Next Article in Journal
The Structure and Functions of PRMT5 in Human Diseases
Next Article in Special Issue
Automated Exploration of Prebiotic Chemical Reaction Space: Progress and Perspectives
Previous Article in Journal
Current Therapeutic Approach to Acute Myocardial Infarction in Patients with Congenital Hemophilia
Previous Article in Special Issue
Understanding Vernal Keratoconjunctivitis: Beyond Allergic Mechanisms
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Uncovering the Roles of MicroRNAs in Major Depressive Disorder: From Candidate Diagnostic Biomarkers to Treatment Response Indicators

1
Department of Biochemistry, University of Medicine and Pharmacy Victor Babes Timisoara, 300041 Timișoara, Romania
2
Discipline of Psychiatry, Department of Neurosciences, University of Medicine and Pharmacy Victor Babes Timisoara, 300041 Timisoara, Romania
3
Eduard Pamfil Psychiatric Clinic, Timisoara County Emergency Clinical Hospital, 300041 Timisoara, Romania
*
Author to whom correspondence should be addressed.
Life 2021, 11(10), 1073; https://doi.org/10.3390/life11101073
Submission received: 16 September 2021 / Revised: 6 October 2021 / Accepted: 8 October 2021 / Published: 11 October 2021
(This article belongs to the Collection Feature Review Papers for Life)

Abstract

:
Major depressive disorder (MDD) is a recurrent debilitating illness that represents a major health burden due to its increasing worldwide prevalence, unclear pathological mechanism, nonresponsive treatment, and lack of reliable and specific diagnostic biomarkers. Recently, microRNA species (miRs) have gained particular interest because they have the ability to post-transcriptionally regulate gene expression by modulating mRNA stability and translation in a cohesive fashion. By regulating entire genetic circuitries, miRs have been shown to have dysregulated expression levels in blood samples from MDD patients, when compared to healthy subjects. In addition, antidepressant treatment (AD) also appears to alter the expression pattern of several miRs. Therefore, we critically and systematically reviewed herein the studies assessing the potential biomarker role of several candidate miRs for MDD, as well as treatment response monitoring indicators, in order to enrich the current knowledge and facilitate possible diagnostic biomarker development for MDD, which could aid in reducing both patients’ burden and open novel avenues toward a better understanding of MDD neurobiology.

1. Introduction

Major depressive disorder (MDD) is a complex neuropsychiatric disorder with an increasing incidence and a 2–20% prevalence in the worldwide general population [1], being the leading cause of disability around the world [2]. A significant decrease in life quality, functional impairment, and other psychosocial aspects, as well as comorbidities are associated with MDD, among others. What is more, a high degree of disability, morbidity, and mortality by suicide (suicidal ideation) causes MDD to be considered a major public health concern in developed countries [3].
Although tremendous efforts have been made in order to better understand and characterize this debilitating illness, current knowledge regarding MDD pathophysiology and neurobiology have failed to completely elucidate its molecular particularities to a greater extent. As a consequence, about 40% of patients with MDD do not respond to antidepressant treatment (AD) and eventually become treatment-resistant as the disease burden increases [4]. In addition, although being diagnosed at relatively early ages in a somewhat efficient fashion, the lack of uniform and accurate diagnostic tools (biomarkers) may lead to difficulties in assessing the differences between MDD and other etiologically related diseases, such as bipolar disorder (BD) [5]. Performing the Diagnosis and Statistical Manual of Mental Disorders (DSM-5) and the 11th Revision of the International Classification of Diseases (ICD 11) as the gold standard diagnostic criterion applied to patients was shown to induce interviewer bias, especially if performed by only one health specialist, which might lead to misdiagnosis in many cases [6]. Moderate reliability has been attributed to the Structured Clinical Interview for DSM-IV Axis I Disorders as well (SCID-I) [7,8,9].
To date, it is known that MDD patients suffer multiple alterations in different regions of the brain, compared to healthy subjects. Studies have shown that qualitatively, synaptic circuits and neural, functional, and structural plasticity are steadily impaired, while connectivity between different brain regions is disrupted. The latter affects communication between subcortical areas involved in modulating negative emotions, the frontal lobe with other brain regions, ultimately affecting cognition, memory, and learning [10,11,12]. Evidence reveals that MDD subjects present a smaller hippocampal volume, a modified morphology (number and shape) of dendrites, and the atrophy of neurons [13,14,15,16,17,18].
Thus, it is becoming increasingly clear that MDD arises primarily due to systematic alterations in the intimate biochemical and signaling pathways involved in mood, cognition, and disposition. Impaired and/or compromised cellular networks involved in MDD remain to be understood and are extensively being investigated in modern research, which has the ability to fine-tune the current approaches and the understanding of MDD neurobiology. However, mounting evidence suggests that no single mechanism can entirely account for and encompass the etiopathogenesis of MDD [11].
Recently, studies of microRNAs (miRs) have been given great attention for their potential role in the etiology and pathophysiology of mental disorders, such as MDD. miRs are small (18–25 nucleotides in length) endogenous noncoding RNA molecules, which post-transcriptionally regulate gene expression by complementarily binding to the 3’UTR regions of target messenger RNAs (mRNAs), ultimately leading either to mRNA degradation or translation inhibition [19,20]. Literature reports have shown that miRs regulate gene expression in a cohesive fashion, being virtually involved in all biological functions [21]. They have been observed to play key roles in physiology, as well as pathological conditions, including neuropsychiatric diseases, such as Alzheimer’s disease, schizophrenia, Parkinson’s disease, and MDD [22,23,24,25,26]. In MDD, miRs act at different levels in the process of neurogenesis and synaptic plasticity, by regulating specific genes that are critical components of various signaling pathways involved in MDD development [27,28]. Being ubiquitously found in numerous body fluids, such as blood, saliva, urine, tears, and cerebrospinal fluid, or even encapsulated in small vesicles (exosomes) [29], they have the great advantage of being easy to collect and analyze. In addition, miRs appear to have a chemically superior stability compared to other RNA-based molecules, such as mRNA, and their expression has been found to be dysregulated in MDD patients when compared to healthy controls. Therefore, taken together, the minimally invasive screening technique of circulating miRs in biological samples of MDD patients might represent a promising tool for the accurate and optimized diagnostic and treatment response monitoring of this neuropsychiatric illness [26,28,29,30,31].
Therefore, the aim of this review article is to critically and systematically review the differences in miR levels in MDD patients vs. controls, at baseline and before and after antidepressant treatment and their potential diagnostic and therapeutic relevance in the context of biomarker development, for a better characterization and reliable diagnosis of MDD, which could open novel horizons in the field of neuropsychiatry.

