Clinical Research, Social and Behavioral Research Branch, National Human Genome Research Institute, Bethesda, Maryland, United States of America
| |
Collapse
|
38
|
Vawter MP, Philibert R, Rollins B, Ruppel PL, Osborn TW. Exon Array Biomarkers for the Differential Diagnosis of Schizophrenia and Bipolar Disorder. MOLECULAR NEUROPSYCHIATRY 2018; 3:197-213. [PMID: 29888231 DOI: 10.1159/000485800] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 11/16/2017] [Indexed: 12/26/2022]
Abstract
This study developed potential blood-based biomarker tests for diagnosing and differentiating schizophrenia (SZ), bipolar disorder type I (BD), and normal control (NC) subjects using mRNA gene expression signatures. A total of 90 subjects (n = 30 each for the three groups of subjects) provided blood samples at two visits. The Affymetrix exon microarray was used to profile the expression of over 1.4 million probesets. We selected potential biomarker panels using the temporal stability of the probesets and also back-tested them at two different visits for each subject. The 18-gene biomarker panels, using logistic regression modeling, correctly differentiated the three groups of subjects with high accuracy across the two different clinical visits (83-88% accuracy). The results are also consistent with the actual data and the "leave-one-out" analyses, indicating that the models should be predictive when applied to independent data cohorts. Many of the SZ and BD subjects were taking antipsychotic and mood stabilizer medications at the time of blood draw, raising the possibility that these drugs could have affected some of the differential transcription signatures. Using an independent Illumina data set of gene expression data from antipsychotic medication-free SZ subjects, the 18-gene biomarker panels produced a receiver operating characteristic curve accuracy greater than 0.866 in patients that were less than 30 years of age and medication free. We confirmed select transcripts by quantitative PCR and the nCounter® System. The episodic nature of psychiatric disorders might lead to highly variable results depending on when blood is collected in relation to the severity of the disease/symptoms. We have found stable trait gene panel markers for lifelong psychiatric disorders that may have diagnostic utility in younger undiagnosed subjects where there is a critical unmet need. The study requires replication in subjects for ultimate proof of the utility of the differential diagnosis.
Collapse
Affiliation(s)
- Marquis Philip Vawter
- Functional Genomics Laboratory, Department of Psychiatry, University of California, Irvine, California, USA
| | - Robert Philibert
- Department of Psychiatry, University of Iowa, Iowa City, Iowa, USA
| | - Brandi Rollins
- Functional Genomics Laboratory, Department of Psychiatry, University of California, Irvine, California, USA
| | | | | |
Collapse
|
39
|
Malamon JS, Kriete A. Integrated Systems Approach Reveals Sphingolipid Metabolism Pathway Dysregulation in Association with Late-Onset Alzheimer's Disease. BIOLOGY 2018; 7:biology7010016. [PMID: 29425116 PMCID: PMC5872042 DOI: 10.3390/biology7010016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 01/22/2018] [Accepted: 02/06/2018] [Indexed: 01/21/2023]
Abstract
Late-onset Alzheimer’s disease (LOAD) and age are significantly correlated such that one-third of Americans beyond 85 years of age are afflicted. We have designed and implemented a pilot study that combines systems biology approaches with traditional next-generation sequencing (NGS) analysis techniques to identify relevant regulatory pathways, infer functional relationships and confirm the dysregulation of these biological pathways in LOAD. Our study design is a most comprehensive systems approach combining co-expression network modeling derived from RNA-seq data, rigorous quality control (QC) standards, functional ontology, and expression quantitative trait loci (eQTL) derived from whole exome (WES) single nucleotide variant (SNV) genotype data. Our initial results reveal several statistically significant, biologically relevant genes involved in sphingolipid metabolism. To validate these findings, we performed a gene set enrichment analysis (GSEA). The GSEA revealed the sphingolipid metabolism pathway and regulation of autophagy in association with LOAD cases. In the execution of this study, we have successfully tested an integrative approach to identify both novel and known LOAD drivers in order to develop a broader and more detailed picture of the highly complex transcriptional and regulatory landscape of age-related dementia.
