RNA Biomarkers in Bipolar Disorder and Response to Mood Stabilizers
Abstract
:1. Introduction
2. RNA Biomarkers and Bipolar Disorder
2.1. Peripheral Levels of RNA Markers in Biofluids or Peripheral Cells from Patients with BD
Ref. | Sample | RNA Source | RNA Type | Method | Targets | Main Findings |
---|---|---|---|---|---|---|
[28] | 15 patients with BD and 17 with MDD during a depression episode and at remission | Whole blood | miRNA | qPCR | miR-499, miR-798 and miR-1908 | Lower levels of all miRNAs in the depressive state compared with remission exclusively in patients with BD |
[27] | 58 patients with BD I (19 manic and 39 euthymic) and 51 HCs | Whole blood | miRNA | qPCR | miR-26b-5p, miR-9-5p, miR-29a-3p, miR-106a-5p, miR-106b-5p, miR-107, miR-125a-3p and miR-125b-5p | Levels of miR-29a-3p, miR-106b-5p, miR-107 and miR-125a-3p were significantly higher in patients with BD compared with HCs. Levels of miR-106a-5p and miR-107 were significantly higher in manic patients compared to euthymic patients |
[25] | 69 patients with BD (15 depressed, 27 manic and 27 euthymic) and 41 HCs | Plasma exosomes | miRNA | qPCR | Genome wide | Levels of miR-484, miR-652-3p and miR-142-3p were lower, while levels of miR-185-5p were higher in patients with BD compared with HCs. No alterations among different states of BD |
[20] | Discovery cohort: 7 patients with BD, 7 with MDD and 6 HCs; validation cohort: 27 patients with BD and 32 with MDD | Plasma | miRNA | Microarray, qPCR | Genome wide | Higher levels of miR-19b-3p in patients with BD compared with patients with MDD |
[33] | 54 patients with BD I, 45 with BD II and 42 HCs | PBMC | mRNA | qPCR | COMT, DNMT, GAD67, MECP2, PDYN and SERT | Lower PDYN expression in patients with BD II but not BD I compared with HCs. Higher levels of genes involved in methylation, such as DNMT3b and MECP2, in patients with BD II compared with HCs |
[34] | 169 patients with BD and 211 HCs | Whole blood | mRNA | qPCR | NOTCH4 | Higher NOTCH4 levels in patients with BD compared with HC |
[18] | 50 patients with BD I and 50 HCs | Whole blood | mRNA, lncRNA | qPCR | SNHG6, MALAT1, Linc00511, Linc00346, VDR and CYP27B1 | Levels of VDR, SNHG6, CYP27B1, MALAT1 and Linc00346 were higher in patients compared with HCs |
[24] | 20 patients with BD I and 21 HCs | Plasma EV | miRNA | Microarray | Genome wide | 33 nominally significant microRNAs altered in patients with BD |
[17] | Discovery cohort: 4 patients with BD and 4 HCs; validation cohort: 16 patients with BD and 16 controls | Whole blood | mRNA, circRNA | NGS and qPCR | Genome wide | 50 circRNAs and 244 mRNAs showed nominally significant higher levels, while 44 circRNAs and 294 mRNAs lower levels in patients with BD compared with HCs |
[16] | Discovery cohort: 18 patients with BD, 44 with MDD, and 31 HCs; validation cohort: 26 patients with BD, 84 with MDD and 74 HCs | Plasma | mRNA, miRNA | NGS and qPCR | Genome wide | Higher levels of hsa-let-7e-5p and hsa-miR-125a-5p in patients with either BD or MDD compared with HCs |
[19] | Discovery cohort: 4 patients with BD and 4 HCs; validation cohort: 130 patients with BD and 116 HCs | Whole blood | mRNA, lncRNA | NGS and qPCR | Genome wide | Higher levels of the NR_028138.1 lncRNA in patients with BD compared with HCs |
[21] | Discovery cohort: 3 patients with BD II and 3 HCs; validation cohort: 99 patients with BD II and 115 HCs | Serum | miRNA | NGS and qPCR | Genome wide | Levels of miR-7-5p, miR-23b-3p, miR-142-3p, miR-221-5p and miR-370-3p were higher in patients with BD II compared with HCs |
[32] | 51 patients with BD and 116 HCs | White blood | mRNA | qPCR | COMT, GCAT, NRG1, PSAT1, SHMT2, SLC1A4 and SRR | A logistic ridge regression model including age, gender and mRNA expression levels of the 7 NMDAR genes showed an AUC of 0.92 in differentiating patients with BD from HCs |
[29] | 20 patients with BD, 20 with MDD and 20 HCs | Whole blood | miRNA | Microarray and qPCR | Genome wide | 5 miRNAs showed higher levels specifically in patients with BD compared with HCs (hsa-miR-140-3p, hsa-miR-30d-5p, hsa-miR-330-5p, hsa-miR-378a-5p and hsa-miR-21-3p), while hsa-miR-330-3p and hsa-miR-345-5p showed higher levels in both BD and MDD |
[30] | 19 patients with BD, 20 with SZ and 20 HCs | PBMC | circRNA | NGS and qPCR | Genome wide | Levels of 30 circRNAs were specifically altered in patients with BD compared with HCs |
[35] | 50 patients with BD and 50 HCs | PBMC | lncRNA | qPCR | 3 lncRNAs related to oxidative stress (lincRNA-p21, lincRNA-ROR and lincRNA-PINT) | Levels of all lncRNAs were lower in patients with BD, and specifically in male patients with BD, compared with HCs |
[36] | 50 patients with BD and 50 HCs | PBMC | lncRNA | qPCR | 5 lncRNAs related to oxidative stress (H19, LUCAT1, RMST, MEG3 and MT1DP) | Levels of LUCAT1, RMST and MEG3 were lower in patients with BD, specifically in male patients with BD, compared with HCs |
[37] | 63 patients with BD, 42 with MDD and 55 HCs | PBMC | miRNA | qPCR | miR-499-5p | Higher levels of miR-499-5p in patients with BD (regardless of disease phase) compared with HCs |
[22] | Serum: 41 patients with BD, 43 with MDD and 93 HCs; fibroblasts: 12 patients with BD, 23 with MDD and 15 HCs | Serum and fibroblasts | mRNA | qPCR | IGFBP2 | Lower IGFBP2 levels exclusively in patients with BD compared with HCs |
[38] | 37 rapid cycling patients with BD in different affective states and 40 HCs | PBMC | mRNA | qPCR | 19 candidate genes | Lower levels of POLG and OGG1 in patients with BD compared with HCs; higher levels of NDUFV2 in patients in a depressed state compared with those in an euthymic state. A gene expression score including all genes showed an AUC of 0.73 in separating patients from HCs |
[39] | 50 patients with BD and 50 HCs | PBMC | lncRNA | qPCR | DISC1 and DISC2 | Lower levels of DISC1 and higher levels of DISC2 in patients with BD compared with HCs (AUC: 0.76 and 0.68, respectively) |