2. Materials and Methods

2.1. Search Strategy and Study Selection

All research articles included in this study were retrieved by two independent investigators by interrogating the PubMed, Web of Knowledge, and DirectScience databases (up to 20 of March 2021) with the following combination of key words: (“depression” or “depressive disorder”), and (“microRNA” or “miR”), and (“blood compartments”), and (“diagnosis”), and (“treatment” or “antidepressant treatment” or “antidepressant” or “therapy”), and (“biomarker”). The references from the articles of interest were analyzed to identify other relevant reports.
After the electronic search, duplicate references were excluded. Titles, abstracts, and study methodologies were screened based on the inclusion and exclusion criteria.

2.2. Inclusion and Exclusion Criteria

We selected studies that contained patients with diagnosed MDD and healthy controls. The index tests for candidate miRs from the selected specimen types (blood compartments) were performed by quantitative real-time PCR. Patients were diagnosed with MDD based on the Structured Clinical Interview for DSM-IV Axis I Disorders (SCID-I) (reference standard).
Research articles’ inclusion criteria were: (1) case-control studies in human subjects on depression assessing miRs’ expression level, with or without AD, (2) studies evaluating the diagnostic potential of different miRs in MDD, (3) MDD diagnosed based on the Structured Clinical Interview for DSM-IV Axis I Disorders (SCID-I), (4) a control group consisting of healthy subjects, and (5) published in the English language.
Research articles’ exclusion criteria were: (1) studies not conducted on human subjects, (2) studies assessing miR expression in body fluids other than blood, (3) nonoriginal papers, such as conference abstracts, letters to editors, and reviews, (4) duplicate studies, and (5) papers not written in the English language.
Disagreement between the two reviewers (D.N. and C.H.) was resolved by discussion. Our study protocol was not prospectively registered.

2.3. Data Collection and Characteristics of the Included Studies

We further considered for analysis only research articles that presented data related to the screening and validation of miRs in MDD from blood compartments (whole blood, serum, total plasma (TP), plasma exosomes, exosome-depleted plasma (EDP), and peripheral blood mononuclear cells (PBMCs)). Extracellular vesicle (EV)-entrapped miRs, such as in exosomes, have also been explored as sources of biomarkers for MDD.
From the initial search, 432 articles were found. After title and abstract screening, a total of 163 articles were included. After full-text reading, 137 studies were excluded, and 19 articles were included for the present paper. Figure 1 summarizes our study selection process.

3. Results

There is a high heterogeneity regarding the analyzed miRs in MDD, in peripheral blood compartments. The included studies revealed dysregulations in the expression levels of miRs in depressed patients compared to healthy controls, as well as before and after AD. Methodological and study design differences were observed among the studies, which can explain the heterogeneity of the analyzed miRs. Not all authors specified the analyzed blood compartment; the specified blood compartments included: whole blood, plasma, serum, and PBMCs.
Table 1 presents for each study the sample size (number of cases/controls), the blood compartment used for the analyses (some authors did not specify this), and miR findings and expression (upregulated, downregulated, or unchanged miRs) in depressive patients compared to healthy controls.
A total number of 106 miRs were identified in the aforementioned studies, out of which 49 were upregulated and 35 downregulated and 22 presented no changes in expression levels (MDD patients vs. healthy controls). The most frequently upregulated miRs were miR-107, miR-132, miR-182, and miR-124, while the most frequently downregulated were miR-381 and miR-451a. There were inconsistencies regarding miR-107, as some studies found it upregulated, others downregulated, or even unchanged in MDD patient samples vs. controls.
Table 2 shows the 49 upregulated miRs and the 35 downregulated miRs in patients with MDD (compared to controls).
Table 3 shows the sample characteristics for each study investigating miRs before and after AD in MDD patients, the blood compartment used for the analyses, and miR findings and expression level changes in depressive patients, before and after AD.
A total number of 88 different miRs were identified in relation to treatment response. There were 61 miRs upregulated and 26 downregulated in MDD patients as a response to AD. We found inconsistent reports regarding several miRs and one unchanged miR (miR-26b) after AD. The most prevalent upregulated miRs after AD treatment were let-7e, miR-183, and miR-335.
There is an increasing interest regarding miR changes towards treatment response. Several antidepressants exert their effects by targeting miRs. This suggests that miRs may be used as biomarkers for monitoring therapy response in patients with MDD. The treatment regimens included in the present studies were mostly SSRI (citalopram, paroxetine, sertralinum) and SNRI (venlafaxine, duloxetine). The average period of treatment was eight weeks. Some analyses were performed after four weeks of treatment, while in other studies, after ten weeks or twelve weeks.
Interestingly, the majority of miRs studies changed their expression pattern after AD treatment, but some maintained their expression level. This is the case of miR-494, -589, -26b, -34a-5p, -124, and -132, which remained upregulated even after treatment, while miR-451a remained downregulated after treatment.
Table 4 presents the common miRs found in MDD patient samples and their expression level before and after antidepressant administration.