Collapse
Affiliation(s)
- John Stephen Malamon
- Bossone Research Center, School of Biomedical Engineering, Science and Health Systems, Drexel University, 3141 Chestnut Street, Philadelphia, PA 19104, USA.
| | - Andres Kriete
- Bossone Research Center, School of Biomedical Engineering, Science and Health Systems, Drexel University, 3141 Chestnut Street, Philadelphia, PA 19104, USA.
| |
Collapse
|
40
|
Izadi F, Soheilifar MH. Exploring Potential Biomarkers Underlying Pathogenesis of Alzheimer's Disease by Differential Co-expression Analysis. Avicenna J Med Biotechnol 2018; 10:233-241. [PMID: 30555656 PMCID: PMC6252023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Alzheimer's Disease (AD) is the most common form of dementia in the elderly. Due to the facts that biological causes of AD are complex in addition to increasing rates of AD worldwide, a deeper understanding of AD etiology is required for AD treatment and diagnosis. METHODS To identify molecular pathological alterations in AD brains, GSE36980 series containing microarray data samples from temporal cortex, frontal cortex and hippocampus were downloaded from Gene Expression Omnibus (GEO) database and valid gene symbols were subjected to building a gene co-expression network by a bioinformatics tool known as differential regulation from differential co-expression (DCGL) software package. Then, a network-driven integrative analysis was performed to find significant genes and underlying biological terms. RESULTS A total of 17088 unique genes were parsed into three independent differential co-expression networks. As a result, a small number of differentially co-regulated genes mostly in frontal and hippocampus lobs were detected as potential biomarkers related to AD brains. Ultimately differentially co-regulated genes were enriched in biological terms including response to lipid and fatty acid and pathways mainly signaling pathway such as G-protein signaling pathway and glutamate receptor groups II and III. By conducting co-expression analysis, our study identified multiple genes that may play an important role in the pathogenesis of AD. CONCLUSION The study aimed to provide a systematic understanding of the potential relationships among these genes and it is hoped that it could aid in AD biomarker discovery.
Collapse
Affiliation(s)
- Fereshteh Izadi
- Department of Genetics, Evolution and Environment, Darwin Building, University College London (UCL), London, UK,Corresponding author: Fereshteh Izadi, PhD, Department of Genetics, Evolution and Environment, Darwin Building, University College London (UCL), Gower Street, London WC1E 6BT, UK, Tel: +44 7846280861, E-mail:
| | | |
Collapse
|
41
|
Chen Y, Liu Y, Du M, Zhang W, Xu L, Gao X, Zhang L, Gao H, Xu L, Li J, Zhao M. Constructing a comprehensive gene co-expression based interactome in Bos taurus. PeerJ 2017; 5:e4107. [PMID: 29226034 PMCID: PMC5719962 DOI: 10.7717/peerj.4107] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 11/08/2017] [Indexed: 01/08/2023] Open
Abstract
Integrating genomic information into cattle breeding is an important approach to exploring genotype-phenotype relationships for complex traits related to diary and meat production. To assist with genomic-based selection, a reference map of interactome is needed to fully understand and identify the functional relevant genes. To this end, we constructed a co-expression analysis of 92 tissues and this represents the systematic exploration of gene-gene relationship in Bos taurus. By using robust WGCNA (Weighted Gene Correlation Network Analysis), we described the gene co-expression network of 5,000 protein-coding genes with majority variations in expression across 92 tissues. Further module identifications found 55 highly organized functional clusters representing diverse cellular activities. To demonstrate the re-use of our interaction for functional genomics analysis, we extracted a sub-network associated with DNA binding genes in Bos taurus. The subnetwork was enriched within regulation of transcription from RNA polymerase II promoter representing central cellular functions. In addition, we identified 28 novel linker genes associated with more than 100 DNA binding genes. Our WGCNA-based co-expression network reconstruction will be a valuable resource for exploring the molecular mechanisms of incompletely characterized proteins and for elucidating larger-scale patterns of functional modulization in the Bos taurus genome.