[40] | 50 patients with BD and 50 HCs | PBMC | lncRNA | qPCR | 6 apoptosis-related lncRNAs (CCAT2, TUG1, PANDA, NEAT1, FAS-AS1 and OIP5-AS1) | Levels of CCAT2, TUG1 and PANDA were higher and levels of OIP5-AS1 were lower in patients with BD patients compared with HCs. CCAT2 and TUG1 expression levels were only different in male subgroups |
[23] | 11 drug-free manic psychotic patients with BD and 9 HCs | Plasma | miRNA | Nanostring and qPCR | Genome wide | Higher levels of hsa-miR-25-3p and hsa-miR-144-3p, and lower levels of hsa-miR-6721-5p in patients with BD compared with HCs |
[41] | 66 patients with BD and 66 HCs | Plasma | miRNA | qPCR | 15 miRNAs | A model including levels of miR-15b-5p, miR-155-5p, miR-134-5p and miR-652-3p showing a 83.3% sensitivity and 78.8% specificity |
[42] | 56 patients with BD I and 52 HCs | Whole blood | miRNA | qPCR | hsa-miR-145-5p, hsa-miR-376a-3p, hsa-miR-3680-5p, hsa-miR-4253-5p, hsa-miR-4482-3p and hsa-miR-4725 | Higher levels of hsa-miR-376a-3p, hsa-miR-3680-5p, hsa-miR-4253-5p, hsa-miR-4482-3p and lower levels of hsa-miR-145-5p in patients with BD compared with HCs |
[43] | 50 patients with BD and 50 HCs | Whole blood | lncRNA | qPCR | Five NF-κB-associated lncRNAs (ANRIL, CEBPA-DT, H19, NKILA and HNF1A-AS1) | Lower levels of ANRIL, CEBPA-DT and HNF1-AS1 and higher levels of NKILA in patients with BD compared with HC. HFN1A-AS1 showed the best diagnostic parameters (AUC: 0.86). |
2.2. Levels of RNA Markers in Cellular Models Derived from Patients with BD
3. RNA Biomarkers and Response to Mood Stabilizers
3.1. Peripheral Levels of RNA Markers in Biofluids or Peripheral Cells from Patients with BD Characterized for Response to Mood Stabilizers
Ref. | Sample | RNA Source | RNA Type | Measurement Method | Targets | Main Findings |
---|---|---|---|---|---|---|
[61] | 20 patients with BD (9 during a hypomanic episode, 11 during a depressive episode) treated with lithium for 8 weeks and 15 HCs. Response was evaluated with the HDRS, YMRS, CGI-I and CGI-S | Whole blood | mRNA | Microarray | Genome wide | 13 genes showed a nominally significant change after treatment, and this change was correlated with a change in the clinical severity measured with the CGI-S score |
[62] | 21 patients treated with lithium for 8 weeks. Response was evaluated with HDRS, YMRS, CGI-I and CGI-S, 16 HCs | Whole blood | mRNA | Microarray | Genome wide | The sphingomyelin metabolism pathway was associated with the change in the HDRS score, and this effect was found to be mediated by the volume of the mediodorsal thalamus measured with brain MRI scans |
[59] | 60 patients treated with OPT or OPT + lithium moderate dose for 6 months. Response was evaluated with the CGI-BP-S | Whole blood | mRNA | Microarray and qPCR | Genome wide | In patients treated with lithium, 62 genes were differentially regulated in responders compared with non-responders, with BCL2L1 showing the greatest difference |
[63] | 21 patients with BD treated with lithium and antipsychotics and 20 HCs. Response was evaluated with the YMRS | Whole blood | mRNA | qPCR | TERT | TERT levels were upregulated both at baseline and at remission in patients with BD compared with HCs |
[60] | 20 patients treated with lithium for 8 weeks and evaluated with the HDRS | Whole blood | mRNA | Microarray | Genome wide | Genes differentially expressed between responders and non-responders were enriched for the regulation of apoptosis pathway. After 4 weeks, anti-apoptotic genes such as BCL2 and IRS2 were upregulated in responders and downregulated in non-responders, while pro-apoptotic genes such as BAK1 and BAD were downregulated in responders and upregulated in non-responders. These changes were not significant anymore at 8 weeks |
[64] | 25 patients with BD during a major depressive episode, treated with lithium for 6 weeks, 31 HCs. Response was evaluated with the HDRS | Whole blood | mRNA | qPCR | 5 genes part of the AKT/mTOR pathway | Significant association between changes in levels of AKT-1, BCL2 and changes in the HDRS score after lithium treatment; baseline BCL2 levels predicted improvement of depressive symptoms after lithium therapy |
[57] | 19 pediatric patients with BD treated with mood stabilizers for 8 weeks and evaluated with the YMRS | Whole blood | mRNA | qPCR | BDNF | Positive correlation between the change in BDNF levels and the change in the YMRS score at 8 weeks |
[58] | 21 patients with BD, drug-free at the first sampling. Response to treatment with antipsychotics and mood stabilizers was evaluated with the BRMS at 2 and 4 weeks | Plasma | mRNA | qPCR | miR-134 | Negative correlation between miR-134 levels at baseline, 2-week or 4-week follow-up and severity of manic symptoms |
[65] | 50 patients with long-term lithium treatment and evaluated with the Alda scale | Whole blood | mRNA | NGS | Genome wide | Nominal association between lithium response and a co-expression module (the central modulators of which were mitochondrially encoded genes). A total of 43 out of the 46 genes included in this module showed reduced levels in responders compared with non-responders |
3.2. Levels of RNA Markers in Cellular Models Derived from Patients with BD Characterized for Response to Mood Stabilizers
Ref. | Sample | RNA Source | RNA Type | Measurement Method | Targets | Main Findings |
---|---|---|---|---|---|---|
[70] | 16 patients treated with lithium. Response was evaluated based on the rate of relapse | LCLs | mRNA | NGS | Genome wide | In vitro treatment with LiCl 1 mM for 1 week modulated 22 coexpression modules |
[66] | 20 patients with BD (11 responders and 9 non-responders). Response was evaluated with the Alda scale | LCLs | mRNA, miRNA | Microarray | Genome wide | 335 genes (217 upregulated and 118 downregulated) and 77 miRNAs (46 upregulated and 31 downregulated) were nominally differentially expressed between responders and non-responders |