4. Discussion

miRs can be detected in different body fluids and in various blood compartments, such as whole blood, plasma, serum, and PBMCs. They have been discovered to have a regulatory role, both in physiological activities and in the pathogenesis of certain diseases. Recent literature reports suggest that miRs may influence the etiology and pathophysiology of psychiatric disorders, including MDD [32].
Mounting evidence suggests that a tremendous number of miR species possess a dysregulated expression pattern in MDD patients relative to healthy controls. miR-132 was among the top-ranked upregulated miRs within the studies, with evidence demonstrating its direct involvement in the pathophysiology of MDD. Animal studies have shown that the increase in miR-132 expression negatively correlated with brain-derived neurotrophic factor (BDNF) expression and that inhibiting miR-132 leads to an increase in BDNF expression and to the reduction of depression symptoms. Moreover, a high miR-132 expression level leads to short-term memory and learning impairment [33].
On the other hand, the most frequently downregulated miRs in MDD patients vs. controls were miR-381 and miR-451a, which are not very well documented to date. It is known, however, that miR-451a downregulates the expression of the macrophage migration inhibitory factor in cells, but its exact mechanistic role in MDD development is still under investigation [38].
Another upregulated miR, miR-155, was also found among the included studies, and mechanistically, it was shown that it represses the expression of the silent information regulator 1 (SIRT1) gene, involved in the circadian rhythm and depression [46].
In addition, some miRs kept their expression levels constant even after administration of AD treatment (let-7e, miR-183, and miR-335); however, contradictory studies exist, and their exact role in MDD etiopathogenesis is yet to be understood [51,52].
Another miR that is well documented in the literature in relation to depression is miR-182. Animal studies performed on rats showed that suppressing miR-182 expression in the hippocampus exerts antidepressant-like effects, thus suggesting that this miR is directly involved in the development of MDD. Overexpression of miR-182 increased the symptoms of depression and decreased the BDNF levels [53]. miR-124 is also involved in the development of MDD and was found by us as one of the most frequently upregulated miRs in patient samples. Wang et al. (2017) showed that, in HEK 293 cell lines, miR-124 targeted the glucocorticoid receptor (GR). Moreover, miR-124 suppression leads to activation of BDNF and induced behavioral improvement of mice, thus suggesting a potential biomarker and therapeutic target for AD drug development, as also suggested by another review report [54,55].
Interestingly, other miRs have been shown to have an inconsistent dysregulation pattern among studies. For example, miR-16 and miR-135a were found to be either upregulated or downregulated, depending on the cohorts examined, the phenotype, and the tissue specimen. This outcome could also possibly occur due to variables that were not taken into account in the literature reports. Both miRs regulate the expression of the serotonin transporter (SERT). Therefore, independent validation of significantly dysregulated individual miRs in blood samples of MDD patients could aid in the biomarker development for the diagnostics of MDD, enriching current diagnostic strategies that remain only moderately reliable to date [9].
Furthermore, we observed that numerous miRs changed their expression pattern after AD. One explanation could be that some miRs are directly involved in the mechanism of action of SSRI and SNRI drugs. Of note, miR-16 targets the SERT and acts as a central effector in mediating the adaptive response to treatment with fluoxetine, of serotoninergic and noradrenergic neurons [56].
Taken together, these studies demonstrate that circulating miRs have a great diagnostic biomarker potential for MDD detection. Moreover, the discovery of altered miR levels between circulating samples of MDD patients compared to healthy controls could lead to a better understanding of the molecular mechanisms involved in the complex neurobiology of this debilitating disorder. Validation of independent miR candidates in individual cohort studies is another crucial step for biomarker development in order to increase the confidence of the results. In addition, as many miRs change their expression pattern after administration of AD, they represent promising candidates not only for treatment response monitoring indicators, but also for the discovery of unknown mechanisms of action.
However, our review has some limitations in assessing the use of miRs as biomarkers of MDD, as some studies are inconsistent and/or conflicting regarding miR expression levels. This heterogeneity could primarily arise due to technical and methodological differences regarding sample types and collection, processing, RNA extraction, and the choice for further downstream statistical analysis. In order to increase the specificity of potential diagnostic biomarkers of MDD, the use of different control groups would be required. However, the latter strategy might lead to difficulties in comparisons between different studies for review reports.