Collapse
Affiliation(s)
- Yan Chen
- Innovation Team of Cattle Genetics and Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yining Liu
- The School of Public Health, Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou, China
| | - Min Du
- Department of Animal Science, Washington State University, Pullman, WA, United States of America
| | - Wengang Zhang
- Innovation Team of Cattle Genetics and Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Ling Xu
- Innovation Team of Cattle Genetics and Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xue Gao
- Innovation Team of Cattle Genetics and Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Lupei Zhang
- Innovation Team of Cattle Genetics and Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Huijiang Gao
- Innovation Team of Cattle Genetics and Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Lingyang Xu
- Innovation Team of Cattle Genetics and Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Junya Li
- Innovation Team of Cattle Genetics and Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Min Zhao
- School of Engineering, Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, Queensland, Australia
| |
Collapse
|
42
|
Gassó P, Mas S, Rodríguez N, Boloc D, García-Cerro S, Bernardo M, Lafuente A, Parellada E. Microarray gene-expression study in fibroblast and lymphoblastoid cell lines from antipsychotic-naïve first-episode schizophrenia patients. J Psychiatr Res 2017; 95:91-101. [PMID: 28822801 DOI: 10.1016/j.jpsychires.2017.08.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 07/25/2017] [Accepted: 08/04/2017] [Indexed: 12/16/2022]
Abstract
Schizophrenia (SZ) is a chronic psychiatric disorder whose onset of symptoms occurs in late adolescence and early adulthood. The etiology is complex and involves important gene-environment interactions. Microarray gene-expression studies on SZ have identified alterations in several biological processes. The heterogeneity in the results can be attributed to the use of different sample types and other important confounding factors including age, illness chronicity and antipsychotic exposure. The aim of the present microarray study was to analyze, for the first time to our knowledge, differences in gene expression profiles in 18 fibroblast (FCLs) and 14 lymphoblastoid cell lines (LCLs) from antipsychotic-naïve first-episode schizophrenia (FES) patients and healthy controls. We used an analytical approach based on protein-protein interaction network construction and functional annotation analysis to identify the biological processes that are altered in SZ. Significant differences in the expression of 32 genes were found when LCLs were assessed. The network and gene set enrichment approach revealed the involvement of similar biological processes in FCLs and LCLs, including apoptosis and related biological terms such as cell cycle, autophagy, cytoskeleton organization and response to stress and stimulus. Metabolism and other processes, including signal transduction, kinase activity and phosphorylation, were also identified. These results were replicated in two independent cohorts using the same analytical approach. This provides more evidence for altered apoptotic processes in antipsychotic-naïve FES patients and other important biological functions such as cytoskeleton organization and metabolism. The convergent results obtained in both peripheral cell models support their usefulness for transcriptome studies on SZ.
Collapse
Affiliation(s)
- Patricia Gassó
- Dept. of Basic Clinical Practice, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | - Sergi Mas
- Dept. of Basic Clinical Practice, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain
| | | | - Daniel Boloc
- Dept. of Basic Clinical Practice, University of Barcelona, Spain
| | | | - Miquel Bernardo
- Barcelona Clínic Schizophrenia Unit (BCSU), Neuroscience Institute, Hospital Clínic de Barcelona, Spain; Dept. of Medicine, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain
| | - Amalia Lafuente
- Dept. of Basic Clinical Practice, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain
| | - Eduard Parellada
- Dept. of Basic Clinical Practice, University of Barcelona, Spain; Barcelona Clínic Schizophrenia Unit (BCSU), Neuroscience Institute, Hospital Clínic de Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain
| |
Collapse
|
43
|
Development of multivariable models to predict change in Body Mass Index within a clinical trial population of psychotic individuals. Sci Rep 2017; 7:14738. [PMID: 29116126 PMCID: PMC5677086 DOI: 10.1038/s41598-017-15137-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 10/18/2017] [Indexed: 12/20/2022] Open
Abstract
Many antipsychotics promote weight gain, which can lead to non-compliance and relapse of psychosis. By developing models that accurately identify individuals at greater risk of weight gain, clinicians can make informed treatment decisions and target intervention measures. We examined clinical, genetic and expression data for 284 individuals with psychosis derived from a previously published randomised controlled trial (IMPACT). These data were used to develop regression and classification models predicting change in Body Mass Index (BMI) over one year. Clinical predictors included demographics, anthropometrics, cardiac and blood measures, diet and exercise, physical and mental health, medication and BMI outcome measures. We included genetic polygenic risk scores (PRS) for schizophrenia, bipolar disorder, BMI, waist-hip-ratio, insulin resistance and height, as well as gene co-expression modules generated by Weighted Gene Co-expression Network Analysis (WGCNA). The best performing predictive models for BMI and BMI gain after one year used clinical data only, which suggests expression and genetic data do not improve prediction in this cohort.