[72] | 36 patients with long-term lithium treatment. Response was evaluated with the Alda scale | LCLs | mRNA | qPCR | 20 circadian genes | Differential temporal evolution between non-responders and responders for levels of different circadian genes |
[67] | 16 patients with long-term lithium treatment. Response was evaluated with the Alda scale | LCLs | mRNA, miRNA | Microarray, qPCR | Genome wide | In vitro treatment with LiCl 1 mM for 1 week induced downregulation of THRAP3 and TFAM in responders |
[77] | 8 patients with BD with long-term lithium treatment. Response was evaluated with the Alda scale | LCLs, fibroblasts | mRNA | Microarray, qPCR | Genome wide | No significant difference in gene expression levels based on lithium response |
[71] | 22 patients treated with lithium for 6 weeks. Response was evaluated with the MADRS and YMRS at 6 weeks | Olfactory neurons | mRNA | qPCR | GSK3B, AKT1, PRKCE and CRMP1 | Treatment-associated downregulation of CRMP1 predicted improvement of both manic and depressive symptoms |
[75] | 6 patients treated with lithium. Response was evaluated with the CGI at 4 months | Neurons differentiated from iPSCs | mRNA | NGS | Genome wide | In vitro treatment with LiCl 1 mM for 1 week modulated 560 genes in responders and 40 genes in non-responders. Genes for which lithium rescued expression in responders were related to the PKA/PKC pathways, action potential firing and mitochondria |
[78] | 17 patients with long-term lithium treatment. Response was evaluated with the Alda scale | LCLs | miRNA | qPCR | let7-c | Nonsignificant trend for higher let-7c expression in non-responders compared with responders |
[79] | 24, 41 and 17 patients with long-term lithium treatment. Response was evaluated with the Alda scale | LCLs | mRNA | NGS, qPCR | Genome wide | 56 genes showed nominal differential expression between responders and non-responders. HDGFRP3 and ID2 were validated in the independent cohorts |
[80] | 36 patients with BD with long-term lithium treatment. Response was evaluated with the Alda scale | LCLs | mRNA | qPCR | GADL1 | No difference in GADL1 levels between responders and non-responders. In vitro treatment with LiCl 1 mM for 4 or 8 days did not modify GADL1 levels |
[74] | 6 patients with BD responders to lithium, 5 patients with BD non-responders to lithium and 6 HCs. Response was evaluated with the Alda scale | Neurons differentiated from iPSCs | mRNA | NGS, qPCR | Genome wide | 41 genes were differentially expressed between responders and non-responders, regardless of in vitro treatment with LiCl 1 mM for 1 week. Focal adhesion and the extracellular matrix were the most significant functions based on functional enrichment analysis of the top 500 proximal network genes |
[81] | 20 patients with long-term lithium treatment. Response was evaluated with the Alda scale | LCLs | mRNA | Microarray, qPCR | RBM3 | RBM3 was upregulated in responders compared with non-responders. In vitro treatment with LiCl 1 mM for 1 week did not modify RBM3 levels |
[82] | LCL: 25 patients with long-term lithium treatment and 12 HCs. NPC: 2 patients with BD. Response was evaluated with the Alda scale | LCLs, NPCs | mRNA | NGS, qPCR | BCL2, GSK3B and NR1D1 | In vitro treatment with LiCl 1 mm for 1 week increased the expression of BCL2 and GSK3B in responders |
[83] | 20 patients with long-term lithium treatment. Response was evaluated with the Alda scale | LCLs | mRNA | Microarray, qPCR | Genome wide | In vitro treatment with LiCl 1 mm for 1 week modified levels of 29 genes, including ZNF493 and ZNF429, in responders |
[73] | 20 patients with long-term lithium treatment. Response was evaluated with the Alda scale | LCLs | mRNA | Microarray | 17 circadian genes | Higher levels of BHLHE40 in responders compared with non-responders |
[68] | 20 patients with long-term lithium treatment. Response was evaluated with the Alda scale | LCLs | miRNA, mRNA | Microarray, NGS, qPCR | Genome wide | miR-320a, miR-155-3p and three of their targeted genes (CAPNS1 and RGS16 for miR-320 and SP4 for miR-155-3p) were differentially expressed between responders and non-responders |
[76] | 3 patients with BD responders to lithium, 3 non-responders and 4 HCs. Response was evaluated with the Alda scale | Neurons differentiated from iPSCs | mRNA | NGS, qPCR | Genome wide | Alterations of the Wnt/β-catenin signaling pathway and decreased levels of the LEF1 transcription factor, which were observed in neurons derived from lithium non-responders |
[84] | 30 patients with long-term lithium treatment. Response was evaluated with the Alda scale | LCLs | mRNA | qPCR | PDLIM5 | No association between PDLIM5 levels and response |
[85] | 20 and 12 patients with long-term lithium treatment. Response was evaluated with the Alda scale | LCLs | mRNA | Microarray, qPCR | Genome wide | 2060 genes were differentially expressed between responders and non-responders; IGF1 was validated in the independent sample |
[86] | 12 patients (all responders to long-term lithium treatment according to [87]) | LCLs | mRNA | Microarray, Northern blot | Genome wide | In vitro treatment with LiCl 1 mM for 1 week decreased the expression of 7 genes |
4. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Grande, I.; Berk, M.; Birmaher, B.; Vieta, E. Bipolar disorder. Lancet 2016, 387, 1561–1572. [Google Scholar] [CrossRef]
- Stahl, E.A.; Breen, G.; Forstner, A.J.; McQuillin, A.; Ripke, S.; Trubetskoy, V.; Mattheisen, M.; Wang, Y.; Coleman, J.R.I.; Gaspar, H.A.; et al. Genome-wide association study identifies 30 loci associated with bipolar disorder. Nat. Genet. 2019, 51, 793–803. [Google Scholar] [CrossRef]
- Mullins, N.; Forstner, A.J.; O’Connell, K.S.; Coombes, B.; Coleman, J.R.I.; Qiao, Z.; Als, T.D.; Bigdeli, T.B.; Børte, S.; Bryois, J.; et al. Genome-wide association study of more than 40,000 bipolar disorder cases provides new insights into the underlying biology. Nat. Genet. 2021, 53, 817–829. [Google Scholar] [CrossRef] [PubMed]