Author Contributions

Conceptualization, C.M. and C.H.; methodology, C.H.; software, C.H.; validation, C.M., C.H. and D.N.; formal analysis, C.M.; investigation, C.H.; resources, C.M.; data curation, C.M.; writing—original draft preparation, D.N. and C.H.; writing—review and editing, C.M., E.S. and V.E.; visualization, V.E.; supervision, C.M., E.S. and V.E.; project administration, C.M.; funding acquisition, C.M., E.S. and C.H. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by an internal grant of “Victor Babes” University of Medicine and Pharmacy Timisoara (Grant Number 4EXP/1232/2020).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data is available in the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Weissman, M.M.; Bland, R.C.; Canino, G.J.; Faravelli, C.; Greenwald, S.; Hwu, H.G.; Joyce, P.R.; Karam, E.G.; Lee, C.K.; Lellouch, J.; et al. Cross-national epidemiology of major depression and bipolar disorder. JAMA 1996, 276, 293–299. [Google Scholar] [CrossRef]
  2. Friedrich, M.J. Depression Is the Leading Cause of Disability Around the World. JAMA 2017, 317, 1517. [Google Scholar] [CrossRef]
  3. Kessler, R.C.; Berglund, P.; Demler, O.; Jin, R.; Merikangas, K.R. Lifetime prevalence and age-of-onset distributions of DSM-IV disorders in the national comorbidity survey replication. Arch. Gen. Psychiatry 2005, 62, 593–602. [Google Scholar] [CrossRef] [Green Version]
  4. Fava, M. Definition and epidemiology of treatment-resistant depression. Psychiatr. Clin. N. Am. 1996, 19, 179–200. [Google Scholar] [CrossRef]
  5. Hashimoto, K. Metabolomics of Major Depressive Disorder and Bipolar Disorder: Overview and Future Perspective. Adv. Clin. Chem. 2018, 84, 81–99. [Google Scholar]
  6. Singh, T. Misdiagnosis of bipolar disorder. Psychiatry (Edgmont) 2006, 3, 57–63. [Google Scholar] [PubMed]
  7. Zajecka, J.M. Treating depression to remission. J. Clin. Psychiatry 2003, 64, 7–12. [Google Scholar] [PubMed]
  8. Schmidt, H.D.; Shelton, R.C. Functional biomarkers of depression: Diagnosis, treatment, and pathophysiology. Neuropsychopharmacology 2011, 36, 2375–2394. [Google Scholar] [CrossRef] [PubMed]
  9. Takahashi, S.; Kitamura, T.; Okano, T.; Tomita, T.; Kikuchi, A. Structured Clinical Interview for DSM-IV Axis I Disorders Version 2.0; Nippon Hyouron Sha: Tokyo, Japan, 2003. [Google Scholar]
  10. Andreasen, N.C. Linking mind and brain in the study of mental illnesses: A project for a scientific psychopathology. Science 1997, 275, 1586–1593. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Honer, W.G. Assessing the machinery of mind: Synapses in neuropsychiatric disorders. J. Psychiatry Neurosci. 1999, 24, 116–121. [Google Scholar] [PubMed]
  12. Anand, A.; Li, Y.; Wang, Y.; Wu, J.W.; Gao, S.J.; Bukhari, L.; Mathews, V.P.; Kalnin, A.; Lowe, M.J. Activity and connectivity of brain mood regulating circuit in depression: A functional magnetic resonance study. Biol. Psychiatry 2005, 57, 1079–1088. [Google Scholar] [CrossRef]
  13. Toni, N.; Buchs, P.A.; Nikonenko, I.; Bron, C.R.; Muller, D. LTP promotes formation of multiple spine synapses between a single axon terminal and a dendrite. Nature 1999, 402, 421–425. [Google Scholar] [CrossRef]
  14. Hajsza, T.; MacLusky, N.J.; Leranth, C. Short-term treatment with the antidepressant fluoxetine triggers pyramidal dendritic spine synapse formation in rat hippocampus. Eur. J. Neurosci. 2005, 21, 1299–1303. [Google Scholar] [CrossRef]
  15. McEwen, B.S. Effects of adverse experiences for brain structure and function. Biol. Psychiatry 2000, 48, 721–731. [Google Scholar] [CrossRef]
  16. Sheline, Y.I. 3D MRI studies of neuroanatomic changes in unipolar major depression: The role of stress and medical comorbidity. Biol. Psychiatry 2000, 48, 791–800. [Google Scholar] [CrossRef]
  17. Sala, M.; Perez, J.; Soloff, P.; Ucelli di Nemi, S.; Caverzasi, E.; Soaresd, J.C.; Brambillab, P. Stress and hippocampal abnormalities in psychiatric disorders. Eur. Neuropsychopharmacol. 2004, 14, 393–405. [Google Scholar] [CrossRef] [PubMed]
  18. Rajkowska, G.; Miguel-Hidalgo, J.J. Gliogenesis and glial pathology in depression. CNS Neurol. Disord.-Drug Targets 2007, 6, 219–233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Bushati, N.; Cohen, S.M. microRNA functions. Annu. Rev. Cell Dev. Biol. 2007, 23, 175–205. [Google Scholar] [CrossRef] [PubMed]