Collapse
|
44
|
Chen J, Schwarz E. The role of blood-based biomarkers in advancing personalized therapy of schizophrenia. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2017. [DOI: 10.1080/23808993.2017.1400906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Junfang Chen
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Emanuel Schwarz
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
45
|
Bo L, Wei B, Wang Z, Kong D, Gao Z, Miao Z. Screening of Critical Genes and MicroRNAs in Blood Samples of Patients with Ruptured Intracranial Aneurysms by Bioinformatic Analysis of Gene Expression Data. Med Sci Monit 2017; 23:4518-4525. [PMID: 28930970 PMCID: PMC5618721 DOI: 10.12659/msm.902953] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Background This study aimed to identify more potential genes and miRNAs associated with the pathogenesis of intracranial aneurysms (IAs). Material/Methods The dataset of GSE36791 (accession number) was downloaded from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) were screened for in the blood samples from patients with ruptured IAs and controls, followed by functional and pathway enrichment analyses. In addition, gene co-expression network was constructed and significant modules were extracted from the network by WGCNA R package. Screening for miRNAs that could regulate DEGs in the modules was performed and an analysis of regulatory relationships was conducted. Results A total of 304 DEGs (167 up-regulated and 137 down-regulated genes) were screened for in blood samples from patients with ruptured IAs compared with those from controls. Functional enrichment analysis showed that the up-regulated genes were mainly associated with immune response and the down-regulated DEGs were mainly concerned with the structure of ribosome and translation. Besides, six functional modules were significantly identified, including four modules enriched by up-regulated genes and two modules enriched by down-regulated genes. Thereinto, the blue, yellow, and turquoise modules of up-regulated genes were all linked with immune response. Additionally, 16 miRNAs were predicted to regulate DEGs in the three modules associated with immune response, such as hsa-miR-1304, hsa-miR-33b, hsa-miR-125b, and hsa-miR-125a-5p. Conclusions Several genes and miRNAs (such as miR-1304, miR-33b, IRS2 and KCNJ2) may take part in the pathogenesis of IAs.
Collapse
Affiliation(s)
- Lijuan Bo
- Department of Infections, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Bo Wei
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Zhanfeng Wang
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Daliang Kong
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Zheng Gao
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Zhuang Miao
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China (mainland)
| |
Collapse
|
46
|
Keck M, Fournier A, Gualtieri F, Walker A, von Rüden EL, Russmann V, Deeg CA, Hauck SM, Krause R, Potschka H. A systems level analysis of epileptogenesis-associated proteome alterations. Neurobiol Dis 2017; 105:164-178. [PMID: 28576708 DOI: 10.1016/j.nbd.2017.05.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 05/22/2017] [Accepted: 05/29/2017] [Indexed: 12/18/2022] Open
Abstract
Despite intense research efforts, the knowledge about the mechanisms of epileptogenesis and epilepsy is still considered incomplete and limited. However, an in-depth understanding of molecular pathophysiological processes is crucial for the rational selection of innovative biomarkers and target candidates. Here, we subjected proteomic data from different phases of a chronic rat epileptogenesis model to a comprehensive systems level analysis. Weighted Gene Co-expression Network analysis identified several modules of interconnected protein groups reflecting distinct molecular aspects of epileptogenesis in the hippocampus and the parahippocampal cortex. Characterization of these modules did not only further validate the data but also revealed regulation of molecular processes not described previously in the context of epilepsy development. The data sets also provide valuable information about temporal patterns, which should be taken into account for development of preventive strategies in particular when it comes to multi-targeting network pharmacology approaches. In addition, principal component analysis suggests candidate biomarkers, which might inform the design of novel molecular imaging approaches aiming to predict epileptogenesis during different phases or confirm epilepsy manifestation. Further studies are necessary to distinguish between molecular alterations, which correlate with epileptogenesis versus those reflecting a mere consequence of the status epilepticus.
Collapse
Affiliation(s)
- Michael Keck
- Institute of Pharmacology, Toxicology and Pharmacy, Ludwig-Maximilians-University (LMU), 80539 Munich, Germany
| | - Anna Fournier
- Bioinformatics Core, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4367 Belvaux, Luxembourg
| | - Fabio Gualtieri
- Institute of Pharmacology, Toxicology and Pharmacy, Ludwig-Maximilians-University (LMU), 80539 Munich, Germany
| | - Andreas Walker
- Institute of Pharmacology, Toxicology and Pharmacy, Ludwig-Maximilians-University (LMU), 80539 Munich, Germany
| | - Eva-Lotta von Rüden
- Institute of Pharmacology, Toxicology and Pharmacy, Ludwig-Maximilians-University (LMU), 80539 Munich, Germany
| | - Vera Russmann
- Institute of Pharmacology, Toxicology and Pharmacy, Ludwig-Maximilians-University (LMU), 80539 Munich, Germany
| | - Cornelia A Deeg
- Institute of Animal Physiology, Department of Veterinary Sciences, Ludwig-Maximilians-University (LMU), 80539 Munich, Germany; Experimental Ophthalmology, Philipps University of Marburg, 35037 Marburg, Germany
| | - Stefanie M Hauck
- Research Unit Protein Science, Helmholtz Center Munich, 85764 Neuherberg, Germany
| | - Roland Krause
- Bioinformatics Core, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4367 Belvaux, Luxembourg.