- Grof, P.; Duffy, A.; Cavazzoni, P.; Grof, E.; Garnham, J.; MacDougall, M.; O’Donovan, C.; Alda, M. Is Response to Prophylactic Lithium a Familial Trait? J. Clin. Psychiatry 2002, 63, 942–947. [Google Scholar] [CrossRef] [PubMed]
- Hou, L.; Heilbronner, U.; Degenhardt, F.; Adli, M.; Akiyama, K.; Akula, N.; Ardau, R.; Arias, B.; Backlund, L.; Banzato, C.E.M.; et al. Genetic variants associated with response to lithium treatment in bipolar disorder: A genome-wide association study. Lancet 2016, 387, 1085–1093. [Google Scholar] [CrossRef] [Green Version]
- Amare, A.T.; Schubert, K.O.; Hou, L.; Clark, S.R.; Papiol, S.; Cearns, M.; Heilbronner, U.; Degenhardt, F.; Tekola-Ayele, F.; Hsu, Y.-H.; et al. Association of polygenic score for major depression with response to lithium in patients with bipolar disorder. Mol. Psychiatry 2021, 26, 2457–2470. [Google Scholar] [CrossRef]
- International Consortium on Lithium Genetics (ConLi+Gen). Association of Polygenic Score for Schizophrenia and HLA Antigen and Inflammation Genes With Response to Lithium in Bipolar Affective Disorder: A Genome-Wide Association Study. JAMA Psychiatry 2018, 75, 65–74. [Google Scholar] [CrossRef]
- Schubert, K.O.; Thalamuthu, A.; Amare, A.T.; Frank, J.; Streit, F.; Adl, M.; Akula, N.; Akiyama, K.; Ardau, R.; Arias, B.; et al. Combining schizophrenia and depression polygenic risk scores improves the genetic prediction of lithium response in bipolar disorder patients. Transl. Psychiatry 2021, 11, 606. [Google Scholar] [CrossRef] [PubMed]
- Pisanu, C.; Heilbronner, U.; Squassina, A. The Role of Pharmacogenomics in Bipolar Disorder: Moving Towards Precision Medicine. Mol. Diagn. Ther. 2018, 22, 409–420. [Google Scholar] [CrossRef] [PubMed]
- Vozikis, A.; Cooper, D.N.; Mitropoulou, C.; Kambouris, M.E.; Brand, A.; Dolzan, V.; Fortina, P.; Innocenti, F.; Lee, M.T.M.; Leyens, L.; et al. Test Pricing and Reimbursement in Genomic Medicine: Towards a General Strategy. Public Health Genom. 2016, 19, 352–363. [Google Scholar] [CrossRef] [Green Version]
- Ha, M.; Kim, V.N. Regulation of microRNA biogenesis. Nat. Rev. Mol. Cell Biol. 2014, 15, 509–524. [Google Scholar] [CrossRef] [PubMed]
- Im, H.-I.; Kenny, P.J. MicroRNAs in neuronal function and dysfunction. Trends Neurosci. 2012, 35, 325–334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vo, J.N.; Cieslik, M.; Zhang, Y.; Shukla, S.; Xiao, L.; Zhang, Y.; Wu, Y.-M.; Dhanasekaran, S.M.; Engelke, C.G.; Cao, X.; et al. The Landscape of Circular RNA in Cancer. Cell 2019, 176, 869–881.e813. [Google Scholar] [CrossRef] [Green Version]
- Chen, W.; Schuman, E. Circular RNAs in Brain and Other Tissues: A Functional Enigma. Trends Neurosci. 2016, 39, 597–604. [Google Scholar] [CrossRef] [PubMed]
- Statello, L.; Guo, C.-J.; Chen, L.-L.; Huarte, M. Gene regulation by long non-coding RNAs and its biological functions. Nat. Rev. Mol. Cell Biol. 2021, 22, 96–118. [Google Scholar] [CrossRef] [PubMed]
- Gecys, D.; Dambrauskiene, K.; Simonyte, S.; Patamsyte, V.; Vilkeviciute, A.; Musneckis, A.; Butkute-Sliuoziene, K.; Lesauskaite, V.; Zemaitis, L.; Usaite, D.; et al. Circulating hsa-let-7e-5p and hsa-miR-125a-5p as Possible Biomarkers in the Diagnosis of Major Depression and Bipolar Disorders. Dis. Markers 2022, 2022, 3004338. [Google Scholar] [CrossRef]
- Fu, Y.; He, W.; Zhou, C.; Fu, X.; Wan, Q.; He, L.; Wei, B. Bioinformatics Analysis of circRNA Expression and Construction of “circRNA-miRNA-mRNA” Competing Endogenous RNAs Networks in Bipolar Disorder Patients. Front. Genet. 2021, 12, 718976. [Google Scholar] [CrossRef]
- Eghtedarian, R.; Ghafouri-Fard, S.; Bouraghi, H.; Hussen, B.M.; Arsang-Jang, S.; Taheri, M. Abnormal pattern of vitamin D receptor-associated genes and lncRNAs in patients with bipolar disorder. BMC Psychiatry 2022, 22, 178. [Google Scholar] [CrossRef]
- He, L.; Zou, P.; Sun, W.; Fu, Y.; He, W.; Li, J. Identification of lncRNA NR_028138.1 as a biomarker and construction of a ceRNA network for bipolar disorder. Sci. Rep. 2021, 11, 15653. [Google Scholar] [CrossRef]
- Chen, Y.; Shi, J.; Liu, H.; Wang, Q.; Chen, X.; Tang, H.; Yan, R.; Yao, Z.; Lu, Q. Plasma microRNA Array Analysis Identifies Overexpressed miR-19b-3p as a Biomarker of Bipolar Depression Distinguishing From Unipolar Depression. Front. Psychiatry 2020, 11, 757. [Google Scholar] [CrossRef]
- Lee, S.-Y.; Lu, R.-B.; Wang, L.-J.; Chang, C.-H.; Lu, T.; Wang, T.-Y.; Tsai, K.-W. Serum miRNA as a possible biomarker in the diagnosis of bipolar II disorder. Sci. Rep. 2020, 10, 1131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Milanesi, E.; Zanardini, R.; Rosso, G.; Maina, G.; Barbon, A.; Mora, C.; Minelli, A.; Gennarelli, M.; Bocchio-Chiavetto, L. Insulin-like growth factor binding protein 2 in bipolar disorder: An expression study in peripheral tissues. World J. Biol. Psychiatry 2018, 19, 610–618. [Google Scholar] [CrossRef] [PubMed]
- Tabano, S.; Caldiroli, A.; Terrasi, A.; Colapietro, P.; Grassi, S.; Carnevali, G.S.; Fontana, L.; Serati, M.; Vaira, V.; Altamura, A.C.; et al. A miRNome analysis of drug-free manic psychotic bipolar patients versus healthy controls. Eur. Arch. Psychiatry Clin. Neurosci. 2020, 270, 893–900. [Google Scholar] [CrossRef] [PubMed]
- Fries, G.R.; Lima, C.N.; Valvassori, S.S.; Zunta-Soares, G.; Soares, J.C.; Quevedo, J. Preliminary investigation of peripheral extracellular vesicles’ microRNAs in bipolar disorder. J. Affect. Disord. 2019, 255, 10–14. [Google Scholar] [CrossRef]
- Ceylan, D.; Tufekci, K.U.; Keskinoglu, P.; Genc, S.; Özerdem, A. Circulating exosomal microRNAs in bipolar disorder. J. Affect. Disord. 2020, 262, 99–107. [Google Scholar] [CrossRef]
- Huo, L.; Du, X.; Li, X.; Liu, S.; Xu, Y. The Emerging Role of Neural Cell-Derived Exosomes in Intercellular Communication in Health and Neurodegenerative Diseases. Front. Neurosci. 2021, 15, 738442. [Google Scholar] [CrossRef]
- Camkurt, M.A.; Karababa, I.F.; Erdal, M.E.; Kandemir, S.B.; Fries, G.R.; Bayazıt, H.; Ay, M.E.; Kandemir, H.; Ay, O.I.; Coşkun, S.; et al. MicroRNA dysregulation in manic and euthymic patients with bipolar disorder. J. Affect. Disord. 2020, 261, 84–90. [Google Scholar] [CrossRef]
- Banach, E.; Dmitrzak-Weglarz, M.; Pawlak, J.; Kapelski, P.; Szczepankiewicz, A.; Rajewska-Rager, A.; Slopien, A.; Skibinska, M.; Czerski, P.; Hauser, J. Dysregulation of miR-499, miR-708 and miR-1908 during a depression episode in bipolar disorders. Neurosci. Lett. 2017, 654, 117–119. [Google Scholar] [CrossRef]
- Maffioletti, E.; Cattaneo, A.; Rosso, G.; Maina, G.; Maj, C.; Gennarelli, M.; Tardito, D.; Bocchio-Chiavetto, L. Peripheral whole blood microRNA alterations in major depression and bipolar disorder. J. Affect. Disord. 2016, 200, 250–258. [Google Scholar] [CrossRef] [Green Version]
- Mahmoudi, E.; Green, M.J.; Cairns, M.J. Dysregulation of circRNA expression in the peripheral blood of individuals with schizophrenia and bipolar disorder. J. Mol. Med. 2021, 99, 981–991. [Google Scholar] [CrossRef]
- Bocchio-Chiavetto, L.; Maffioletti, E.; Bettinsoli, P.; Giovannini, C.; Bignotti, S.; Tardito, D.; Corrada, D.; Milanesi, L.; Gennarelli, M. Blood microRNA changes in depressed patients during antidepressant treatment. Eur. Neuropsychopharmacol. 2013, 23, 602–611. [Google Scholar] [CrossRef] [PubMed]
- Lin, E.; Lin, C.-H.; Lane, H.-Y. Logistic ridge regression to predict bipolar disorder using mRNA expression levels in the N-methyl-D-aspartate receptor genes. J. Affect. Disord. 2021, 297, 309–313. [Google Scholar] [CrossRef] [PubMed]
- D’Addario, C.; Palazzo, M.C.; Benatti, B.; Grancini, B.; Pucci, M.; Di Francesco, A.; Camuri, G.; Galimberti, D.; Fenoglio, C.; Scarpini, E.; et al. Regulation of gene transcription in bipolar disorders: Role of DNA methylation in the relationship between prodynorphin and brain derived neurotrophic factor. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2018, 82, 314–321. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dieset, I.; Djurovic, S.; Tesli, M.; Hope, S.; Mattingsdal, M.; Michelsen, A.; Joa, I.; Larsen, T.K.; Agartz, I.; Melle, I.; et al. Up-Regulation of NOTCH4 Gene Expression in Bipolar Disorder. Am. J. Psychiatry 2012, 169, 1292–1300. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Maloum, Z.; Ramezani, S.; Taheri, M.; Ghafouri-Fard, S.; Shirvani-Farsani, Z. Downregulation of long non-coding RNAs in patients with bipolar disorder. Sci. Rep. 2022, 12, 7479. [Google Scholar] [CrossRef] [PubMed]
- Maloum, Z.; Taheri, M.; Ghafouri-Fard, S.; Shirvani-Farsani, Z. Significant reduction of long non-coding RNAs expression in bipolar disorder. BMC Psychiatry 2022, 22, 256. [Google Scholar] [CrossRef]
- Martins, H.C.; Gilardi, C.; Sungur, A.; Winterer, J.; Pelzl, M.A.; Bicker, S.; Gross, F.; Kisko, T.M.; Malikowska-Racia, N.; Braun, M.D.; et al. Bipolar-associated miR-499-5p controls neuroplasticity by downregulating the Cav1.2 subunit CACNB2. EMBO Rep. 2022, 23, e54420. [Google Scholar] [CrossRef]
- Munkholm, K.; Peijs, L.; Vinberg, M.; Kessing, L.V. A composite peripheral blood gene expression measure as a potential diagnostic biomarker in bipolar disorder. Transl. Psychiatry 2015, 5, e614. [Google Scholar] [CrossRef] [Green Version]
- Naghavi-Gargari, B.; Zahirodin, A.; Ghaderian, S.M.H.; Shirvani-Farsani, Z. Significant increasing of DISC2 long non-coding RNA expression as a potential biomarker in bipolar disorder. Neurosci. Lett. 2019, 696, 206–211. [Google Scholar] [CrossRef]
- Sayad, A.; Taheri, M.; Omrani, M.D.; Fallah, H.; Oskooei, V.K.; Ghafouri-Fard, S. Peripheral expression of long non-coding RNAs in bipolar patients. J. Affect. Disord. 2019, 249, 169–174. [Google Scholar] [CrossRef]
- Tekdemir, R.; Selvi, Y.; Altınbaş, K.; Koçak, N. Decreased miR-15b-5p/miR-155-5p levels and increased miR-134-5p/miR-652-3p levels among BD patients under lithium treatment. J. Affect. Disord. 2022, 317, 6–14. [Google Scholar] [CrossRef] [PubMed]
- Tekin, S.S.; Erdal, M.E.; Asoğlu, M.; Ay, I.; Ay, M.E.; Yılmaz, G. Biomarker potential of hsa-miR-145-5p in peripheral whole blood of manic bipolar I patients. Braz. J. Psychiatry 2022, 40, 378–387. [Google Scholar] [CrossRef] [PubMed]
- Teshnizi, S.A.; Shahani, P.; Taheri, M.; Hussen, B.M.; Eslami, S.; Sadeghzadeh, Z.; Ghafouri-Fard, S.; Sayad, A. Expression analysis of NF-ƙB-related long non-coding RNAs in bipolar disorder. Sci. Rep. 2022, 12, 20941. [Google Scholar] [CrossRef]
- Fries, G.R.; Colpo, G.D.; Monroy-Jaramillo, N.; Zhao, J.; Zhao, Z.; Arnold, J.G.; Bowden, C.L.; Walss-Bass, C. Distinct lithium-induced gene expression effects in lymphoblastoid cell lines from patients with bipolar disorder. Eur. Neuropsychopharmacol. 2017, 27, 1110–1119. [Google Scholar] [CrossRef] [PubMed]
- Courtin, C.; Marie-Claire, C.; Gross, G.; Hennion, V.; Mundwiller, E.; Guégan, J.; Meyrel, M.; Bellivier, F.; Etain, B. Gene expression of circadian genes and CIART in bipolar disorder: A preliminary case-control study. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2023, 122, 110691. [Google Scholar] [CrossRef] [PubMed]
- Costa, M.; Squassina, A.; Piras, I.S.; Pisanu, C.; Congiu, D.; Niola, P.; Angius, A.; Chillotti, C.; Ardau, R.; Severino, G.; et al. Preliminary Transcriptome Analysis in Lymphoblasts from Cluster Headache and Bipolar Disorder Patients Implicates Dysregulation of Circadian and Serotonergic Genes. J. Mol. Neurosci. 2015, 56, 688–695. [Google Scholar] [CrossRef]
- Squassina, A.; Manchia, M.; Chillotti, C.; Deiana, V.; Congiu, D.; Paribello, F.; Roncada, P.; Soggiu, A.; Piras, C.; Urbani, A.; et al. Differential effect of lithium on spermidine/spermine N1-acetyltransferase expression in suicidal behaviour. Int. J. Neuropsychopharmacol. 2013, 16, 2209–2218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fiori, L.M.; Turecki, G. Implication of the polyamine system in mental disorders. J. Psychiatry Neurosci. 2008, 33, 102–110. [Google Scholar]