  20. Bartel, D.P. MicroRNAs: Target recognition and regulatory functions. Cell 2009, 136, 215–233. [Google Scholar] [CrossRef] [Green Version]
  21. Malphettes, L.; Fussenegger, M. Impact of RNA interference on gene networks. Metab. Eng. 2006, 8, 672–683. [Google Scholar] [CrossRef]
  22. Li, M.; Marin-Muller, C.; Bharadwaj, U.; Chow, K.W.; Yao, Q.Z.; Chen, C.Y. MicroRNAs: Control and loss of control in human physiology and disease. World J. Surg. 2009, 33, 667–684. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Wang, M.; Qin, L.; Tang, B. MicroRNAs in Alzheimer’s Disease. Front. Genet. 2019, 10, 153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. He, K.; Guo, C.; He, L.; Shi, Y. MiRNAs of peripheral blood as the biomarker of schizophrenia. Hereditas 2017, 155, 9. [Google Scholar] [CrossRef]
  25. Goh, S.Y.; Chao, Y.X.; Dheen, S.T.; Tan, E.K.; Tay, S.S.W. Role of MicroRNAs in Parkinson’s Disease. Int. J. Mol. Sci. 2019, 20, 5649. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Dwivedi, Y. Emerging role of microRNAs in major depressive disorder: Diagnosis and therapeutic implications. Dialogues Clin. Neurosci. 2014, 16, 43–61. [Google Scholar]
  27. Serafini, G.; Pompili, M.; Innamorati, M.; Giordano, G.; Montebovi, F.; Sher, L.; Dwivedi, Y.; Girardi, P. The role of microRNAs in synaptic plasticity, major affective disorders and suicidal behavior. Neurosci. Res. 2012, 73, 179–190. [Google Scholar] [CrossRef] [PubMed]
  28. Saavedra, K.; Molina-Márquez, A.M.; Saavedra, N.; Zambrano, T.; Salazar, L.A. Epigenetic modifications of major depressive disorder. Int. J. Mol. Sci. 2016, 17, 1279. [Google Scholar] [CrossRef] [PubMed]
  29. Lu, T.X.; Rothenberg, M.E. MicroRNA. J. Allergy Clin. Immunol. 2018, 141, 1202–1207. [Google Scholar] [CrossRef] [Green Version]
  30. Enatescu, V.R.; Papava, I.; Enatescu, I.; Antonescu, M.; Anghel, A.; Seclaman, E.; Sirbu, I.O.; Marian, C. Circulating Plasma Miro RNAs in Patients with Major Deppresive Disorder Treated with Antidepressants: A Pilot Study. Psychiatry Investig. 2016, 13, 549–557. [Google Scholar] [CrossRef] [Green Version]
  31. Dwivedi, Y. microRNAs as Biomarker in Depression Pathogenesis. Ann. Psychiatry Ment. Health 2013, 1, 1003. [Google Scholar]
  32. Belzeaux, R.; Bergon, A.; Jeanjean, V.; Loriod, B.; Formisano-Tréziny, C.; Verrier, L.; Loundou, A.; Baumstarck-Barrau, K.; Boyer, L.; Gall, V.; et al. Responder and nonresponder patients exhibit different peripheral transcriptional signatures during major depressive episode. Transl. Psychiatry 2012, 2, e185. [Google Scholar] [CrossRef]
  33. Li, Y.J.; Xu, M.; Gao, Z.H.; Wang, Y.Q.; Yue, Z.; Zhang, Y.X.; Li, X.X.; Zhang, C.; Xie, S.Y.; Wang, P.Y. Alterations of serum levels of BDNF-related miRNAs in patients with depression. PLoS ONE 2013, 8, e63648. [Google Scholar] [CrossRef] [Green Version]
  34. Fan, H.M.; Sun, X.Y.; Guo, W.; Zhong, A.F.; Niu, W.; Zhao, L.; Dai, Y.H.; Guo, Z.M.; Zhang, L.Y.; Lu, J. Differential expression of microRNA in peripheral blood mononuclear cells as specific biomarker for major depressive disorder patients. J. Psychiatr. Res. 2014, 59, 45–52. [Google Scholar] [CrossRef]
  35. Li, J.; Meng, H.; Cao, W.; Qiu, T. MiR-335 is involved in major depression disorder and antidepressant treatment through targeting GRM4. Neurosci. Lett. 2015, 606, 167–172. [Google Scholar] [CrossRef]
  36. Camkurt, M.A.; Acar, S.; Coskun, S.; Güneş, M.; Güneş, S.; Yılmaz, M.F.; Görür, A.; Tamer, L. Comparison of plasma MicroRNA levels in drug naive, first episode depressed patients and healthy controls. J. Psychiatr. Res. 2015, 69, 67–71. [Google Scholar] [CrossRef] [PubMed]
  37. Wan, Y.; Liu, Y.; Wang, X.; Wu, J.L.; Liu, K.Z.; Zhou, J.; Liu, L.; Zhang, C.H. Identification of differential microRNAs in cerebrospinal fluid and serum of patients with major depressive disorder. PLoS ONE 2015, 10, e0121975. [Google Scholar] [CrossRef] [PubMed]