| | - Heidrun Potschka
- Institute of Pharmacology, Toxicology and Pharmacy, Ludwig-Maximilians-University (LMU), 80539 Munich, Germany.
| |
Collapse
|
47
|
Identification of Key Modules and Hub Genes of Keloids with Weighted Gene Coexpression Network Analysis. Plast Reconstr Surg 2017; 139:376-390. [PMID: 28121871 DOI: 10.1097/prs.0000000000003014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND Keloid scarring impairs patients' quality of life, and although many therapeutic strategies have been developed, most remain unsatisfactory because of limited understanding of the mechanisms underlying keloid development. METHODS A microarray gene expression data set from keloid tissue was acquired from the Gene Expression Omnibus. Differentially expressed genes in fibroblasts and keratinocytes underwent functional annotation and pathway analysis. Weighted gene coexpression network analysis was applied to identify the gene targets of keloid scars within differentially expressed genes. Modules and hub genes for keloids were identified. Enrichment analysis was undertaken to verify the modules' and hub genes' relationship with keloids. RESULTS Enrichment analysis and pathway analysis showed gene ontology terms and pathways related to keloids. Each cell type generated three modules in weighted gene coexpression network analysis, with one module most related to keloids. Enrichment analysis showed that the modules concerned are enriched with terms related to keloids. Three hub genes were selected for fibroblasts and keratinocytes, and their relationship to keloids was verified. Immunohistochemical staining verified expression change of some hub genes. CONCLUSIONS This is the first study to describe the gene networks underlying keloids. Modules and hub genes generated in the present study are highly related to keloids and may identify novel therapeutic targets for treatment of keloids. CLINICAL QUESTION/LEVEL OF EVIDENCE Therapeutic, V.
Collapse
|
48
|
A Four-Biomarker Blood Signature Discriminates Systemic Inflammation Due to Viral Infection Versus Other Etiologies. Sci Rep 2017; 7:2914. [PMID: 28588308 PMCID: PMC5460227 DOI: 10.1038/s41598-017-02325-8] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 04/10/2017] [Indexed: 02/07/2023] Open
Abstract
The innate immune system of humans and other mammals responds to pathogen-associated molecular patterns (PAMPs) that are conserved across broad classes of infectious agents such as bacteria and viruses. We hypothesized that a blood-based transcriptional signature could be discovered indicating a host systemic response to viral infection. Previous work identified host transcriptional signatures to individual viruses including influenza, respiratory syncytial virus and dengue, but the generality of these signatures across all viral infection types has not been established. Based on 44 publicly available datasets and two clinical studies of our own design, we discovered and validated a four-gene expression signature in whole blood, indicative of a general host systemic response to many types of viral infection. The signature’s genes are: Interferon Stimulated Gene 15 (ISG15), Interleukin 16 (IL16), 2′,5′-Oligoadenylate Synthetase Like (OASL), and Adhesion G Protein Coupled Receptor E5 (ADGRE5). In each of 13 validation datasets encompassing human, macaque, chimpanzee, pig, mouse, rat and all seven Baltimore virus classification groups, the signature provides statistically significant (p < 0.05) discrimination between viral and non-viral conditions. The signature may have clinical utility for differentiating host systemic inflammation (SI) due to viral versus bacterial or non-infectious causes.