- Squassina, A.; Niola, P.; Lopez, J.P.; Cruceanu, C.; Pisanu, C.; Congiu, D.; Severino, G.; Ardau, R.; Chillotti, C.; Alda, M.; et al. MicroRNA expression profiling of lymphoblasts from bipolar disorder patients who died by suicide, pathway analysis and integration with postmortem brain findings. Eur. Neuropsychopharmacol. 2020, 34, 39–49. [Google Scholar] [CrossRef]
- Bavamian, S.; Mellios, N.; Lalonde, J.; Fass, D.M.; Wang, J.; Sheridan, S.D.; Madison, J.M.; Zhou, F.; Rueckert, E.H.; Barker, D.; et al. Dysregulation of miR-34a links neuronal development to genetic risk factors for bipolar disorder. Mol. Psychiatry 2015, 20, 573–584. [Google Scholar] [CrossRef] [Green Version]
- Kathuria, A.; Lopez-Lengowski, K.; Vater, M.; McPHIE, D.; Cohen, B.M.; Karmacharya, R. Transcriptome analysis and functional characterization of cerebral organoids in bipolar disorder. Genome Med. 2020, 12, 34. [Google Scholar] [CrossRef] [Green Version]
- Bame, M.; McInnis, M.G.; O’Shea, K.S. MicroRNA Alterations in Induced Pluripotent Stem Cell-Derived Neurons from Bipolar Disorder Patients: Pathways Involved in Neuronal Differentiation, Axon Guidance, and Plasticity. Stem Cells Dev. 2020, 29, 1145–1159. [Google Scholar] [CrossRef] [PubMed]
- Ishima, T.; Illes, S.; Iwayama, Y.; Dean, B.; Yoshikawa, T.; Ågren, H.; Funa, K.; Hashimoto, K. Abnormal gene expression of BDNF, but not BDNF-AS, in iPSC, neural stem cells and postmortem brain samples from bipolar disorder. J. Affect. Disord. 2021, 290, 61–64. [Google Scholar] [CrossRef]
- Kim, H.K.; Tyryshkin, K.; Elmi, N.; Dharsee, M.; Evans, K.R.; Good, J.; Javadi, M.; McCormack, S.; Vaccarino, A.L.; Zhang, X.; et al. Plasma microRNA expression levels and their targeted pathways in patients with major depressive disorder who are responsive to duloxetine treatment. J. Psychiatr. Res. 2019, 110, 38–44. [Google Scholar] [CrossRef] [PubMed]
- Madison, J.M.; Zhou, F.; Nigam, A.; Hussain, A.; Barker, D.D.; Nehme, R.; van der Ven, K.; Hsu, J.; Wolf, P.; Fleishman, M.; et al. Characterization of bipolar disorder patient-specific induced pluripotent stem cells from a family reveals neurodevelopmental and mRNA expression abnormalities. Mol. Psychiatry 2015, 20, 703–717. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sawada, T.; Chater, T.E.; Sasagawa, Y.; Yoshimura, M.; Fujimori-Tonou, N.; Tanaka, K.; Benjamin, K.J.M.; Paquola, A.C.M.; Erwin, J.A.; Goda, Y.; et al. Developmental excitation-inhibition imbalance underlying psychoses revealed by single-cell analyses of discordant twins-derived cerebral organoids. Mol. Psychiatry 2020, 25, 2695–2711. [Google Scholar] [CrossRef]
- Pandey, G.N.; Rizavi, H.S.; Dwivedi, Y.; Pavuluri, M.N. Brain-Derived Neurotrophic Factor Gene Expression in Pediatric Bipolar Disorder: Effects of Treatment and Clinical Response. J. Am. Acad. Child Adolesc. Psychiatry 2008, 47, 1077–1085. [Google Scholar] [CrossRef]
- Rong, H.; Liu, T.B.; Yang, K.J.; Yang, H.C.; Wu, D.H.; Liao, C.P.; Hong, F.; Yang, H.Z.; Wan, F.; Ye, X.Y.; et al. MicroRNA-134 plasma levels before and after treatment for bipolar mania. J. Psychiatr. Res. 2011, 45, 92–95. [Google Scholar] [CrossRef]
- Beech, R.D.; Leffert, J.J.; Lin, A.; Sylvia, L.G.; Umlauf, S.; Mane, S.; Zhao, H.; Bowden, C.; Calabrese, J.R.; Friedman, E.S.; et al. Gene-expression differences in peripheral blood between lithium responders and non-responders in the Lithium Treatment-Moderate dose Use Study (LiTMUS). Pharmacogenom. J. 2013, 14, 182–191. [Google Scholar] [CrossRef]
- Lowthert, L.; Leffert, J.; Lin, A.; Umlauf, S.; Maloney, K.; Muralidharan, A.; Lorberg, B.; Mane, S.; Zhao, H.; Sinha, R.; et al. Increased ratio of anti-apoptotic to pro-apoptotic Bcl2 gene-family members in lithium-responders one month after treatment initiation. Biol. Mood Anxiety Disord. 2012, 2, 15. [Google Scholar] [CrossRef] [Green Version]
- Anand, A.; McClintick, J.N.; Murrell, J.; Karne, H.; Nurnberger, J.I.; Edenberg, H.J. Effects of Lithium Monotherapy for Bipolar Disorder on Gene Expression in Peripheral Lymphocytes. Complex Psychiatry 2016, 2, 115–123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Anand, A.; Nakamura, K.; Spielberg, J.M.; Cha, J.; Karne, H.; Hu, B. Integrative analysis of lithium treatment associated effects on brain structure and peripheral gene expression reveals novel molecular insights into mechanism of action. Transl. Psychiatry 2020, 10, 103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Çinar, R.K. Telomere length and hTERT in mania and subsequent remission. Braz. J. Psychiatry 2018, 40, 19–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Machado-Vieira, R.; Zanetti, M.V.; Teixeira, A.L.; Uno, M.; Valiengo, L.L.; Soeiro-De-Souza, M.G.; Oba-Shinjo, S.M.; de Sousa, R.T.; Zarate, C.A.; Gattaz, W.F.; et al. Decreased AKT1/mTOR pathway mRNA expression in short-term bipolar disorder. Eur. Neuropsychopharmacol. 2015, 25, 468–473. [Google Scholar] [CrossRef] [Green Version]
- Stacey, D.; Schubert, K.O.; Clark, S.R.; Amare, A.T.; Milanesi, E.; Maj, C.; Leckband, S.G.; Shekhtman, T.; Kelsoe, J.R.; Gurwitz, D.; et al. A gene co-expression module implicating the mitochondrial electron transport chain is associated with long-term response to lithium treatment in bipolar affective disorder. Transl. Psychiatry 2018, 8, 183. [Google Scholar] [CrossRef] [Green Version]
- Cattane, N.; Courtin, C.; Mombelli, E.; Maj, C.; Mora, C.; Etain, B.; Bellivier, F.; Marie-Claire, C.; Cattaneo, A. Transcriptomics and miRNomics data integration in lymphoblastoid cells highlights the key role of immune-related functions in lithium treatment response in Bipolar disorder. BMC Psychiatry 2022, 22, 665. [Google Scholar] [CrossRef]