  38. Wang, X.; Sundquist, K.; Hedelius, A.; Palmér, K.; Memon, A.A.; Sundquist, J. Circulating microRNA-144-5p is associated with depressive disorders. Clinical. Epigenet. 2015, 7, 69. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Maffioletti, E.; Cattaneo, A.; Rosso, G.; Maina, G.; Maj, C.; Gennarellia, 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]
  40. Sun, N.; Lei, L.; Wang, Y.; Yang, C.X.; Liu, Z.F.; Li, X.R.; Zhang, K. Preliminary comparison of plasma notch-associated microRNA-34b and -34c levels in drug naive, first episode depressed patients and healthy controls. J. Affect. Disord. 2016, 194, 109–114. [Google Scholar] [CrossRef]
  41. He, S.; Liu, X.; Jiang, K.; Peng, D.H.; Hong, W.; Fang, Y.R.; Qian, Y.P.; Yu, S.Y.; Li, H.F. Alterations of microRNA-124 expression in peripheral blood mononuclear cells in pre- and post-treatment patients with major depressive disorder. J. Psychiatr. Res. 2016, 78, 65–71. [Google Scholar] [CrossRef]
  42. Roy, B.; Dunbar, M.; Shelton, R.; Dwivedi, Y. Identification of MicroRNA-124-3p as a Putative Epigenetic Signature of Major Depressive Disorder. Neuropsychopharmacol 2017, 42, 864–875. [Google Scholar] [CrossRef] [Green Version]
  43. Kuang, W.H.; Dong, Z.Q.; Tian, L.T.; Li, J. MicroRNA-451a, microRNA-34a-5p, and microRNA-221-3p as predictors of response to antidepressant treatment. Braz. J. Med. Biol. Res. 2018, 51, e7212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Fang, Y.; Qiu, Q.; Zhang, S.; Sun, L.; Li, G.J.; Xiao, S.F.; Lia, X. Changes in miRNA-132 and miR-124 levels in non-treated and citalopram-treated patients with depression. J. Affect. Disord. 2018, 227, 745–751. [Google Scholar] [CrossRef] [PubMed]
  45. Gheysarzadeh, A.; Sadeghifard, N.; Afraidooni, L.; Pooyan, F.; Mofid, M.R.; Valadbeigi, H.; Bakhtiari, H.; Keikhavani, S. Serum-based microRNA biomarkers for major depression: MiR-16, miR-135a, and miR-1202. J. Res. Med. Sci. 2018, 23, 69. [Google Scholar] [CrossRef] [PubMed]
  46. Hung, Y.Y.; Wu, M.K.; Tsai, M.C.; Huang, Y.L.; Kang, H.Y. Aberrant Expression of Intracellular let-7e, miR-146a, and miR-155 Correlates with Severity of Depression in Patients with Major Depressive Disorder and Is Ameliorated after Antidepressant Treatment. Cells 2019, 8, 647. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Bocchio-Chiavetto, L.; Maffioletti, E.; Bettinsoli, P.; Giovannini, C.; Bignotti, S.; Tardito, D.; Corrada, D.; Milanesi, L.; Gennarellie, M. Blood microRNA changes in depressed patients during antidepressant treatment. Eur. Neuropsychopharmacol. 2013, 23, 602–611. [Google Scholar] [CrossRef]
  48. Zhang, Q.L.; Lu, J.; Sun, X.Y.; Guo, W.; Zhao, L.; Fan, H.M.; Zhong, A.F.; Niu, W.; Dai, Y.H.; Zhang, L.Y.; et al. A preliminary analysis of association between plasma microRNA expression alteration and symptomatology improvement in Major Depressive Disorder (MDD) patients before and after antidepressant treatment. Eur. J. Psychiatry 2014, 28, 252–264. [Google Scholar]
  49. Lopez, J.P.; Fiori, L.M.; Cruceanu, C.; Lin, R.; Labonte, B.; Cates, H.M.; Heller, A.E.; Vialou, V.; Ku, S.M.; Gerald, C.; et al. MicroRNAs 146a/b-5 and 425-3p and 24-3p are markers of antidepressant response and regulate MAPK/Wnt-system genes. Nat. Commun. 2017, 8, 15497. [Google Scholar] [CrossRef] [Green Version]
  50. Lin, C.C.; Tsai, M.C.; Lee, C.T.; Sun, M.H.; Huang, T.L. Antidepressant treatment increased serum miR-183 and miR-212 levels in patients with major depressive disorder. Psychiatry Res. 2018, 270, 232–237. [Google Scholar] [CrossRef]
  51. Scott, H.L.; Tamagnini, F.; Narduzzo, K.E.; Howarth, J.L.; Lee, Y.B.; Wong, L.F.; Brown, M.W.; Warburton, E.C.; Bashir, Z.I.; Uney, J.B. MicroRNA-132 regulates recognition memory and synaptic plasticity in the perirhinal cortex. Eur. J. Neurosci. 2012, 36, 2941–2948. [Google Scholar] [CrossRef] [Green Version]
  52. Griffiths, S.; Scott, H.; Glover, C.; Bienemann, A.; Ghorbel, M.T.; Uney, J.; Brown, M.W.; Warburton, E.C.; Bashir, Z.I. Expression of long-term depression underlies visual recognition memory. Neuron 2008, 58, 186–194. [Google Scholar] [CrossRef] [Green Version]
  53. Li, Y.; Li, S.; Yan, J.; Wang, D.Q.; Yin, R.G.; Zhao, L.; Zhu, Y.; Zhu, X.L. miR-182 (microRNA-182) suppression in the hippocampus evokes antidepressant-like effects in rats. Prog. Neuropsychopharmacol. Biol. Psychiatry 2016, 65, 96–103. [Google Scholar] [CrossRef] [PubMed]