Collapse
|
49
|
Zhang Y, Wang J, Ji LJ, Li L, Wei M, Zhen S, Wen CC. Identification of Key Gene Modules of Neuropathic Pain by Co-Expression Analysis. J Cell Biochem 2017; 118:4436-4443. [PMID: 28460420 DOI: 10.1002/jcb.26098] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 04/27/2017] [Indexed: 12/26/2022]
Abstract
Neuropathic pain (NP) is a substantial clinical problem causing great injury to people word-widely. Although gene expression analyses had been performed previously, the mechanisms underlying the etiology and development of NP are still poorly understood. To understand the function genes involved in the etiology and development of NP, we built the co-expression modules and performed function enrichment analysis for neuropathic pain. In the present study, from a public microarray data set (GSE69901) from NCBI, gene co-expression modules were contributed with the help of WGCNA for 12 neuropathic pain samples and 13 control samples, respectively. And functional enrichment analyses were followed by DAVID database. Firstly, we established 21 co-expression modules and 19 co-expression modules out of 5,000 high-express genes in NP and control samples, respectively. Then, it showed great difference in interaction relationships of total genes and hub-genes between pairwise modules, which indicated the high confidence of gene co-expression modules. Finally, functional enrichment analysis of the top five co-expression modules in NP exhibited great differences and significant enrichment in transcription regulation of RNA polymerase II promoter and ubiquitin mediated proteolysis pathway. RNA polymerase II promoter and ubiquitin-mediated proteolysis pathway played important role in etiology and development of NP. Anyhow, our findings provided the framework of gene co-expression modules of NP and furthered the understanding of these modules from functional aspect. J. Cell. Biochem. 118: 4436-4443, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Anesthesiology, Huai'an First People's Hospital, Nanjing Medical University, 6 Beijing Road West, Huai'an, Jiangsu, 223300, China
| | - Jinlin Wang
- Department of Anesthesiology, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Li-Juan Ji
- Department of Sport Medicine and Pain Clinic, Center of Sports Rehabilitation, School of Sport Science, Shanghai University of Sport, Shanghai, 200438, China
| | - Lin Li
- Department of Anesthesiology, Huai'an First People's Hospital, Nanjing Medical University, 6 Beijing Road West, Huai'an, Jiangsu, 223300, China
| | - Meng Wei
- Department of Anesthesiology, Huai'an First People's Hospital, Nanjing Medical University, 6 Beijing Road West, Huai'an, Jiangsu, 223300, China
| | - Su Zhen
- Department of Anesthesiology, Huai'an First People's Hospital, Nanjing Medical University, 6 Beijing Road West, Huai'an, Jiangsu, 223300, China
| | - Cheng-Cai Wen
- Department of Rehabilitation, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, Huai'an, China
| |
Collapse
|
50
|
Liu X, Hu AX, Zhao JL, Chen FL. Identification of Key Gene Modules in Human Osteosarcoma by Co-Expression Analysis Weighted Gene Co-Expression Network Analysis (WGCNA). J Cell Biochem 2017; 118:3953-3959. [PMID: 28398605 DOI: 10.1002/jcb.26050] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 04/10/2017] [Indexed: 12/21/2022]
Abstract
Osteosarcoma is the eighth-most common form of childhood cancer, comprising about 20% of all primary bone cancers. To date, systemic co-expression analysis for this cancer is still insufficient to explain the pathogenesis of poorly understood OC. The objective of this study was to construct a gene co-expression network to predict clusters of candidate genes involved in the pathogenesis of osteosarcoma. First, we contributed co-expression modules via weighted gene co-expression network analysis (WGCNA) and investigated the functional enrichment analysis of co-expression genes in terms of GO and KEGG. In result, seven co-expression modules were identified, containing 2,228 differentially expressed genes identified from the 22 human osteosarcoma samples. Subsequently, correlation study showed that the hub-genes between pair-wise modules displayed significant differences. Lastly, functional enrichment analysis of the co-expression modules showed that the module 5 enriched in progresses of immune response, antigen processing, and presentation. In conclusion, we identified essential genes in module 5 which were associated to human osteosarcoma. The key genes in our findings might provide the framework of co-expression gene modules of human osteosarcoma. Further, the functional analysis of these associated genes provides references to understand the mechanism of Osteosarcoma. J. Cell. Biochem. 118: 3953-3959, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Xiangsheng Liu
- The Department of Orthopaedics, The Fifth People's Hospital of Fudan University, Heqing Road No.801, Minghangqu, Shanghai, 200240, People's Republic of China
| | - Ai-Xin Hu
- The Department of Orthopedic Surgery, People's Hospital of Three Gorges University, YiChang, Hubei Province, People's Republic of China
| | - Jia-Li Zhao
- Department of Orthopaedics, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, Huai'an, Jiangsu, 223002, People's Republic of China
| | - Feng-Li Chen
- Central Laboratory, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, 223300, People's Republic of China
| |
Collapse
|