- Hunsberger, J.G.; Chibane, F.L.; Elkahloun, A.G.; Henderson, R.; Singh, R.; Lawson, J.; Cruceanu, C.; Nagarajan, V.; Turecki, G.; Squassina, A.; et al. Novel integrative genomic tool for interrogating lithium response in bipolar disorder. Transl. Psychiatry 2015, 5, e504. [Google Scholar] [CrossRef] [Green Version]
- Pisanu, C.; Papadima, E.M.; Melis, C.; Congiu, D.; Loizedda, A.; Orrù, N.; Calza, S.; Carcassi, C.; Severino, G.; Ardau, R.; et al. Whole Genome Expression Analyses of miRNAs and mRNAs Suggest the Involvement of miR-320a and miR-155-3p and their Targeted Genes in Lithium Response in Bipolar Disorder. Int. J. Mol. Sci. 2019, 20, 6040. [Google Scholar] [CrossRef] [Green Version]
- Manchia, M.; Adli, M.; Akula, N.; Ardau, R.; Aubry, J.-M.; Backlund, L.; Banzato, C.E.; Baune, B.T.; Bellivier, F.; Bengesser, S.; et al. Assessment of Response to Lithium Maintenance Treatment in Bipolar Disorder: A Consortium on Lithium Genetics (ConLiGen) Report. PLoS ONE 2013, 8, e65636. [Google Scholar] [CrossRef] [Green Version]
- Breen, M.S.; White, C.H.; Shekhtman, T.; Lin, K.; Looney, D.; Woelk, C.H.; Kelsoe, J.R. Lithium-responsive genes and gene networks in bipolar disorder patient-derived lymphoblastoid cell lines. Pharmacogenom. J. 2016, 16, 446–453. [Google Scholar] [CrossRef]
- McLean, C.K.; Narayan, S.; Lin, S.Y.; Rai, N.; Chung, Y.; Hipolito, M.S.; Cascella, N.G.; Nurnberger, J.I.; Ishizuka, K.; Sawa, A.S.; et al. Lithium-associated transcriptional regulation of CRMP1 in patient-derived olfactory neurons and symptom changes in bipolar disorder. Transl. Psychiatry 2018, 8, 81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Geoffroy, P.A.; Curis, E.; Courtin, C.; Moreira, J.; Morvillers, T.; Etain, B.; Laplanche, J.-L.; Bellivier, F.; Marie-Claire, C. Lithium response in bipolar disorders and core clock genes expression. World J. Biol. Psychiatry 2018, 19, 619–632. [Google Scholar] [CrossRef]
- Pisanu, C.; Congiu, D.; Melis, C.; Severino, G.; Angius, A.; Ardau, R.; Chillotti, C.; Del Zompo, M.; Squassina, A. Involvement of core clock genes in lithium response. World J. Biol. Psychiatry 2018, 19, 645–646. [Google Scholar] [CrossRef] [PubMed]
- Niemsiri, V.; Rosenthal, S.B.; Nievergelt, C.M.; Maihofer, A.X.; Marchetto, M.C.; Santos, R.; Shekhtman, T.; Alliey-Rodriguez, N.; Anand, A.; Balaraman, Y.; et al. Focal adhesion is associated with lithium response in bipolar disorder: Evidence from a network-based multi-omics analysis. Mol. Psychiatry 2023. [Google Scholar] [CrossRef] [PubMed]
- Mertens, J.; Wang, Q.W.; Kim, Y.; Yu, D.X.; Pham, S.; Yang, B.; Zheng, Y.; Diffenderfer, K.E.; Zhang, J.; Soltani, S.; et al. Differential responses to lithium in hyperexcitable neurons from patients with bipolar disorder. Nature 2015, 527, 95–99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Santos, R.; Linker, S.B.; Stern, S.; Mendes, A.P.D.; Shokhirev, M.N.; Erikson, G.; Randolph-Moore, L.; Racha, V.; Kim, Y.; Kelsoe, J.R.; et al. Deficient LEF1 expression is associated with lithium resistance and hyperexcitability in neurons derived from bipolar disorder patients. Mol. Psychiatry 2021, 26, 2440–2456. [Google Scholar] [CrossRef]
- Kittel-Schneider, S.; Hilscher, M.; Scholz, C.-J.; Weber, H.; Grünewald, L.; Schwarz, R.; Chiocchetti, A.G.; Reif, A. Lithium-induced gene expression alterations in two peripheral cell models of bipolar disorder. World J. Biol. Psychiatry 2019, 20, 462–475. [Google Scholar] [CrossRef]
- Milanesi, E.; Hadar, A.; Maffioletti, E.; Werner, H.; Shomron, N.; Gennarelli, M.; Schulze, T.G.; Costa, M.; Del Zompo, M.; Squassina, A.; et al. Insulin-like Growth Factor 1 Differentially Affects Lithium Sensitivity of Lymphoblastoid Cell Lines from Lithium Responder and Non-responder Bipolar Disorder Patients. J. Mol. Neurosci. 2015, 56, 681–687. [Google Scholar] [CrossRef]
- Milanesi, E.; Voinsky, I.; Hadar, A.; Srouji, A.; Maj, C.; Shekhtman, T.; Gershovits, M.; Gilad, S.; Chillotti, C.; Squassina, A.; et al. RNA sequencing of bipolar disorder lymphoblastoid cell lines implicates the neurotrophic factor HRP-3 in lithium’s clinical efficacy. World J. Biol. Psychiatry 2019, 20, 449–461. [Google Scholar] [CrossRef]
- Moreira, J.; Courtin, C.; Geoffroy, P.A.; Curis, E.; Bellivier, F.; Cynthia, M.-C. Lithium response in bipolar disorder: No difference in GADL1 gene expression between cell lines from excellent-responders and non-responders. Psychiatry Res. 2017, 251, 217–220. [Google Scholar] [CrossRef]
- Papadima, E.M.; Niola, P.; Melis, C.; Pisanu, C.; Congiu, D.; Cruceanu, C.; Lopez, J.P.; Turecki, G.; Ardau, R.; Severino, G.; et al. Evidence towards RNA Binding Motif (RNP1, RRM) Protein 3 (RBM3) as a Potential Biomarker of Lithium Response in Bipolar Disorder Patients. J. Mol. Neurosci. 2017, 62, 304–308. [Google Scholar] [CrossRef] [PubMed]
- Paul, P.; Iyer, S.; Nadella, R.K.; Nayak, R.; Chellappa, A.S.; Ambardar, S.; Sud, R.; Sukumaran, S.K.; Purushottam, M.; Jain, S.; et al. Lithium response in bipolar disorder correlates with improved cell viability of patient derived cell lines. Sci. Rep. 2020, 10, 7428. [Google Scholar] [CrossRef]
- Pisanu, C.; Congiu, D.; Costa, M.; Chillotti, C.; Ardau, R.; Severino, G.; Angius, A.; Heilbronner, U.; Hou, L.; McMahon, F.J.; et al. Convergent analysis of genome-wide genotyping and transcriptomic data suggests association of zinc finger genes with lithium response in bipolar disorder. Am. J. Med. Genet. Part B Neuropsychiatr. Genet. 2018, 177, 658–664. [Google Scholar] [CrossRef] [PubMed]
- Squassina, A.; Congiu, D.; Manconi, F.; Manchia, M.; Chillotti, C.; Lampus, S.; Severino, G.; Del Zompo, M. The PDLIM5 gene and lithium prophylaxis: An association and gene expression analysis in Sardinian patients with bipolar disorder. Pharmacol. Res. 2008, 57, 369–373. [Google Scholar] [CrossRef] [PubMed]
- Squassina, A.; Costa, M.; Congiu, D.; Manchia, M.; Angius, A.; Deiana, V.; Ardau, R.; Chillotti, C.; Severino, G.; Calza, S.; et al. Insulin-like growth factor 1 (IGF-1) expression is up-regulated in lymphoblastoid cell lines of lithium responsive bipolar disorder patients. Pharmacol. Res. 2013, 73, 1–7. [Google Scholar] [CrossRef]
- Sun, X.; Young, L.T.; Wang, J.-F.; Grof, P.; Turecki, G.; Rouleau, G.A.; Alda, M. Identification of Lithium-Regulated Genes in Cultured Lymphoblasts of Lithium Responsive Subjects with Bipolar Disorder. Neuropsychopharmacology 2004, 29, 799–804. [Google Scholar] [CrossRef]
- Grof, P.; Alda, M.; Gror, E.; Zvolsky, P.; Walsh, M. Lithium response and genetics of affective disorders. J. Affect. Disord. 1994, 32, 85–95. [Google Scholar] [CrossRef]
- Altamura, A.C.; Dell’Osso, B.; Berlin, H.A.; Buoli, M.; Bassetti, R.; Mundo, E. Duration of untreated illness and suicide in bipolar disorder: A naturalistic study. Eur. Arch. Psychiatry Clin. Neurosci. 2010, 260, 385–391. [Google Scholar] [CrossRef]
- Bessonova, L.; Ogden, K.; Doane, M.J.; O’Sullivan, A.K.; Tohen, M. The Economic Burden of Bipolar Disorder in the United States: A Systematic Literature Review. Clin. Outcomes Res. 2020, 12, 481–497. [Google Scholar] [CrossRef]
Ref. | Sample | RNA Source | RNA Type | Measurement Method | Targets | Main Findings |
---|---|---|---|---|---|---|
[52] | 3 patients with BD and 3 HCs | Neurons differentiated from iPSC-derived NPCs | mRNA, miRNA | Microarray, qPCR | Genome wide | Six (miR-128-3p, miR-138-2-3p, miR-195-5p, miR-382-5p, miR-487b-3p and miR-744-3p) and two miRNAs (miR-10b-5p and miR-10b-3p) showed higher or lower levels, respectively, in neurons derived from patients with BD compared with HCs |
[50] | iPSC-derived NPCs from 1 patient with BD and 1 HC; post-mortem brain samples from 29 patients with BD and 34 HCs | NPCs, post-mortem brain samples | mRNA, miRNA | Nanostring, qPCR | miR-34a and predicted targets | Increased miR-34a levels in the cerebellum of patients with BD compared with HCs. In NPCs, the enhancement of miR-34a expression impaired neuronal differentiation, expression of synaptic proteins and neuronal morphology |
[45] | 37 euthymic patients with BD I and 20 HCs | LCLs | mRNA | NGS, qPCR | 19 circadian genes | Lower levels of ARNTL and higher levels of CIART and BHLHE41 in patients with BD compared with HCs |
[44] | 62 patients with BD I and 17 HCs | LCLs | mRNA | Microarray, qPCR | Genome wide | No significant difference between patients with BD and HCs |
[53] | iPSC and NPCs from 6 patients with BD and 4 HCs; post-mortem brain samples from 35 patients with BD and 34 HCs (BA46), 15 patients with BD and 15 HCs (corpus callosum and BA8) | NPCs, iPSCs and post-mortem brain samples | mRNA, lncRNA | qPCR | BDNF and BDNF-AS | BDNF expression was lower in iPSCs but higher in NPCs from BD patients compared with HCs. BDNF expression was lower in BA46 but not in BA8 or corpus callosum from patients with BD compared with HCs |
[51] | 8 patients with BD I and 8 HCs | Brain organoids generated from iPSCs | mRNA | NGS | Genome wide | Downregulation of pathways involved in cell adhesion, neurodevelopment and synaptic biology and upregulation of genes involved in immune signaling in organoids from patients with BD compared with HCs. The central hub in the network analysis was the neurocan gene, located in a locus with evidence for genome-wide significant association for BD |
[54] | 4 patients with BD and 4 HCs | Neurons and NPCs derived from iPSCs | mRNA | Microarray, qPCR | Genome wide | 328 genes were differentially expressed neurons from patients with BD and HCs. These genes were enriched for alterations in RNA biosynthesis and metabolism, protein trafficking and receptor signaling pathways. Higher levels of GAD1 in neurons from patients with BD were confirmed with qPCR |
[55] | 2 brothers with BD and 2 unaffected parents | NPCs derived from iPSCs | mRNA | Nanostring, NGS | Genome wide | NPCs expressing CXCR4 from both BD patients compared to their unaffected parents showed differences in the expression of genes critical for neuroplasticity, including Wnt pathway components and ion channel subunits |
[56] | 2 monozygotic twins discordant for schizoaffective disorder, bipolar type, and 2 pairs of monozygotic twins discordant for SZ | Brain organoids generated from iPSCs | mRNA | scRNAseq | Genome wide | Enhanced GABAergic specification and reduced cell proliferation following diminished Wnt signaling in the patient with BD, which was confirmed in iPSC-derived forebrain neuronal cells |
[47] | 9 patients with BD who died by suicide, 17 at low risk of suicide, 17 at high risk of suicide and 21 HCs | LCLs | mRNA | qPCR | SAT1 | In vitro treatment with LiCl 1 mM increased SAT1 expression in the high and low risk groups as well as in HCs, but not in suicide completers |
[49] | 7 patients with BD who died by suicide, 11 patients at low risk of suicide and 12 HCs | LCLs, NPCs and post-mortem brain samples | miRNA | Nanostring, qPCR | Genome wide | Higher levels of miR-4286 and lower levels of miR-186-5p in LCLs from patients who died by suicide compared with patients at low risk of suicide and HCs. In vitro treatment with lithium reduced miR-4286 expression in human NPCs |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Pisanu, C.; Squassina, A. RNA Biomarkers in Bipolar Disorder and Response to Mood Stabilizers. Int. J. Mol. Sci. 2023, 24, 10067. https://doi.org/10.3390/ijms241210067
Pisanu C, Squassina A. RNA Biomarkers in Bipolar Disorder and Response to Mood Stabilizers. International Journal of Molecular Sciences. 2023; 24(12):10067. https://doi.org/10.3390/ijms241210067
Chicago/Turabian StylePisanu, Claudia, and Alessio Squassina. 2023. "RNA Biomarkers in Bipolar Disorder and Response to Mood Stabilizers" International Journal of Molecular Sciences 24, no. 12: 10067. https://doi.org/10.3390/ijms241210067