  54. Wang, S.S.; Mu, R.H.; Li, C.F.; Dong, S.Q.; Geng, D.; Liu, Q.; Yi, L.T. microRNA-124 targets glucocorticoid receptor and is involved in depression-like behaviors. Prog. Neuropsychopharmacol. Biol. Psychiatry 2017, 79, 417–425. [Google Scholar] [CrossRef] [PubMed]
  55. Dwivedi, Y. microRNA-124: A putative therapeutic target and biomarker for major depression. Expert Opin. Ther. Targets 2017, 21, 653–656. [Google Scholar] [CrossRef] [Green Version]
  56. Launay, J.M.; Mouillet-Richard, S.; Baudry, A.; Pietri, M.; Kellermann, O. Raphe-mediated signals control the hippocampal response to SRI antidepressants via miR-16. Transl. Psychiatry 2011, 1, e56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Flow diagram of the study selection process.
Figure 1. Flow diagram of the study selection process.
Life 11 01073 g001
Table 1. miRs’ expression in different blood compartments of patients with MDD compared to healthy controls.
Table 1. miRs’ expression in different blood compartments of patients with MDD compared to healthy controls.
Study (Year, Reference No)PatientsControlsBlood CompartmentUpregulated miRsDownregulated miRsUnchanged miRsTotal
Belzeaux et al., 2012 [32]1613PBMCsmiR-107
miR-133a
miR-148a
miR-425-3p
miR-494
miR-579
miR-652
miR-941
miR-589
miR-200c
miR-381
miR-571
miR-636
miR-1243
-9 upregulated, 5 downregulated
Li YJ et al., 2013 [33]4040SerummiR-132
miR-182
--2 upregulated
Fan et al., 2014 [34]8146PBMCsmiR-26b
miR-1972
miR-4485
miR-4498
miR-4743
--5 upregulated
Li J et al., 2015 [35]1818Whole bloodmiR-644
miR-450b
miR-328
miR-182
miR-335
miR-583
miR-708a
miR-650
miR-654a
miR-541
miR-663
miR-578
4 upregulated, 5 downregulated, 3 unchanged
Camkurt et al., 2015 [36]5041PlasmamiR-451a
miR-17-5p
miR-223-3p
miR-320amiR-25-3p
miR-126-3p
miR-16-5p
miR-93-5p
3 upregulated, 1 downregulated, 4 unchanged
Wan et al., 2015 [37]3827Serumlet-7d-3p
miR-34a-5p
miR-221-3p
miR-125a-5p
miR-30a-5p
miR-29b-3p
miR-10a-5p
miR-375
miR-155–5p
miR-33a-5p
miR-139–5p
miR-451a
miR-15b-5p
miR-106-5p
miR-590-5p
miR-185-5p
-11 upregulated, 5 downregulated
Wang X et al., 2015 [38]16952Plasma-miR-144-5p-1 downregulated
Mafioletti et al., 2016 [39]2020Peripheral venous bloodhsa-miR-199a-5p
hsa-miR-24-3p
hsa-miR-425-3p
hsa-miR-29c-5p
hsa-miR-330-3p
hsamiR-345-5p
hsa-let-7a-5p
hsa-let-7d-5p
has-let-7f-5p
has-miR-1915-3p
hsa-miR-720
hsa-miR-140-3p
hsa-miR-1973
hsa-miR-30d-5p
hsa-miR-3158-3p
hsa-miR-330-5p
hsa-miR-378a-5p
hsa-miR-1915-5p
hsa-miR-1972
hsa-miR-21-3p
hsa-miR-4521
hsa-miR-4793-3p
hsa-miR-4440
6 upregulated, 4 downregulated, 13 unchanged
Sun et al., 2016 [40]3232Peripheral blood leukocytesmiR-34b-5p
miR-34c-5p
-miR-369–3p
miR-381
miR-107
2 upregulated, 3 unchanged
He et al., 2016 [41]3230PBMCsmiR-124--1 upregulated
Roy et al., 2017 [42]1817SerummiR-124-3p--1 upregulated
Kuang et al., 2018 [43]8478SerummiR-34a-5p
miR-221-3p
miR-451a-2 upregulated, 1 downregulated
Fang Y et al., 2018 [44]4532PlasmamiR-124
miR-132
--2 upregulated
Gheysarzadeh et al., 2018 [45]3936Serum-miR-16
miR-135a
miR-1202
-3 downregulated
Hung et al., 2019 [46]8443PBMCsmiR-21-5p
miR-145
miR-223
miR-146a
miR-155
let-7e
-3 upregulated, 3 downregulated
Table 2. Upregulated and downregulated miRs in MDD patients’ vs. healthy controls from all studies included in this review (miR names in bold were identified by more than one independent study).
Table 2. Upregulated and downregulated miRs in MDD patients’ vs. healthy controls from all studies included in this review (miR names in bold were identified by more than one independent study).
Upregulated miRsDownregulated miRs
miR-107miR-200c
miR-133amiR-381
miR-148amiR-571
miR-425-3pmiR-636
miR-494miR-1243
miR-579hsa-let-7f-5p
miR-652hsa-miR-1285-5p
miR-941hsa-miR-107
miR-589hsa-miR-26a-5p
hsa-miR-5010-3phsa-miR-26b-5p
hsa-miR-151a-3pbrain-miR-161
miR-132brain-miR-112
miR-182hsa-let-7d-3p
miR-26bhsa-miR-103a-3p
miR-1972hsa-miR-532-5p
miR-4485miR-335
miR-4498miR-583
miR-4743miR-708a
miR-644miR-650
miR-450bmiR-320a
miR-328miR-15b-5p
miR-451amiR-106-5p
miR-17-5pmiR-590-5p
let-7d-3pmiR-185-5p
miR-223-3pmiR-144-5p
miR-34a-5phsa-let-7a-5p
miR-221-3phsa-let-7d-5p
miR-125a-5phas-let-7f-5p
