| Peer-Reviewed

The NRF1/miR-4514/SOCS3 Pathway Is Associated with Schizophrenia Pathogenesis

Received: 27 September 2021    Accepted: 15 October 2021    Published: 28 October 2021
Views:       Downloads:
Abstract

Background: Schizophrenia (SZ) is a common and severe mental disease. However, its etiology and pathogenesis have not been fully established. In this study, bioinformatics was used to identify SZ-related genes and reveal the potential mechanisms of them. Methods: Gene expression profiles were obtained from the GSE46509 dataset. Differentially expressed genes (DEGs) were analyzed by Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment databases. A protein-protein interaction (PPI) network was established. TargetScan and miRGen, which are based on bioinformatics algorithms, were used to predict potential candidate target miRNAs and transcription factors. Results: Compared to healthy people controls, a total of 1422 DEGs were identified in SZ patient samples. Functional enrichment analysis revealed that these DEGs were significantly enriched in RNA processing, mRNA binding, and cell adhesion molecules. In addition, in the PPI network, SOCS3, FBXO9, ASB17, FBXO10, and ASB4 were identified as hub genes. In the predicted TF-miRNA-mRNA targeting regulatory network, hsa-miR-4514 was up-regulated by the highly expressed transcription factor (TF) NRF1, which down-regulated multiple hubs genes such as SOCS3, FBXO9, and FBXO10. Conclusions: Several potential biomarkers involved in SZ development were identified by bioinformatics analyses. Furthermore, our findings revealed the underpinning mechanisms of these potential biomarkers in the pathogenesis of SZ. And these results suggest a potential application value in clinical practice.

Published in Clinical Neurology and Neuroscience (Volume 5, Issue 4)
DOI 10.11648/j.cnn.20210504.13
Page(s) 82-97
Creative Commons

This is an Open Access article, distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution and reproduction in any medium or format, provided the original work is properly cited.

Copyright

Copyright © The Author(s), 2021. Published by Science Publishing Group

Keywords

Schizophrenia, Bioinformatics, Regulatory Network, MicroRNAs, Transcription Factors

References
[1] Charlson, F. J., et al., Global Epidemiology and Burden of Schizophrenia: Findings From the Global Burden of Disease Study 2016. Schizophrenia Bulletin, 2018. 44 (6).
[2] Wolfgang Fleischhacker, W., et al., Schizophrenia—Time to Commit to Policy Change. Schizophrenia Bulletin, 2014. 40 (3): p. 165-194.
[3] Maziade, M., At Risk for Serious Mental Illness - Screening Children of Patients with Mood Disorders or Schizophre nia. N Engl J Med, 2017. 376 (10): p. 910-912.
[4] Dietz, A. G., S. A. Goldman, and M. Nedergaard, Glial cells in schizophrenia: a unified hypothesis. Lancet Psychiatry, 2020. 7 (3): p. 272-281.
[5] Bartel, D. P., Metazoan MicroRNAs. Cell, 2018. 173 (1): p. 20.
[6] Berg, M. M. J. V. D., et al., Circulating microRNAs as potential biomarkers for psychiatric and neurodegenerative disorders. Progress in Neurobiology, 2019. 185: p. 101732.
[7] Smigielski, L., et al., Epigenetic mechanisms in schizophrenia and other psychotic disorders: a systematic review of empirical human findings. Molecular Psychiatry, 2020 (11): p. 1-31.
[8] He, K., et al., Identification of serum microRNAs as diagnostic biomarkers for schizophrenia. Hereditas, 2019. 156.
[9] Lai, C. Y., et al., MicroRNA Expression Aberration as Potential Peripheral Blood Biomarkers for Schizophrenia. Plos One, 2011. 6.
[10] Ozsolak, F., et al., Chromatin structure analyses identify miRNA promoters. Genes & Development, 2008. 22 (22): p. 3172-3183.
[11] Wang, W., et al., The p53/miR-193a/EGFR feedback loop function as a driving force for non-small cell lung carcinoma tumorigenesis. Therapeutic Advances in Medical Oncology, 2019. 11.
[12] Xu, Y., et al., Exploring Transcription Factors-microRNAs Co-regulation Networks in Schizophrenia. Schizophr Bull, 2016. 42 (4): p. 1037-45.
[13] Chen, C., et al., TBtools: An Integrative Toolkit Developed for Interactive Analyses of Big Biological Data. Molecular Plant, 2020. 13 (8).
[14] Huang, D. W., et al., DAVID Bioinformatics Resources: expanded annotation database and novel algorithms to better extract biology from large gene lists. Nucleic Acids Research, 2007. 35 (suppl_2): p. W169-W175.
[15] Damian, S., et al., STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic acids research, 2018 (D1): p. D607.
[16] Ba Der, G. D. and C. Hogue, An Automated Method for Finding Molecular Complexes in Large Protein Interaction Networks. BMC Bioinformatics, 2003. 4 (1, article 2): p. 2.
[17] Gabriela, B., et al., ClueGO: a Cytoscape plug-in to decipher functionally grouped gene ontology and pathway annotation networks. Bioinformatics, 2009. 25 (8): p. 1091-1093.
[18] Bindea, G., J. Galon, and B. Mlecnik, CluePedia Cytoscape plugin: pathway insights using integrated experimental and in silico data. Bioinformatics, 2013. 29 (5): p. 661-663.
[19] Chin, C. H., et al., cytoHubba: identifying hub objects and sub-networks from complex interactome. BMC Systems Biology, 2014. 8 (S4): p. S11.
[20] Vikram, A., et al., Predicting effective microRNA target sites in mammalian mRNAs. eLife, 2015. 4: p. e05005.
[21] Georgakilas, G., et al., DIANA-miRGen v3.0: accurate characterization of microRNA promoters and their regulators. Nucleic Acids Res, 2016. 44 (D1): p. D190-5.
[22] Andlauer, T., et al., Genetic factors influencing a neurobiological substrate for psychiatric disorders. 2019.
[23] Richetto, J. and U. Meyer, Epigenetic Modifications in Schizophrenia and Related Disorders: Molecular Scars of Environmental Exposures and Source of Phenotypic Variability. Biological Psychiatry, 2020. 89 (3).
[24] Uher, R. and A. Zwicker, Etiology in psychiatry: embracing the reality of polyene-environmental causation of mental illness. World Psychiatry, 2017. 16 (2): p. 121-129.
[25] Mathoux, J., D. C. Henshall, and G. P. Brennan, Regulatory Mechanisms of the RNA Modification m6A and Significance in Brain Function in Health and Disease. Frontiers in cellular neuroscience, 2021. 15: p. 671932.
[26] Chapman, R., et al., Convergent Evidence That ZNF804A Is a Regulator of Pre-messenger RNA Processing and Gene Expression. Schizophrenia Bulletin, 2018. 45 (6255).
[27] Kanakry, C. G., et al., Neuregulin-1 regulates cell adhesion via an ErbB2/phosphoinositide-3 kinase/Akt-dependent pathway: po tential implications for schizophrenia and cancer. PLoS One, 2007. 2 (12): p. e1369.
[28] Nogami, M., et al., DGCR8-dependent efficient pri-miRNA processing of human pri-miR-9-2. Journal of Biological Chemistry.
[29] Moretti, P. N., et al., Accessing Gene Expression in Treatment-Resistant Schizophrenia. Molecular Neurobiology, 2018.
[30] Papadima, E. M., et al., Evidence towards RNA Binding Motif (RNP1, RRM) Protein 3 (RBM3) as a Potential Biomarker of Lithium Response in Bipolar Disorder Patients. Journal of Molecular Neuroence, 2017.
[31] Lelieveld, S. H., et al., Meta-analysis of 2,104 trios provides support for 10 new genes for intellectual disability. Nature Neuroscience, 2016.
[32] Bronicki, L. M. and B. J. Jasmin, Emerging complexity of the HuD/ELAVl4 gene; implications for neuronal development, function, and dysfunction. Rna-a Publication of the Rna Society, 2013. 19 (8): p. 1019-1037.
[33] Budzinska, M. I., et al., PTPN23 binds the dynein adaptor BICD1 and is required for endocytic sorting of neurotrophin receptors. Journal of Cell Science. 133 (6).
[34] Hauberg, M. E., et al., Analyzing the Role of MicroRNAs in Schizophrenia in the Context of Common Genetic Risk Variants. JAMA Psychiatry, 2016. 73 (4): p. 369-377.
[35] Kubo, M., T. Hanada, and A. Yoshimura, Suppressors of cytokine signaling and immunity. Nat Immunol, 2003. 4 (12): p. 1169-76.
[36] Ni, C., et al., LncRNA-AC006129.1 reactivates a SOCS3-mediated anti-inflammatory response through DNA methylation-mediated CIC downregulation in schizophrenia. Molecular Psychiatry, 2020.
[37] John, P. S., et al., Spatiotemporal separation of PER and CRY posttranslational regulation in the mammalian circadian clock. Proc Natl Acad Sci U S A, 2014. 111 (5): p. 2040-2045.
[38] Agapito, M. A., et al., Fetal alcohol exposure disrupts metabolic signaling in hypothalamic proopiomelanocortin neurons via a circadian mechanism in male mice. Endocrinology, 2014 (7): p. 2578-2588.
[39] Quartier, A., et al., Genes and Pathways Regulated by Androgens in Human Neural Cells, Potential Candidates for the Male Excess in Autism Spectrum Disorder. Biological Psychiatry, 2018: p. S0006322318300088.
[40] Hülskamp, G., et al., Deficiency of UBR1, a ubiquitin ligase of the N-end rule pathway, causes pancreatic dysfunction, malformations and mental retardation (Johanson-Blizzard syndrome). Nature Genetics.
[41] Florentinus-Mefailoski, A., et al., The plasma peptides of Alzheimer's disease. Clin Proteomics, 2021. 18 (1): p. 17.
[42] Li, R., et al., A Potential Autophagy-Related Competing Endogenous RNA Network and Corresponding Diagnostic Efficacy in Schizophrenia. Frontiers in Psychiatry, 2021. 12.
[43] Zhang, H. M., et al., MKL1/miR-5100/CAAP1 loop regulates autophagy and apoptosis in gastric cancer cells. Neoplasia (New York, N.Y.), 2020. 22 (5): p. 220-230.
[44] Kmioek, T., et al., The Interplay between Transcriptional Factors and MicroRNAs as an Important Factor for Th17/Treg Balance in RA Patients. International Journal of Molecular Sciences, 2020. 21 (19): p. 7169.
[45] Aja, A., et al., Further insights into the molecular complexity of the human sinus node – The role of 'novel' transcription factors and microRNAs. Progress in Biophysics and Molecular Biology, 2021.
[46] Bang-Shun, et al., Association of the DISC1 and NRG1 genetic polymorphisms with schizophrenia in a Chinese population. Gene, 2016. 590 (2): p. 293-7.
[47] Buxbaum, J. D., G. Corfas, and K. Roy, Neuregulin 1-erbB signaling and the molecular/cellular basis of schizophrenia. Nature Neuroscience, 2004. 7 (6): p. 575-580.
[48] Falls, D. L., Neuregulins: functions, forms, and signaling strategies. Experimental Cell Research, 2003. 284 (1): p. 14-30.
[49] Bertram, I., et al., Immunohistochemical evidence for impaired neuregulin-1 signaling in the prefrontal cortex in schizophrenia and in unipolar depression. Annals of the New York Academy of Sciences, 2010. 1096 (1): p. 147-156.
[50] A, V. Z. C., et al., Elevated neuregulin-1 and ErbB4 protein in the prefrontal cortex of schizophrenic patients. Schizophrenia Research, 2008. 100 (1–3): p. 270-280.
Cite This Article
  • APA Style

    Yilin Liu, Shujun Li, Xiao Ma, Qing Long, Lei Yu, et al. (2021). The NRF1/miR-4514/SOCS3 Pathway Is Associated with Schizophrenia Pathogenesis. Clinical Neurology and Neuroscience, 5(4), 82-97. https://doi.org/10.11648/j.cnn.20210504.13

    Copy | Download

    ACS Style

    Yilin Liu; Shujun Li; Xiao Ma; Qing Long; Lei Yu, et al. The NRF1/miR-4514/SOCS3 Pathway Is Associated with Schizophrenia Pathogenesis. Clin. Neurol. Neurosci. 2021, 5(4), 82-97. doi: 10.11648/j.cnn.20210504.13

    Copy | Download

    AMA Style

    Yilin Liu, Shujun Li, Xiao Ma, Qing Long, Lei Yu, et al. The NRF1/miR-4514/SOCS3 Pathway Is Associated with Schizophrenia Pathogenesis. Clin Neurol Neurosci. 2021;5(4):82-97. doi: 10.11648/j.cnn.20210504.13

    Copy | Download

  • @article{10.11648/j.cnn.20210504.13,
      author = {Yilin Liu and Shujun Li and Xiao Ma and Qing Long and Lei Yu and Yatang Chen and Wenzhi Wu and Zhichao Guo and Zhaowei Teng and Yong Zeng},
      title = {The NRF1/miR-4514/SOCS3 Pathway Is Associated with Schizophrenia Pathogenesis},
      journal = {Clinical Neurology and Neuroscience},
      volume = {5},
      number = {4},
      pages = {82-97},
      doi = {10.11648/j.cnn.20210504.13},
      url = {https://doi.org/10.11648/j.cnn.20210504.13},
      eprint = {https://article.sciencepublishinggroup.com/pdf/10.11648.j.cnn.20210504.13},
      abstract = {Background: Schizophrenia (SZ) is a common and severe mental disease. However, its etiology and pathogenesis have not been fully established. In this study, bioinformatics was used to identify SZ-related genes and reveal the potential mechanisms of them. Methods: Gene expression profiles were obtained from the GSE46509 dataset. Differentially expressed genes (DEGs) were analyzed by Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment databases. A protein-protein interaction (PPI) network was established. TargetScan and miRGen, which are based on bioinformatics algorithms, were used to predict potential candidate target miRNAs and transcription factors. Results: Compared to healthy people controls, a total of 1422 DEGs were identified in SZ patient samples. Functional enrichment analysis revealed that these DEGs were significantly enriched in RNA processing, mRNA binding, and cell adhesion molecules. In addition, in the PPI network, SOCS3, FBXO9, ASB17, FBXO10, and ASB4 were identified as hub genes. In the predicted TF-miRNA-mRNA targeting regulatory network, hsa-miR-4514 was up-regulated by the highly expressed transcription factor (TF) NRF1, which down-regulated multiple hubs genes such as SOCS3, FBXO9, and FBXO10. Conclusions: Several potential biomarkers involved in SZ development were identified by bioinformatics analyses. Furthermore, our findings revealed the underpinning mechanisms of these potential biomarkers in the pathogenesis of SZ. And these results suggest a potential application value in clinical practice.},
     year = {2021}
    }
    

    Copy | Download

  • TY  - JOUR
    T1  - The NRF1/miR-4514/SOCS3 Pathway Is Associated with Schizophrenia Pathogenesis
    AU  - Yilin Liu
    AU  - Shujun Li
    AU  - Xiao Ma
    AU  - Qing Long
    AU  - Lei Yu
    AU  - Yatang Chen
    AU  - Wenzhi Wu
    AU  - Zhichao Guo
    AU  - Zhaowei Teng
    AU  - Yong Zeng
    Y1  - 2021/10/28
    PY  - 2021
    N1  - https://doi.org/10.11648/j.cnn.20210504.13
    DO  - 10.11648/j.cnn.20210504.13
    T2  - Clinical Neurology and Neuroscience
    JF  - Clinical Neurology and Neuroscience
    JO  - Clinical Neurology and Neuroscience
    SP  - 82
    EP  - 97
    PB  - Science Publishing Group
    SN  - 2578-8930
    UR  - https://doi.org/10.11648/j.cnn.20210504.13
    AB  - Background: Schizophrenia (SZ) is a common and severe mental disease. However, its etiology and pathogenesis have not been fully established. In this study, bioinformatics was used to identify SZ-related genes and reveal the potential mechanisms of them. Methods: Gene expression profiles were obtained from the GSE46509 dataset. Differentially expressed genes (DEGs) were analyzed by Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment databases. A protein-protein interaction (PPI) network was established. TargetScan and miRGen, which are based on bioinformatics algorithms, were used to predict potential candidate target miRNAs and transcription factors. Results: Compared to healthy people controls, a total of 1422 DEGs were identified in SZ patient samples. Functional enrichment analysis revealed that these DEGs were significantly enriched in RNA processing, mRNA binding, and cell adhesion molecules. In addition, in the PPI network, SOCS3, FBXO9, ASB17, FBXO10, and ASB4 were identified as hub genes. In the predicted TF-miRNA-mRNA targeting regulatory network, hsa-miR-4514 was up-regulated by the highly expressed transcription factor (TF) NRF1, which down-regulated multiple hubs genes such as SOCS3, FBXO9, and FBXO10. Conclusions: Several potential biomarkers involved in SZ development were identified by bioinformatics analyses. Furthermore, our findings revealed the underpinning mechanisms of these potential biomarkers in the pathogenesis of SZ. And these results suggest a potential application value in clinical practice.
    VL  - 5
    IS  - 4
    ER  - 

    Copy | Download

Author Information
  • The Sixth Affiliated Hospital of Kunming Medical University, Yuxi, China

  • The Sixth Affiliated Hospital of Kunming Medical University, Yuxi, China

  • The Sixth Affiliated Hospital of Kunming Medical University, Yuxi, China

  • The Sixth Affiliated Hospital of Kunming Medical University, Yuxi, China

  • The Sixth Affiliated Hospital of Kunming Medical University, Yuxi, China

  • The Sixth Affiliated Hospital of Kunming Medical University, Yuxi, China

  • The Sixth Affiliated Hospital of Kunming Medical University, Yuxi, China

  • The Sixth Affiliated Hospital of Kunming Medical University, Yuxi, China

  • The Sixth Affiliated Hospital of Kunming Medical University, Yuxi, China

  • The Sixth Affiliated Hospital of Kunming Medical University, Yuxi, China

  • Sections