miR-30a-5phas-miR-1915-3p
miR-29b-3pmiR-451a
hsa-miR-199a-5pmiR-16
hsa-miR-24-3pmiR-135a
hsa-miR-425-3pmiR-1202
hsa-miR-29c-5pmiR-146a
hsa-miR-330-3pmiR-155
miR-10a-5plet-7e
miR-375
hsamiR-345-5p
miR-155–5p
miR-33a-5p
miR-139–5p
miR-34b-5p
miR-34c-5p
miR-124
miR-124-3p
miR-34a-5p
miR-221-3p
miR-21-5p
miR-145
miR-223
Table 3. miR changes in expression levels before and after antidepressant (AD) treatment.
Table 3. miR changes in expression levels before and after antidepressant (AD) treatment.
StudyPatientsAD Treatment and DurationBlood CompartmentUpregulated miRsDownregulated miRUnchanged miRsTotal
Enatescu et al., 2016 [30]5Escitalopram 12 weeksPlasmamiR-1193
miR-3173-3p
miR-3154
miR-129-5p
miR-3661
miR-1287
miR-532-3p
miR-2278
miR-3150a-3p
miR-3909
miR-937
miR-676
miR-489
miR-637
miR-608
miR-4263
miR-382
miR-3691-5p
miR-375
miR-433
miR-1298
miR-1909
miR-1471
miR-99b
miR-151-5p
miR-223
miR-181b
miR-26a
miR-744
miR-301b
miR-27a
miR-24
miR-146a-
miR-146b-5p
miR-126
miR-151-3p
let-7d
miR-221
miR-125a-5p
miR-652
-23 upregulated, 17 downregulated
Li J et al., 2015 [35]18Citalopram, 1 weekWhole bloodmiR-335--1 upregulated
Wang X et al., 2015 [38]169Not mentioned, 8 weeksPlasmamiR-144-5p
miR-30a-5p
--2 upregulated
He et al., 2016 [41]32Venlafaxine (N = 7), paroxetine (N = 7), fluoxetine (N = 3), escitalopram (N = 11), duloxetine (N = 1), sertraline (N = 3), mirtazapine (N = 2)PBMCs-miR-124-1 downregulated
Kuang et al., 2018 [43]84Paroxetine 8 weeksSerummiR-34a-5p
miR-221a-3p
miR-451a-2 upregulated, 1 downregulated
Fang Y et al., 2018 [44]32Citalopram 8 weeksPlasmamiR-124miR-132-1 upregulated, 1 downregulated
Hung YY et al., 2019 [46]84Not mentioned, 4 weeksPBMCsmiR-146a
miR-155
let-7e
--3 upregulated
Bocchio-Chiavetto et al., 2013 [47]10Escitalopram 10 weeksWhole bloodmiR-130b
miR-505
miR-29b-2
miR-26b
miR-22
miR-26a
miR-64
miR-494
let-7d
let-7g
let-7e
let-7f
miR-629
miR-106b
miR-103
miR-191
miR-128
miR-502-3p
miR-374b
miR-132
miR-30d
miR-500
miR-589
miR-183
miR-574-3p
miR-140-3p
miR-335
miR-361-5p
miR-34c-5p
miR-770-5p
-26 upregulated, 2 downregulated
Zhang et al., 2014 [48]20Venlafaxine, sertraline, mirtazapine 6 weeksPBMCs-miR-1972
miR-4485
miR-4498
miR-4743
miR-26b4 upregulated, 1 downregulated
Lopez et al., 2017 [49]23Escitalopram 8 weeksPeripheral bloodmiR-1202--1 upregulated
Lin CC et al., 2018 [50]33Not mentioned, 4 weeksWhole bloodmiR-16
miR-183
miR-212
--3 upregulated
Table 4. Expression level of common miRs before and after AD treatment.
Table 4. Expression level of common miRs before and after AD treatment.
Common miRs in MDD (No Treatment and After ADT)miRs of MDD Patients Before Antidepressant Treatment vs. ControlsmiRs of MDD Patients After Antidepressant Treatment vs. Controls
let-7dup, downup, down
let-7edownup
let-7fdownup
miR-16downup
miR-24updown
miR-26adownup, down
miR-26bupup
miR-29bupup
miR-30a-5pupup
miR-34a-5pupup
miR-34c-5pupdown
miR-106bdownup
miR-124upup, down
miR-125a-5pupdown
miR-132upup, down
miR-144-5pdownup
miR-146a-3pdownup, down
miR-151-3pupdown
miR-155up, downup, down
miR-221upup, down
miR-223updown
miR-335downup
miR-375upup
miR-425-3pupdown
miR-451aup, downdown
miR-494upup
miR-532downup
miR-589upup
miR-652updown
miR-1202downup
miR-1972downdown
miR-4485updown
miR-4498updown
miR-4743updown
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Homorogan, C.; Nitusca, D.; Seclaman, E.; Enatescu, V.; Marian, C. Uncovering the Roles of MicroRNAs in Major Depressive Disorder: From Candidate Diagnostic Biomarkers to Treatment Response Indicators. Life 2021, 11, 1073. https://doi.org/10.3390/life11101073

AMA Style

Homorogan C, Nitusca D, Seclaman E, Enatescu V, Marian C. Uncovering the Roles of MicroRNAs in Major Depressive Disorder: From Candidate Diagnostic Biomarkers to Treatment Response Indicators. Life. 2021; 11(10):1073. https://doi.org/10.3390/life11101073

Chicago/Turabian Style

Homorogan, Claudia, Diana Nitusca, Edward Seclaman, Virgil Enatescu, and Catalin Marian. 2021. "Uncovering the Roles of MicroRNAs in Major Depressive Disorder: From Candidate Diagnostic Biomarkers to Treatment Response Indicators" Life 11, no. 10: 1073. https://doi.org/10.3390/life11101073

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop