MicroRNA expression profiles and type 1 diabetes mellitus: systematic review and bioinformatic analysis

in Endocrine Connections
Authors:
Taís S Assmann Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
Postgraduation Program in Medical Sciences: Endocrinology, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil

Search for other papers by Taís S Assmann in
Current site
Google Scholar
PubMed
Close
,
Mariana Recamonde-Mendoza Institute of Informatics, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil

Search for other papers by Mariana Recamonde-Mendoza in
Current site
Google Scholar
PubMed
Close
,
Bianca M De Souza Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
Postgraduation Program in Medical Sciences: Endocrinology, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil

Search for other papers by Bianca M De Souza in
Current site
Google Scholar
PubMed
Close
, and
Daisy Crispim Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
Postgraduation Program in Medical Sciences: Endocrinology, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil

Search for other papers by Daisy Crispim in
Current site
Google Scholar
PubMed
Close

Open access

Sign up for journal news

Growing evidence indicates that microRNAs (miRNAs) have a key role in processes involved in type 1 diabetes mellitus (T1DM) pathogenesis, including immune system functions and beta-cell metabolism and death. Although dysregulated miRNA profiles have been identified in T1DM patients, results are inconclusive; with only few miRNAs being consistently dysregulated among studies. Thus, we performed a systematic review of the literature on the subject, followed by bioinformatic analysis, to point out which miRNAs are dysregulated in T1DM-related tissues and in which pathways they act. PubMed and EMBASE were searched to identify all studies that compared miRNA expressions between T1DM patients and non-diabetic controls. Search was completed in August, 2017. Those miRNAs consistently dysregulated in T1DM-related tissues were submitted to bioinformatic analysis, using six databases of miRNA–target gene interactions to retrieve their putative targets and identify potentially affected pathways under their regulation. Thirty-three studies were included in the systematic review: 19 of them reported miRNA expressions in human samples, 13 in murine models and one in both human and murine samples. Among 278 dysregulated miRNAs reported in these studies, 25.9% were reported in at least 2 studies; however, only 48 of them were analyzed in tissues directly related to T1DM pathogenesis (serum/plasma, pancreas and peripheral blood mononuclear cells (PBMCs)). Regarding circulating miRNAs, 11 were consistently dysregulated in T1DM patients compared to controls: miR-21-5p, miR-24-3p, miR-100-5p, miR-146a-5p, miR-148a-3p, miR-150-5p, miR-181a-5p, miR-210-5p, miR-342-3p, miR-375 and miR-1275. The bioinformatic analysis retrieved a total of 5867 validated and 2979 predicted miRNA–target interactions for human miRNAs. In functional enrichment analysis of miRNA target genes, 77 KEGG terms were enriched for more than one miRNA. These miRNAs are involved in pathways related to immune system function, cell survival, cell proliferation and insulin biosynthesis and secretion. In conclusion, eleven circulating miRNAs seem to be dysregulated in T1DM patients in different studies, being potential circulating biomarkers of this disease.

Abstract

Growing evidence indicates that microRNAs (miRNAs) have a key role in processes involved in type 1 diabetes mellitus (T1DM) pathogenesis, including immune system functions and beta-cell metabolism and death. Although dysregulated miRNA profiles have been identified in T1DM patients, results are inconclusive; with only few miRNAs being consistently dysregulated among studies. Thus, we performed a systematic review of the literature on the subject, followed by bioinformatic analysis, to point out which miRNAs are dysregulated in T1DM-related tissues and in which pathways they act. PubMed and EMBASE were searched to identify all studies that compared miRNA expressions between T1DM patients and non-diabetic controls. Search was completed in August, 2017. Those miRNAs consistently dysregulated in T1DM-related tissues were submitted to bioinformatic analysis, using six databases of miRNA–target gene interactions to retrieve their putative targets and identify potentially affected pathways under their regulation. Thirty-three studies were included in the systematic review: 19 of them reported miRNA expressions in human samples, 13 in murine models and one in both human and murine samples. Among 278 dysregulated miRNAs reported in these studies, 25.9% were reported in at least 2 studies; however, only 48 of them were analyzed in tissues directly related to T1DM pathogenesis (serum/plasma, pancreas and peripheral blood mononuclear cells (PBMCs)). Regarding circulating miRNAs, 11 were consistently dysregulated in T1DM patients compared to controls: miR-21-5p, miR-24-3p, miR-100-5p, miR-146a-5p, miR-148a-3p, miR-150-5p, miR-181a-5p, miR-210-5p, miR-342-3p, miR-375 and miR-1275. The bioinformatic analysis retrieved a total of 5867 validated and 2979 predicted miRNA–target interactions for human miRNAs. In functional enrichment analysis of miRNA target genes, 77 KEGG terms were enriched for more than one miRNA. These miRNAs are involved in pathways related to immune system function, cell survival, cell proliferation and insulin biosynthesis and secretion. In conclusion, eleven circulating miRNAs seem to be dysregulated in T1DM patients in different studies, being potential circulating biomarkers of this disease.

Introduction

Type 1 diabetes mellitus (T1DM) is characterized by autoimmune destruction of pancreatic beta-cells by T lymphocytes and macrophages (1). The disease is usually diagnosed when over 80–90% of beta-cells have been destructed by the infiltrating immune system. T1DM development is slow, providing a potentially long window of time in which it is possible to identify and theoretically treat individuals at risk (2, 3).

The first sign of autoimmunity against beta-cells, frequently detectable a few months/years before the appearance of clinical symptoms, is the occurrence of antibodies against beta-cell antigens (4). These autoantibodies are used as biomarkers of T1DM risk and are directed against insulin, glutamic acid decarboxylase, zinc cation efflux transporter and tyrosine phosphatases-2 and -2β (4). The presence of more than two of these autoantibodies indicates high risk for T1DM development (5, 6). However, the use of islet autoantibodies as biomarkers of T1DM progression has some limitations, especially because a subset of children with new-onset T1DM is negative for islet autoantibodies (6), and many autoantibody-positive subjects will never develop T1DM (2, 7). Moreover, autoantibodies cannot be used as markers to initiate a potential treatment at earlier stages of the disease when many beta-cells are still present (2, 7). Thus, new biomarkers of T1DM are necessary to complement the information obtained from the presence of autoantibodies together with genetic and environmental risk factors (8).

In this context, several microRNAs (miRNAs) are released in the circulation and might be used as biomarkers to evaluate health status and disease progression (2). miRNAs are a class of small noncoding RNAs that negatively regulate gene expression by partially pairing to the 3′, 5′ untranslated regions of their target mRNAs, leading to translation repression and/or transcript degradation (9, 10, 11). They have recognized roles in the regulation of various processes, such as cellular differentiation, proliferation, metabolism, aging and apoptosis (10, 12). miRNAs are estimated to regulate the expression of more than 60% of protein-coding genes (9); consequently, changes in their expressions have been linked to many diseases, including cancer, endocrine disorders and autoimmune diseases (13, 14, 15).

Growing evidence suggests that miRNAs also play a key role in immune system functions as well as in beta-cell metabolism, proliferation and death, which are processes involved in T1DM pathogenesis (2, 10, 16, 17). Indeed, IL-1β and TNF inflammatory cytokines were reported to induce miR-21-5p, miR-30b-3p, miR-34, miR-101a and miR-146a-5p expressions in MIN6 cells and human pancreatic islets (18, 19), suggesting that these miRNAs may have a role in cytokine-mediated beta-cell destruction. miRNA-specific profiles were observed in PBMCs or serum from T1DM patients (20, 21, 22, 23, 24), and some miRNAs seem to modulate mRNA expressions of the major T1DM autoantigens (24, 25).

Several studies identified a large number of miRNAs as being differentially expressed in T1DM samples (2, 10). These studies were performed in cultured cells, body fluids or solid tissue samples from T1DM patients or murine models of the disease, using different techniques to quantify gene expression. Consequently, findings are inconsistent among studies; with only few miRNAs actually being important signatures of T1DM. Therefore, to further investigate which miRNAs may be used as new potential biomarkers of T1DM, we performed a systematic review of the literature on the subject. Additionally, bioinformatic analyses were performed to investigate the regulatory and functional roles of miRNAs in T1DM. For this, six databases of miRNA–target gene interactions were queried, including experimentally validated and computationally predicted miRNA–target gene interactions. The functional enrichment analysis of miRNAs target genes was performed using pathways annotation from the KEGG Pathway Database.

Methods

Search strategies and eligibility of relevant studies

This systematic literature search was designed and described in accordance with current guidelines (26, 27). PubMed and EMBASE repositories were searched to identify all studies that evaluated miRNA expressions in T1DM samples. The following medical subject headings (MeSH) were used: (‘diabetes mellitus’ OR ‘type 1, diabetes mellitus’) AND (‘microRNA’ OR ‘RNA, small untranslated’). The search was restricted to English, Portuguese or Spanish language papers and was completed on August, 2017. We also manually checked the reference lists of all articles retrieved to identify other important citations. To ensure that relevant studies were not overlooked, searches in Gene Expression Omnibus (www.ncbi.nlm.nih.gov/geo) and Array Express (www.ebi.ac.uk/arrayexpress) databases were also performed.

We included original reports that analyzed miRNA expressions in T1DM patients (cases) and non-diabetic subjects (controls) or in murine model of this disease. Studies that did not have a control group or studies performed in cell lines were excluded. Two investigators (T S A and B M S) independently reviewed titles and abstracts of articles retrieved in order to evaluate whether the studies were eligible for inclusion in this review.

Data extraction and quality assessment of each individual study

Data were independently extracted by two investigators (B M S and T S A) using a standardized abstraction form (26), and consensus was sought in all extracted items. Information extracted from each study in humans were as follows: (1) characteristics of studies and samples; (2) information regarding miRNA expression (method used for quantification, tissue analyzed, number of miRNAs analyzed) and (3) miRNA expression in groups. For those studies performed in mice/rats, we also collected information about the murine model analyzed. All miRNA names were standardized based on miRBase v21 prior to analysis.

Two investigators (T S A and B M S) assessed the quality of each eligible study using The Quality Assessment of Diagnostic Accuracy Studies-2 (QUADAS-2) (28). This tool comprises 4 key domains (patient selection, index test, reference standard and flow/timing) supported by 7 questions to aid judgment on risk of bias, rating risk of bias and concerns about applicability of studies. Each question can be answered with ‘yes’, ‘no’ or ‘unclear’. Then, a score of 1 is given for each ‘yes’ (low risk/high concern), a score of 0.5 for each ‘unclear’ and a score of 0 for each ‘no’ (high risk/low concern). Quality scores range from 0 to 7, with studies being classified as having ‘good quality’ (scores 6–7), ‘fair’ (scores 4–5) and ‘poor quality’ (scores <3).

Additionally, we checked if the articles were performed in accordance to Minimum Information about a Microarray Experiment (MIAME) guideline, version 2.0 (29) or Minimum Information for Publication of Quantitative Real-time PCR Experiments (MIQE) guideline (30). Only articles in accordance with these guidelines were included in the systematic review.

Bioinformatic prediction and analysis of miRNA’s target genes

To investigate in greater depth the functional involvement of miRNAs in T1DM, we selected those miRNAs that were consistently dysregulated in T1DM-related tissues (PBMCs, serum/plasma and pancreas) and performed bioinformatic analysis to retrieve their putative targets and identify potentially affected biological pathways under their regulation (Supplementary Fig. 1, see section on supplementary data given at the end of this article). For this, we queried the databases miRTarbase release 6.1 (31) and starBase, v2.0 (32) of experimentally validated data concerning miRNA–target interactions, restricting the search for interactions classified as functional in miRTarBase (33), and interactions predicted by two or more software with at least one supporting experiment in starBase. Moreover, we obtained the complete collection of validated targets provided by miRecords. The union of all interactions retrieved from the 3 queried sources was considered as the set of validated miRNA–target gene interactions in our study.

To complement the information derived from experimental validation and search for additional miRNA targets, we also applied in silico target prediction algorithms for selected miRNA sequences using web-based tools TargetScan, v7.1 (34), Diana MicroT-CDS (35) and miRanda-mirSVR (August 2010 Release) (36, 37). To control for false-positive rates, we adopted the following filtering criteria: (1) for TargetScan, v7.1, we considered interactions involving conserved miRNA sites and with context++ scores <−0.1; (2) for Diana MicroT-CDS, we kept interactions with prediction scores ≥0.7; (3) for miRanda-mirSVR, we selected interactions involving conserved miRNAs and with scores <−0.1; (4) the compilation of miRNA–target interactions gathered from in silico analysis was built based on target genes predicted by at least 2 adopted computational tools. The combination of validated and predicted miRNA–target interactions was used for further analyses. miRNAs and gene identifiers were mapped to miRBase, v21 and Human Gene Nomenclature Committee (38, 39) or Mouse Genome Information nomenclature (40, 41).

Next, we implemented functional enrichment analysis of miRNAs target genes using pathways annotation from the KEGG Pathway Database (42, 43) and the clusterProfiler package in R/Bioconductor environment (44). This investigation was performed for targets of each individual miRNA as well as for targets of miRNAs grouped by tissue (PBMCs, serum/plasma or pancreas). Significance for KEGG pathways enrichment was estimated with a hypergeometric test and adjusted to account for multiple hypotheses using the false discovery rate (FDR) procedure implemented in the q-value R package (45). Pathways with a q-value <0.05 were considered strongly enriched for the genes targeted by selected miRNAs.

Results

Literature search, characteristics of the eligible studies and quality assessment

The flow diagram showing the strategy used to identify and select studies for inclusion in this systematic review is depicted in Fig. 1. According to the search criteria, a total of 1738 publications were retrieved from databases; however, after full text analysis, only 33 articles (20, 22, 23, 24, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74) fulfilled the eligibility criteria and were included in the review. The main characteristics of these 33 articles are shown in Table 1. Among these studies, 19 reported miRNA expression profiles in human, 13 focused on miRNA profiles in murine models, and only one analyzed both human and murine samples (46). Sample sizes ranged from 10 to 162 in studies that analyzed human samples and from 6 to 60 in studies with murine models. The number of miRNAs analyzed ranged from 1 to 847, with the number of miRNAs differentially expressed between groups varying from 1 to 136 (Table 1).

Figure 1
Figure 1

Flowchart illustrating the search strategy used to identify association studies of miRNAs expression with type 1 diabetes for inclusion in the systematic review.

Citation: Endocrine Connections 6, 8; 10.1530/EC-17-0248

Table 1

Characteristics of studies included in the systematic review.

T1DM Differentially expressed microRNAs
First author, year (Ref) Country Diabetic sample Tissue Sample size Case/Control Method Cut-off criteria Total Increased Decreased Quality/QUADAS-2
Alipour et al. 2013 (58) Iran Male Sprague-Dawley rats induced with STZ Kidney 6/6 RT-PCR 0.05 1 1 0 5.5
Bacon et al. 2015 (59) Ireland T1DM patients Urine 44/26 RT-PCR N/A 2 2 0 6.0
Barutta et al. 2013 (46) Italy Normoalbuminuric T1DM patients Urinary Exossome 12/12 RT-PCR 2-fold 2 2 0 6.0
Barutta et al. 2013 (46) Italy Male C57BL6/J mice induced with STZ Glomeruli 30/30 RT-PCR 2-fold 1 1 0 5.5
Barutta et al. 2013 (46) Italy Male C57BL6/J mice induced with STZ Exossome 30/30 RT-PCR 2-fold 1 1 0 5.5
Diao et al. 2011 (47) China Male C57BL/6 mice induced with STZ Heart 15/10 Microarray analysis 0.05 16 10 6 6.0
Emadi et al. 2014 (48) Iran Male Wistar rats Aorta 6/6 RT-PCR 0.05 1 0 1 6.0
Erener et al. 2013 (63) Canada C57BL/6 mice induced with STZ and NOD mice Plasma 6/6 RT-PCR 0.05 1 1 0 5.5
Erener et al. 2017 (71) Canada Recent-onset T1DM patients Serum 10/7 RT-PCR N/A 35 27 8 6.0
Estrella et al. 2016 (65) Chile T1DM patients PBMCs 20/20 RT-PCR N/A 2 1 1 6.0
Garcia de la Torre et al. 2015 (64) Spain T1DM patients without DR EPC 76/38 RT-PCR 2-fold 1 1 0 6.0
Garcia-Contreras et al. 2017 (72) USA T1DM patients Plasma-derived exosome 36/36 Microarray and RT-PCR N/A 7 1 6 5.5
Hezova et al. 2010 (49) Czech Republic T1DM patients T cells 5/5 TLDA 0.05 3 1 2 5.5
Kato et al. 2010 (50) USA C57BL/6 mice induced with STZ Kidney 3/3 RT-PCR 0.05 1 1 0 6.0
Kovacs et al. 2011 (51) USA Male Sprague-Dawley rats induced with STZ Retina 3/3 Microarray 0.05 17 14 3 5.5
Li et al. 2009 (67) China C57BL/6 mice induced wit STZ Liver 8/8 Microarray 0.05 2 1 1 5.5
Ma et al. 2016 (61) China NOD mice Pancreas 6/6 RT-PCR N/A 1 0 1 6.0
Marchand et al. 2016 (66) France T1DM patients recently diagnosed Serum 22/10 RT-PCR 0.05 1 1 0 6.0
Nabih et al. 2016 (62) Egypt Children and adolescents with T1DM Serum 40/40 RT-PCR N/A 1 1 0 6.0
Nielsen et al. 2012 (22) Denmark Children with newly diagnosed T1DM Serum 108/54 Solexa sequencing/RT-PCR 2-fold 24 24 0 6.5
Osipova et al. 2014 (52) Germany T1DM pediatric patients Serum 68/79 RT-PCR 0.05 2 2 0 6.0
Osipova et al. 2014 (52) Germany T1DM pediatric patients Urine 68/79 RT-PCR 0.05 3 2 1 6.0
*Perez-Bravo et al. 2014 (69) Chile T1DM patients PBMCs 5/5 RT-PCR N/A 1 0 1
Qing et al. 2014 (56) China T1DM patients Retina 90/20 TLDA N/A 3 3 0 5.0
Salas-Perez et al. 2013 (20) Chile T1DM pediatric patients PBMCs 20/20 RT-PCR 0.05 2 0 2 5.0
*Sebastiani et al. 2012 (68) Italy T1DM patients Serum 20/20 TLDA 0.05 64 21 43
Sebastiani et al. 2017 (74) Italy T1DM patients T-cells from pancreatic lymphnodes Megaplex RT-stem-loop microRNA Pool A v2.1 N/A 1 1 0 6.0
Seyhan et al. 2016 (70) USA T1DM patients Plasma 16/27 RT-PCR 0.05 4 4 0 6.5
Silva et al. 2011 (53) Brazil Male Wistar rats, induced with STZ Retina 3/3 RT-PCR 0.05 1 1 0 6.5
Takahashi et al. 2014 (23) Brazil T1DM patients PBMCs 6/6 Microarray N/A 44 35 9 6.0
Tian et al. 2015 (60) China Male NIH mice induced with STZ Pancreas 3/3 Microarray 2-fold 136 64 72 6.0
Wang et al. 2014 (54) USA Male Long Evans rats induced with STZ Retina 3/3 RT-PCR N/A 1 1 0 5.5
Wang et al. 2017 (73) China T1DM patients PBMCs 78/56 RT-PCR N/A 3 0 3 6.0
Xiong et al. 2014 (57) China Rat induced with STZ Retina 6/6 RT-PCR 0.05 17 15 2 6.0
Yang et al. 2015 (24) China Newly diagnosed T1DM patients PBMCs 12/10 Microarray/ RT-PCR 0.05 24 5 19 6.5
Yousefzadeh et al. 2015 (55) Iran Male Sprague-Dawley rats induced with STZ Sciatic nerve 6/6 RT-PCR 0.05 1 1 0 6.0

DR, diabetic retinopathy; EPC, Endothelial Progenitor cells; N/A, not available; QUADAS-2, Quality Assessment of Diagnostic Accuracy Studies 2; RT-PCR, Reverse transcription polymerase chain reaction; STZ, streptozotocin; T1DM, Type 1 diabetes mellitus; TLDA, TaqMan Low Density Array. *Abstract from Congress.

Regarding tissues analyzed, 24.1% of the studies evaluated miRNA expression in serum/plasma samples, 20.7% in PBMCs/T cells, and 6.9% in pancreas tissue. The remaining studies evaluated other tissues related to T1DM chronic complications, such as urine, kidney, heart and retina (Table 1). Two articles analyzed different tissues (46, 52) and were considered separately, totalizing 36 studies.

Quality of each study included in this review was assessed using QUADAS-2, as reported in the Methods section. Overall, most studies were considered as having a good quality since 62.5% of studies received QUADAS-2 scores between 6 and 7 (Table 1). No study scored less than 5.0.

Dysregulated miRNAs in T1DM-related tissues

Out of 278 dysregulated miRNAs reported in 36 studies that compared T1DM patients and controls, 72 miRNAs (25.9%) were reported in at least two studies (Supplementary Table 1). However, only 48 of them were analyzed in tissues directly related to T1DM pathogenesis (PBMCs, serum/plasma and pancreas). Hence, these 48 miRNAs were chosen for further evaluation (Table 2).

Table 2

miRNAs differently expressed in tissues related to T1DM analyzed in at least two studies.

miRNA ID First author (ref.) Species Sample type Change of expression
let-7a-5p Tian et al. (60) Mice Pancreas Down
Yang et al. (24) Human Pancreas Down
let-7c-5p Tian et al. (60) Mice Pancreas Down
Yang et al. (24) Human Pancreas Down
let-7f-5p Takahashi et al. (23) Human PBMCs Up
Yang et al. (24) Human PBMCs Down
Tian et al. (60) Mice Pancreas Down
let-7g-5p Takahashi et al. (23) Human PBMCs Up
Erener et al. (71) Human Serum Up
Yang et al. (24) Human PBMCs Down
Tian et al. (60) Mice Pancreas Down
miR-10a-5p Nielsen et al. (22) Human Serum Up
Takahashi et al. (23) Human PBMCs Up
Tian et al. (60) Mice Pancreas Down
miR-100-5p Hezova et al. (49) Human T cells Down
Erener et al. (71) Human Serum Down
miR-126-3p Takahashi et al. (23) Human PBMCs Up
Tian et al. (60) Mice Pancreas Down
miR-1275 Takahashi et al. (23) Human PBMCs Down
Yang et al. (24) Human PBMCs Down
miR-146a-5p Yang et al. (24) Human PBMCs Down
Hezova et al. (49) Human T cells Up
Sebastiani et al. (68) Human Serum Down
Perez-Bravo et al. (69) Human PBMCs Down
Wang et al. 2017 (73) Human PBMCs Down
miR-148a-3p Nielsen et al. (22) Human Serum Up
Takahashi et al. (23) Human PBMCs Up
Seyhan et al. (70) Human Plasma Up
miR-148b-3p Takahashi et al. (23) Human PBMCs Up
Tian et al. (60) Mice Pancreas Down
miR-150-5p Estrella et al. (65) Human PBMCs Down
Wang et al. 2017 (73) Human PBMCs Down
miR-151-3p Hezova et al. (49) Human T cells Down
Tian et al. (60) Mice Pancreas Down
miR-154-3p Tian et al. (60) Mice Pancreas Up
Erener et al. (71) Human Serum Down
miR-15b Takahashi et al. (23) Human PBMCs Up
Yang et al. (24) Human PBMCs Down
miR-16-5p Takahashi et al. (23) Human PBMCs Up
Garcia-Contreras et al. (72) Human Plasma-derived exosome Down
Tian et al. (60) Mice Pancreas Down
miR-181a-5p Nielsen et al. (22) Human Serum Up
Nabih et al. (62) Human Serum Up
miR-199a-3p Takahashi et al. (23) Human PBMCs Up
Sebastiani et al. (68) Human Serum Down
miR-19a-3p Takahashi et al. (23) Human PBMCs Up
Tian et al. (60) Mice Pancreas Down
miR-200c-3p Nielsen et al. (22) Human Serum Up
Yang et al. (24) Human PBMCs Down
miR-20b-5p Hezova et al. (49) Human T cells Down
Takahashi et al. (23) Human PBMCs Up
miR-210-5p Nielsen et al. (22) Human Serum Up
Osipova a et al. (52) Human Serum Up
miR-21-5p Seyhan et al. (70) Human Plasma Up
Nielsen et al. (22) Human Serum Up
Osipova et al. (52) Human Serum Up
Takahashi et al. (23) Human PBMCs Up
miR-221-3p Erener et al. 2017 (71) Human Serum Up
Yang et al. (24) Human PBMCs Down
miR-22-3p Yang et al. (24) Human PBMCs Down
Estrella et al. (65) Human PBMCs Up
miR-24-3p Seyhan et al. (70) Human Plasma Up
Erener et al. 2017 (71) Human Serum Up
Nielsen et al. (22) Human Serum Up
miR-25-3p Nielsen et al. (22) Human Serum Up
Garcia-Contreras et al. (72) Human Plasma-derived exosome Up
Erener et al. 2017 (71) Human Serum Up
Yang et al. (24) Human PBMCs Down
miR-26a-5p Nielsen et al. (22) Human Serum Up
Ma et al. (61) Mice Pancreas Down
Tian et al. (60) Mice Pancreas Down
miR-26b-5p Nielsen et al. (22) Human Serum Up
Takahashi et al. (23) Human PBMCs Up
Tian et al. (60) Mice Pancreas Down
miR-27a-3p Nielsen et al. (22) Human Serum Up
Tian et al. (60) Mice Pancreas Down
miR-27b-3p Nielsen et al. (22) Human Serum Up
Takahashi et al. (23) Human PBMCs Up
miR-30b-3p Tian et al. (60) Mice Pancreas Up
Yang et al. (24) Human PBMCs Down
miR-324-3p Tian et al. (60) Mice Pancreas Down
Erener et al. 2017 (71) Human Serum Up
miR-324-5p Takahashi et al. (23) Human PBMCs Down
Erener et al. 2017 (71) Human Serum Up
Tian et al. (60) Mice Pancreas Down
miR-32-5p Takahashi et al. (23) Human PBMCs Up
Tian et al. (60) Mice Pancreas Down
miR-335-5p Hezova et al. (49) Human T cells Down
Takahashi et al. (23) Human PBMCs Up
miR-342-3p Takahashi et al. (23) Human PBMCs Down
Sebastiani et al. (68) Human Serum Up
Yang et al. (24) Human PBMCs Down
miR-375 Erener et al. (63) Mice Plasma Up
Marchand et al. (66) Human Serum Up
Sebastiani et al. (68) Human Serum Down
Seyhan et al. (70) Human Plasma Up
miR-377-3p Sebastiani et al. (68) Human Serum Down
Erener et al. 2017 (71) Human Serum Up
miR-378 Erener et al. (63) Mice Plasma Up
Garcia-Contreras et al. (72) Human Plasma-derived exosome Down
miR-424-5p Wang et al. 2017 (73) Human PBMCs Down
Takahashi et al. (23) Human PBMCs Up
miR-450a-2-3p Takahashi et al. (23) Human PBMCs Up
Tian et al. (60) Mice Pancreas Up
miR-454-3p Takahashi et al. (23) Human PBMCs Up
Erener et al. 2017 (71) Human Serum Up
miR-490-5p Tian et al. (60) Mice Pancreas Up
Erener et al. 2017 (71) Human Serum Down
miR-574-3p Garcia-Contreras et al. (72) Human Plasma-derived exosome Down
Tian et al. (60) Mice Pancreas Down
miR-720 Takahashi et al. (23) Human PBMCs Down
Erener et al. 2017 (71) Human Serum Down
miR-9-3p Tian et al. (60) Mice Pancreas Up
Sebastiani et al. (68) Human Serum Down
miR-98-5p Takahashi et al. (23) Human PBMCs Up
Tian et al. (60) Mice Pancreas Down

Eight miRNAs were consistently downregulated in T1DM-related tissues from patients compared to controls (miR-100-5p, miR-1275, miR-150-5p, miR-151-3p, miR-146a-5p, miR-151-3p, miR-574-3p and miR-720), while 10 miRNAs were upregulated in cases (miR-21-5p, miR-24-3p, miR-25-3p, miR-27b-3p, miR-148a-3p, miR-181a-5p, miR-210-5p, miR-375, miR-450a-2-3p and miR-454-3p) (Fig. 2A and Table 2). Thirty miRNAs were reported as being downregulated in cases from one study and upregulated in cases from another study, possibly due to the different tissues or species that were analyzed (Table 2).

Figure 2
Figure 2

miRNA expression analyzed in at least two studies considering their expression profile in all tissues analyzed independently of the species (A). miRNA expression profile between the species (B). miRNA expression profile in tissues related to T1DM pathogenesis (C).

Citation: Endocrine Connections 6, 8; 10.1530/EC-17-0248

miRNA expression profiles according to species

In subgroup analysis of species, 19 studies reported expressions of 139 miRNAs in different tissues from T1DM patients and controls, with 36 of these miRNAs being reported by at least two studies (Supplementary Table 1). One study analyzed both human and murine samples. Additionally, 13 miRNA profile studies were performed in murine models of T1DM, identifying 173 dysregulated miRNAs in different tissues, with only 45 of them being reported by at least two studies (Supplementary Table 1).

Considering only the 48 miRNAs expressed in serum/plasma, PBMCs or pancreas, 12 miRNAs were dysregulated exclusively in human samples, with 4 miRNAs (miR-100-5p, miR-146a-5p miR-150-5p and miR-1275) being downregulated and 8 upregulated (miR-10a-5p, miR-21-5p, miR-24-3p miR-26b-5p, miR-27b-3p, miR-148a-3p, miR-181a-5p and miR-210-5p) in T1DM patients compared to controls. Only one miRNA (mmu-miR-26a-5p) was consistently downregulated in pancreas from murine models of T1DM. Four miRNAs (miR-151-3p, miR-324-5p, let-7a-5p and let-7c-5p) were shown to be downregulated in tissues from both human and mice with diabetes, and only miR-375 was upregulated in tissues from human and mice (Fig. 2B and Table 2).

Dysregulated miRNAs as circulating and tissue biomarkers of T1DM

Several miRNAs are released into the bloodstream or expressed in blood cells and might be used as circulating biomarkers of T1DM (2). Among miRNAs that were analyzed in more than one study, 21 (miR-15b, miR-20b-5p, miR-21-5p, miR-22-3p, miR-24-3p, miR-25-3p, miR-26b-5p, miR-27b-3p, miR-100-5p, miR-146a-5p, miR-148a-3p, miR-150-5p, miR-181a-5p, miR-200c-3p, miR-210-5p, miR-335-5p, miR-342-3p, miR-375, miR-1275, let-7f-5p and let-7g-5p) were expressed in serum/plasma or PBMCs/T cells and, therefore, have the potential to be circulating biomarkers of T1DM (Table 2). Nevertheless, only 11 of them were consistently dysregulated, being analyzed in the same tissue by at least two studies: miR-146a-5p, miR-150-5p, miR-342-3p and miR-1275 were downregulated in PBMCs from T1DM cases compared to controls, while miR-21-5p, miR-24-3p, miR-100-5p, miR-148a-3p, miR-181a-5p, miR-210-5p and miR-375 were upregulated in serum/plasma from cases (Fig. 2C and Table 2).

Two studies evaluated miRNA expression profiles in pancreas from murine models of T1DM (60, 61) and showed that miR-26a-5p expression was downregulated in pancreas from NOD or streptozotocin (STZ)-induced diabetes mice compared to control mice (Fig. 2C and Table 2). No study has evaluated miRNA expressions in human pancreas from T1DM patients and non-diabetic controls.

Moreover, 3 miRNAs (miR-151-3p, let-7a-5p and let-7c-5p) were downregulated in pancreas from diabetic mice as well as PBMCs/T cells from T1DM patients compared to the respective control groups. Inversely, several miRNAs were downregulated in pancreas from diabetic mice but upregulated in PBMCs or serum/plasma from T1DM patients (Table 2), which might reflect differential expression in tissues and/or species.

Perturbed pathways in type 1 diabetes mellitus

Bioinformatic analyses were performed to retrieve putative targets and pathways potentially modulated by 12 miRNAs (hsa-miR-21-5p, hsa-miR-24-3p, mmu-miR-26a-5p, hsa-miR-100-5p, hsa-miR-146a-5p, hsa-miR-148a-3p, has-miR-150-5p, hsa-miR-181a-5p, hsa-miR-210-5p, hsa-miR-342-3p, hsa-miR-375 and hsa-miR-1275) consistently dysregulated in T1DM-related tissues. Species prefixes were used in miRNA identifiers to clearly designate the species under consideration while reporting these results. First, we searched for targets of these miRNAs using 6 distinct resources, including experimentally validated databases and prediction programs (Supplementary Fig. 1). A total of 5867 validated and 2979 predicted miRNA–target interactions were retrieved for human miRNAs, while 573 validated and 453 predicted interactions were retrieved for the mmu-miR-26a-5p (Table 3; Supplementary Table 2).

Table 3

Number of miRNA-target interactions for each analyzed miRNA considered individually and grouped by tissue related to T1DM.

miRNA/tissue Validated interactions Predicted interactions
Analysis by miRNA hsa-miR-1275 121
has-miR-100-5p 279 14
hsa-miR-146a-5p 300 409
hsa-miR-148a-3p 621 375
has-miR-150-5p 637 433
hsa-miR-181a-5p 1159 361
hsa-miR-21-5p 725 198
has-miR-24-3p 1052 450
hsa-miR-210-5p 40
hsa-miR-342-3p 433 436
hsa-miR-375 500 303
mmu-miR-26a-5p 573 453
Analysis by tissue related to T1DM PBMCs 1491 1278
Serum/Plasma 4376 1701
Pancreas 573 453

PBMCs, peripheral blood mononuclear cells.

After target prediction, we performed functional enrichment analysis of miRNA target genes using pathway maps from the KEGG Pathway Database, aiming to better understand the biological pathways affected by the selected miRNAs. Out of 518 pathways annotated in KEGG Database (accessed in August 2017), a total of 127 pathways were significantly overrepresented (q-value <0.05) in the putative target lists analyzed, and 77 KEGG terms were enriched for more than one miRNA. Targets of hsa-miR-21-5p, hsa-miR-24-3p, mmu-miR-26a-5p, hsa-miR-100-5p, hsa-miR-146a-5p, hsa-miR-148a-3p, hsa-miR-150-5p hsa-miR-181a-5p, hsa-miR-342-5p and hsa-miR-375 are involved in several pathways (Supplementary Table 3), many of them having a recognized role in T1DM pathogenesis, such as TNF, MAPK, Jak-STAT, PI3K-Akt, apoptosis, insulin, toll-like receptors (TLRs) and T cell receptor (TCR) signaling pathways (Supplementary Table 3). No significantly enriched KEGG terms were found for hsa-miR-210-5p and hsa-miR-1275, probably due to the small number of retrieved targets (40 and 121, respectively), as well as for hsa-miR-150-5p despite its broad regulatory action.

Considering different T1DM-related tissues, results indicated 2539 targets in PBMCs, 4665 targets in serum/plasma and 1026 targets in pancreas for the selected miRNAs (Table 3), where numbers reflect the size of the non-redundant set of target genes found for the group of miRNAs differentially expressed in each T1DM-related tissue. Forty-five significant KEGG pathways were found for targets of miRNAs dysregulated in PBMCs, which included NF-ΚB, apoptosis and neurotrophin signaling pathways. Similarly, 17 KEGG terms were found in pancreas, including Wnt and phosphatidylinositol signaling pathways. For serum and plasma, 94 significant KEGG terms were found, comprising signaling pathways by TNF, Jak-STAT, MAPK, TCR and insulin as well as pathways associated to protein processing in endoplasmic reticulum and apoptosis, which have key roles in T1DM pathogenesis (Supplementary Table 4; Fig. 3).

Figure 3
Figure 3

KEGG pathway functional annotation of the differentially miRNAs expressed in PBMCs (A), serum/plasma (B) and pancreas (C). Enrichment scores corresponding to each pathway are displayed as number of targets.

Citation: Endocrine Connections 6, 8; 10.1530/EC-17-0248

Next, we searched for KEGG terms linked to T1DM pathogenesis regardless of the functional enrichment analysis of miRNA targets and found 5 significant signaling pathways associated with this disease: type 1 diabetes (KEGG hsa04949), TCR (KEGG hsa04660), cytokine–cytokine receptor interaction (KEGG hsa04060), Jak-STAT (KEGG hsa04630) and neurotrophin (KEGG hsa04722). Then, TCR and Jak-STAT pathways were selected for further detailed analysis since they are targeted by most of the miRNAs in the list of interest (11 miRNAs each).

In the TCR signaling pathway (Fig. 4), miR-181a-5p directly targets mRNAs for CD4+ and CD8+ cell receptors. Moreover, this miRNA post-transcriptionally regulates genes associated with PI3K-Akt (PI3K, Akt, COT), actin cytoskeleton (PAK4) and ubiquitin mediated proteolysis (CBL) pathways, which are triggered after activation of T cell and co-stimulatory receptors. miR-21-5p targets mRNAs from MAPK (Ras, ErkSOS and RasGRP1) and ubiquitin-mediated proteolysis (FYN) pathways. Furthermore, miR-146a-5p targets mRNAs from MAPK (Ras) and NF-κB signaling pathways. miR-148a-5p, miR-100-5p, miR-24-3p and miR-150-5p target mRNAs from ubiquitin-mediated proteolysis (CBL), PI3K-Akt, NF-κB and MAPK pathways (Fig. 4).

Figure 4
Figure 4

Schematic diagram of the selected miRNA-mRNA interaction networks involved in T cell receptor signaling pathway. The network was built based on KEGG pathway map (KEGG: hsa04660). The miRNAs are indicated by rhombus. The predicted miRNA-mRNA interactions are indicated by dotted lines and the thick lines indicate validated miRNA-mRNA interactions.

Citation: Endocrine Connections 6, 8; 10.1530/EC-17-0248

In the Jak-STAT signaling pathway (Fig. 5), miR-21-5p, miR-24-3p, miR-181a-5p and miR-210-5p target mRNAs for different cytokine and hormone receptors, such as IL6R, LIFR, IL2RB and IFNLR1. miR-375 targets JAK2 mRNA while miR-181a-5p and miR-21-5p bind to STAT3 mRNA. In addition, miR-375, miR-181a-5p, miR-146a-5p, miR-148a-3p, miR-100-5p, miR-150-5p and miR-21-5p target different mRNAs codifying proteins related to apoptosis (BCL2, SOCS, PIAS and MCL1), cell cycle progression (cMyc), cell cycle inhibition (p21), proliferation and differentiation (SHP2, SOS and Ras) and cell survival (PI3K and Akt) (Fig. 5).

Figure 5
Figure 5

Schematic diagram of the selected miRNA-mRNA interaction networks involved in Jak-STAT signaling pathway. The network was built based on KEGG pathway map (KEGG: hsa04630). The miRNAs are indicated by rhombus. The predicted miRNA-mRNA interactions are indicated by dotted lines and the thick lines indicate validated miRNA-mRNA interactions.

Citation: Endocrine Connections 6, 8; 10.1530/EC-17-0248

Discussion

Since the exact origin of T1DM remains uncertain, the discovery of novel biomarkers and their implications in T1DM pathogenesis may contribute to a better understanding of the mechanisms involved in this disease. Clinically, new biomarkers might enable an earlier T1DM diagnosis as well as a more adequate treatment of T1DM patients, improving their quality of life (23). Circulating miRNAs are ideal biomarkers because they are stable and resistant to degradation by ribonucleases or repeated freezing/thawing cycles and can be easily detected in body fluids by highly sensitive and specific quantitative RT-PCR (2). Thus, as part of the ongoing effort to identify a profile of circulating miRNAs as biomarkers of T1DM, we performed a systematic review of studies that evaluated miRNA expressions in tissues from T1DM patients and non-diabetic controls. Eleven circulating miRNAs (miR-21-5p, miR-24-3p, miR-100-5p, miR-146a-5p, miR-148a-3p, miR-150-5p, miR-181a-5p, miR-210-5p, miR-342-3p, miR-375 and miR-1275) were consistently dysregulated in T1DM patients, suggesting that they may be potential minimally invasive biomarkers of this disease.

miR-21-5p, miR-24-3p, miR-148a-3p, miR-181a-5p, miR-210-5p and miR-375 seem to be upregulated in serum/plasma or PBMCs from T1DM compared to controls (Table 2). Emerging studies have indicated diverse roles of miR-21-5p in immunity (75). Particularly, this miRNA acts in TCR signaling transduction, augmenting T cell proliferation (76); regulates Th1 vs Th2 responses (77) and Treg development (78) and is a key mediator of the anti-inflammatory response in macrophages (79). miR-21-5p also appears to have anti-inflammatory and anti-apoptotic effects since it inhibits the proinflammatory tumor suppressor programed cell death protein 4 (PDCD4), which promotes the activation of NF-ΚB and suppresses IL-10 (80). Several studies have reported increased miR-21-5p expression in diseases characterized by impaired immune responses, including asthma, cancer, rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), chronic bacterial and viral infections (14, 79, 81, 82) and T1DM (reviewed here).

The role of miR-21-5p in beta-cells has not yet been clearly elucidated, but its function also seems to rely on the effect of cytokines via NF-ΚB (83). IL-1β and TNF strongly induce miR-21-5p expression in both insulin-secreting MIN6 cells and human islets (18). Moreover, miR-21-5p expression was increased during the development of pre-diabetic insulitis in islets from NOD mice, possibly as a protective response since miR-21-5p knockdown in MIN-6 cells promoted apoptosis (18). Accordingly, Ruan and coworkers (84) reported that NF-ΚB activated miR-21-5p in beta-cells, decreasing PDCD4 levels, inhibiting NF-ΚB activity in a negative regulatory loop, thus rendering beta-cells resistant to death. In contrast, Backe and coworkers (85) showed that miR-21-5p overexpression potentiated cell death after exposure to proinflammatory cytokines, leading to a reduced beta-cell number. Also, overexpression of miR-21-5p led to impaired glucose-stimulated insulin secretion through decreased VAMP2 expression, a secretory granule protein that is essential for insulin exocytosis (18).

miR-148a-3p is a potent regulator of B cell tolerance and autoimmunity through suppression of GADD45a, PTEN and BCL2l11 expressions (86). In agreement with this study, Pan and coworkers (81) showed elevated miR-148a-3p and miR-21-5p expression in CD4+ T cells of SLE patients, which contributed to DNA hypomethylation by targeting the DNA methyltransferase 1 (DNMT1). Melkman-Zehavi and coworkers (87) reported that knockdown of miR-148a-3p in primary islets from mice or cultured beta-cells decreased insulin promoter activity and insulin mRNA levels.

miR-181a-5p also has a recognized role in the immune system (17, 88). This miRNA increases CD19+ B populations and regulates T cell function (17, 89, 90, 91). miR-181a-5p seems to ‘tune’ TCR signal strength by targeting tyrosine phosphatases SHP-2, PTPN22 and DUSP5/6, which enhances the basal activation of the TCR signaling molecules LcK and Erk, thus having an important role in thymic positive and negative selection (17, 88, 90). Xie and coworkers (92) reported that LPS and STZ strongly induced miR-181a-5p in macrophages from mice. Moreover, this miRNA was upregulated in patients with RA, which correlated with levels of proinflammatory cytokines (92). In this context, miR-181a-5p seems to have an anti-inflammatory function since it inhibited the increase of IL-1β, IL-6 and TNF in macrophages treated with LPS (93).

miR-210-5p is currently considered as a ‘master miRNA’ of hypoxic response as it was found upregulated by hypoxia in several cell types analyzed (94, 95). Consequently, miR-210-5p has been linked to various cancers and cardiovascular diseases (95, 96, 97, 98). Targets of this miRNA are involved in mitochondrial metabolism, angiogenesis, DNA repair and cell survival (95). Given that miR-210-5p targets many mitochondrial components, it is not surprising that manipulation of this miRNA leads to mitochondrial dysfunction and oxidative stress (94).

miR-375 is the most abundant miRNA detected in islets and is important for the development and maintenance of normal alpha- and beta-cell mass and insulin synthesis and secretion (16, 99, 100). Consequently, this miRNA has been proposed as a biomarker to detect beta-cell death and to predict the development of T1DM (2, 63, 66). Accordingly, massive beta-cell loss elicited by administration of STZ in C57BL/6 mice caused a dramatic increase in circulating levels of miR-375 (63). Moreover, plasma levels of this miRNA were increased in NOD mice 2 weeks before the onset of T1DM (63).

miR-24-3p seems to mark better preserved beta-cell function and/or insulin sensitivity 12 months after diagnosis (101). Moreover, this miRNA was elevated in serum of T1DM children (22), and its overexpression has been shown to inhibit beta-cell proliferation and insulin secretion (102).

miR-146a-5p, miR-150-5p, miR-342-3p, miR-1275 and miR-100-5p seem to be downregulated in serum/plasma or PBMCs from T1DM patients (Table 2). miR-146a-5p regulates Treg-mediated suppression of IFNγ-dependent Th1 responses and associated autoimmunity by directly targeting STAT-1 (103), which was confirmed in our bioinformatic analysis (Supplementary Table 3). This miRNA has also a recognized role in innate immunity by negatively regulating the inflammatory response after recognition of bacterial components by TLRs on monocytes and macrophages (104). Upon activation by TNF and IL-1β (18), this miRNA downregulates TRAF-6 and IRAK-1 expressions, decreasing NF-ΚB activity, which seems to be a fine-tuning mechanism that prevents the overstimulation of the TLR pathway (104, 105). In disagreement with the downregulation of miR-146-5p in serum/PBMCs from T1DM patients, Roggli and coworkers (18) reported that this miRNA was increased in islets from NOD mice during development of insulitis. Blocking miR-146a-5p protected MIN6 cells from cytokine-induced apoptosis and also prevented the reduction in glucose-stimulated insulin secretion observed after IL-1β exposure (18).

Although literature on miR-100 is sparse, it has been implicated in some types of cancer (106, 107, 108) and tissue differentiation (109, 110, 111). Expression of circulating miR-100-5p was significantly decreased in obese normoglycemic subjects and subjects with type 2 diabetes mellitus (T2DM) compared to healthy, lean individuals. Moreover, in visceral adipose tissue, expression of miR-100 was lower in obese subjects with T2DM compared to obese subjects without T2DM (112).

miR-342-3p has been found to be dysregulated in different cancers (113, 114, 115), SLE (14), obesity (116, 117), diabetic kidney disease (118), T2DM and gestational diabetes (119). It is involved in beta-cell differentiation and maturation by targeting FOXA2 and MAFB (120). Furthermore, miR-342-3p was downregulated in human leukocytes in response to LPS (121) and in Tregs from T1DM patients (49), suggesting that this miRNA may be involved in the development of autoimmunity and inflammation in T1DM patients.

Only a few studies have analyzed miR-1275 expression in different diseases. Due to its downregulation in certain cancers, miR-1275 has been referred to as a tumor suppressor (122, 123, 124, 125). In a HuH-7 hepatocarcinome cell line, miR-1275 overexpression suppressed IGF2BP expression, effectively impairing tumor cell proliferation, migration, viability and colony formation (124). The role of this miRNA in autoimmunity and beta-cell function is unknown.

Bezman and coworkers (126) showed that miR-150 plays a critical role in the innate immune system, and decreased expression level of miR-150-5p was negatively associated with GADA autoantibody titers, independently of hyperglycemia and disease duration (73).

Our bioinformatic analysis suggest that miR-21-5p, miR-24-3p, miR-100-5p, miR-146a-5p, miR-148a-3p, miR-150-5p, miR-181a-5p, miR-210-5p, miR-342-3p, miR-375 and miR-1275 significantly regulate several mRNAs from pathways related to immune system and T1DM pathogenesis, such as MAPK, Jak-STAT, NF-ΚB, PI3K-Akt, apoptosis, TNF, TLRs, insulin and TCR signaling pathways (Figs 3, and ). This might help to raise hypothesis about genes and pathways under influence of these circulating miRNAs.

Regarding the pathway analysis methods, there are many tools in literature that provide support for this type of investigation. Nonetheless, they are very similar as they all calculate the enrichment P values of pathways for a user pre-selected list of genes using a statistical test and a database with functional annotation that links genes to biological pathways. In addition, the hypergeometric test, which we adopted in our paper, has been widely applied in literature and previous studies discuss that when identifying significant pathways, the differences among the statistical methods will not be dramatic (127). The functional annotation has been traditionally performed in literature using annotations derived either from Gene Ontology (GO) or KEGG Pathway. KEGG Pathway Database has less annotated terms compared to GO, but it covers a wide range of molecular mechanisms and diseases, providing a graphical description of pathways, which is an important resource in the interpretation of results. Thus, bioinformatic tools used in this study provide robust data, which might be very similar to those generated using different tools.

In conclusion, this systematic review and bioinformatic analysis suggest that 11 circulating miRNAs (miR-21-5p, miR-24-3p, miR-100-5p, miR-146a-5p, miR-148a-3p, miR-150-5p, miR-181a-5p, miR-210-5p, miR-342-3p, miR-375 and miR-1275) are consistently dysregulated in T1DM patients. Further studies aiming at clarifying the specific role of these 11 miRNAs in pancreatic islets and islet-infiltrating immune cells are needed to shed light if they are biomarkers of T1DM and which are their specific roles in beta-cell function.

Supplementary data

This is linked to the online version of the paper at http://dx.doi.org/10.1530/EC-17-0248.

Declaration of interest

The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of the research reported.

Funding

This study was supported by grants from the Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq), Fundação de Amparo à Pesquisa do Estado do Rio Grande do Sul (Fapergs), Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (Capes), Fundo de Incentivo à Pesquisa e Eventos (FIPE) at Hospital de Clínicas de Porto Alegre and Post-graduation program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul. D C and T S A are the recipients of a scholarship from CNPq.

References

  • 1

    American Diabetes Association. Classification and diagnosis of diabetes. Diabetes Care 2015 38 S8S16. (doi:10.2337/dc15-S005)

  • 2

    Guay C & Regazzi R. Circulating microRNAs as novel biomarkers for diabetes mellitus. Nature Reviews Endocrinology 2013 9 513521. (doi:10.1038/nrendo.2013.86)

  • 3

    Atkinson MA & Eisenbarth GS. Type 1 diabetes: new perspectives on disease pathogenesis and treatment. Lancet 2001 358 221229. (doi:10.1016/S0140-6736(01)05415-0)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 4

    Pirot P, Cardozo AK & Eizirik DL. Mediators and mechanisms of pancreatic beta-cell death in type 1 diabetes. Arquivos Brasileiros De Endocrinologia e Metabologia 2008 52 156165. (doi:10.1590/S0004-27302008000200003)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 5

    Ziegler AG, Rewers M, Simell O, Simell T, Lempainen J, Steck A, Winkler C, Ilonen J, Veijola R, Knip M et al.. Seroconversion to multiple islet autoantibodies and risk of progression to diabetes in children. JAMA 2013 309 24732479. (doi:10.1001/jama.2013.6285)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 6

    Eisenbarth GS & Jeffrey J. The natural history of type 1A diabetes. Arquivos Brasileiros De Endocrinologia e Metabologia 2008 52 146155. (doi:10.1590/S0004-27302008000200002)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 7

    Purohit S & She JX. Biomarkers for type 1 diabetes. International Journal of Clinical and Experimental Medicine 2008 1 98116.

  • 8

    Bonifacio E. Predicting type 1 diabetes using biomarkers. Diabetes Care 2015 38 989996. (doi:10.2337/dc15-0101)

  • 9

    Esteller M. Non-coding RNAs in human disease. Nature Reviews Genetics 2011 12 861874. (doi:10.1038/nrg3074)

  • 10

    Butz H, Kinga N, Racz K & Patocs A. Circulating miRNAs as biomarkers for endocrine disorders. Journal of Endocrinological Investigation 2016 39 110. (doi:10.1007/s40618-015-0316-5)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 11

    Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell 2009 136 215233. (doi:10.1016/j.cell.2009.01.002)

  • 12

    Carrington JC & Ambros V. Role of microRNAs in plant and animal development. Science 2003 301 336338. (doi:10.1126/science.1085242)

  • 13

    Lin S & Gregory RI. MicroRNA biogenesis pathways in cancer. Nature Reviews Cancer 2015 15 321333. (doi:10.1038/nrc3932)

  • 14

    Pauley KM, Cha S & Chan EK. MicroRNA in autoimmunity and autoimmune diseases. Journal of Autoimmunity 2009 32 189194. (doi:10.1016/j.jaut.2009.02.012)

  • 15

    Zalts H & Shomron N. The impact of microRNAs on endocrinology. Pediatric Endocrinology Reviews 2011 8 354362.

  • 16

    Guay C, Roggli E, Nesca V, Jacovetti C & Regazzi R. Diabetes mellitus, a microRNA-related disease? Translational Research 2011 157 253264. (doi:10.1016/j.trsl.2011.01.009)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 17

    Baltimore D, Boldin MP, O'Connell RM, Rao DS & Taganov KD. MicroRNAs: new regulators of immune cell development and function. Nature Immunology 2008 9 839845. (doi:10.1038/ni.f.209)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 18

    Roggli E, Britan A, Gattesco S, Lin-Marq N, Abderrahmani A, Meda P & Regazzi R. Involvement of microRNAs in the cytotoxic effects exerted by proinflammatory cytokines on pancreatic beta-cells. Diabetes 2010 59 978986. (doi:10.2337/db09-0881)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 19

    Zheng Y, Wang Z, Tu Y, Shen H, Dai Z, Lin J & Zhou Z. miR-101a and miR-30b contribute to inflammatory cytokine-mediated beta-cell dysfunction. Laboratory Investigation 2015 95 13871397. (doi:10.1038/labinvest.2015.112)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 20

    Salas-Perez F, Codner E, Valencia E, Pizarro C, Carrasco E & Perez-Bravo F. MicroRNAs miR-21a and miR-93 are down regulated in peripheral blood mononuclear cells (PBMCs) from patients with type 1 diabetes. Immunobiology 2013 218 733737. (doi:10.1016/j.imbio.2012.08.276)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 21

    Sebastiani G, Grieco FA, Spagnuolo I, Galleri L, Cataldo D & Dotta F. Increased expression of microRNA miR-326 in type 1 diabetic patients with ongoing islet autoimmunity. Diabetes/Metabolism Research and Reviews 2011 27 862866. (doi:10.1002/dmrr.1262)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 22

    Nielsen LB, Wang C, Sorensen K, Bang-Berthelsen CH, Hansen L, Andersen ML, Hougaard P, Juul A, Zhang CY, Pociot F et al.. Circulating levels of microRNA from children with newly diagnosed type 1 diabetes and healthy controls: evidence that miR-25 associates to residual beta-cell function and glycaemic control during disease progression. Experimental Diabetes Research 2012 2012 896362. (doi:10.1155/2012/672865)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 23

    Takahashi P, Xavier DJ, Evangelista AF, Manoel-Caetano FS, Macedo C, Collares CV, Foss-Freitas MC, Foss MC, Rassi DM, Donadi EA et al.. MicroRNA expression profiling and functional annotation analysis of their targets in patients with type 1 diabetes mellitus. Gene 2014 539 213223. (doi:10.1016/j.gene.2014.01.075)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 24

    Yang M, Ye L, Wang B, Gao J, Liu R, Hong J, Wang W, Gu W & Ning G. Decreased miR-146 expression in peripheral blood mononuclear cells is correlated with ongoing islet autoimmunity in type 1 diabetes patients. Journal of Diabetes 2015 7 158165. (doi:10.1111/1753-0407.12163)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 25

    Abuhatzira L, Xu H, Tahhan G, Boulougoura A, Schaffer AA & Notkins AL. Multiple microRNAs within the 14q32 cluster target the mRNAs of major type 1 diabetes autoantigens IA-2, IA-2beta, and GAD65. FASEB Journal 2015 29 43744383. (doi:10.1096/fj.15-273649)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 26

    Moher D, Liberati A, Tetzlaff J, Altman DG & Group P. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. Journal of Clinical Epidemiology 2009 62 10061012. (doi:10.1016/j.jclinepi.2009.06.005)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 27

    Stroup DF, Berlin JA, Morton SC, Olkin I, Williamson GD, Rennie D, Moher D, Becker BJ, Sipe TA & Thacker SB. Meta-analysis of observational studies in epidemiology: a proposal for reporting Meta-analysis Of Observational Studies in Epidemiology (MOOSE) group. JAMA 2000 283 20082012. (doi:10.1001/jama.283.15.2008)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 28

    Whiting PF, Rutjes AW, Westwood ME, Mallett S, Deeks JJ, Reitsma JB, Leeflang MM, Sterne JA, Bossuyt PM & Group Q-. QUADAS-2: a revised tool for the quality assessment of diagnostic accuracy studies. Annals of Internal Medicine 2011 155 529536. (doi:10.7326/0003-4819-155-8-201110180-00009)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 29

    Brazma A, Hingamp P, Quackenbush J, Sherlock G, Spellman P, Stoeckert C, Aach J, Ansorge W, Ball CA, Causton HC et al.. Minimum information about a microarray experiment (MIAME)-toward standards for microarray data. Nature Genetics 2001 29 365371. (doi:10.1038/ng1201-365)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 30

    Bustin SA, Benes V, Garson JA, Hellemans J, Huggett J, Kubista M, Mueller R, Nolan T, Pfaffl MW, Shipley GL et al.. The MIQE guidelines: minimum information for publication of quantitative real-time PCR experiments. Clinical Chemistry 2009 55 611622. (doi:10.1373/clinchem.2008.112797)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 31

    Chou CH, Chang NW, Shrestha S, Hsu SD, Lin YL, Lee WH, Yang CD, Hong HC, Wei TY, Tu SJ et al.. miRTarBase 2016: updates to the experimentally validated miRNA-target interactions database. Nucleic Acids Research 2016 44 D239D247. (doi:10.1093/nar/gkv1258)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 32

    Li JH, Liu S, Zhou H, Qu LH & Yang JH. starBase v2.0: decoding miRNA-ceRNA, miRNA-ncRNA and protein-RNA interaction networks from large-scale CLIP-Seq data. Nucleic Acids Research 2014 42 D92D97. (doi:10.1093/nar/gkt1248)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 33

    Xiao F, Zuo Z, Cai G, Kang S, Gao X & Li T. miRecords: an integrated resource for microRNA-target interactions. Nucleic Acids Research 2009 37 D105D110. (doi:10.1093/nar/gkn851)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 34

    Agarwal V, Bell GW, Nam JW & Bartel DP. Predicting effective microRNA target sites in mammalian mRNAs. eLife 2015 4 e05005. (doi:10.7554/elife.05005)

  • 35

    Paraskevopoulou MD, Georgakilas G, Kostoulas N, Vlachos IS, Vergoulis T, Reczko M, Filippidis C, Dalamagas T & Hatzigeorgiou AG. DIANA-microT web server v5.0: service integration into miRNA functional analysis workflows. Nucleic Acids Research 2013 41 W169W173. (doi:10.1093/nar/gkt393)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 36

    Betel D, Koppal A, Agius P, Sander C & Leslie C. Comprehensive modeling of microRNA targets predicts functional non-conserved and non-canonical sites. Genome Biology 2010 11 R90. (doi:10.1186/gb-2010-11-8-r90)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 37

    Betel D, Wilson M, Gabow A, Marks DS & Sander C. The microRNA.org resource: targets and expression. Nucleic Acids Research 2008 36 D149D153. (doi:10.1093/nar/gkm995)

  • 38

    Wain HM, Bruford EA, Lovering RC, Lush MJ, Wright MW & Povey S. Guidelines for human gene nomenclature. Genomics 2002 79 464470. (doi:10.1006/geno.2002.6748)

  • 39

    Gray KA, Yates B, Seal RL, Wright MW & Bruford EA. Genenames.org: the HGNC resources in 2015. Nucleic Acids Research 2015 43 D1079D1085. (doi:10.1093/nar/gku1071)

  • 40

    Bult CJ, Eppig JT, Blake JA, Kadin JA, Richardson JE & Mouse Genome Database G. Mouse genome database 2016. Nucleic Acids Research 2016 44 D840D847. (doi:10.1093/nar/gkv1211)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 41

    Eppig JT, Blake JA, Bult CJ, Kadin JA, Richardson JE & Mouse Genome Database G. The Mouse Genome Database (MGD): facilitating mouse as a model for human biology and disease. Nucleic Acids Research 2015 43 D726D736. (doi:10.1093/nar/gku967)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 42

    Kanehisa M, Sato Y, Kawashima M, Furumichi M & Tanabe M. KEGG as a reference resource for gene and protein annotation. Nucleic Acids Research 2016 44 D457D462. (doi:10.1093/nar/gkv1070)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 43

    Kanehisa M & Goto S. KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Research 2000 28 2730. (doi:10.1093/nar/28.1.27)

  • 44

    Yu G, Wang LG, Han Y & He QY. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS 2012 16 284287. (doi:10.1089/omi.2011.0118)

  • 45

    The R Core Team. R: A Language and Environment for Statistical Computing Vienna, Austria: R Foundation for Statistical Computing, 2013.

  • 46

    Barutta F, Tricarico M, Corbelli A, Annaratone L, Pinach S, Grimaldi S, Bruno G, Cimino D, Taverna D, Deregibus MC et al.. Urinary exosomal microRNAs in incipient diabetic nephropathy. PLoS ONE 2013 8 e73798. (doi:10.1371/journal.pone.0073798)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 47

    Diao X, Shen E, Wang X & Hu B. Differentially expressed microRNAs and their target genes in the hearts of streptozotocin-induced diabetic mice. Molecular Medicine Reports 2011 4 633640.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 48

    Emadi SS, Soufi FG, Khamaneh AM & Alipour MR. MicroRNA-146a expression and its intervention in NF-small ka, CyrillicB signaling pathway in diabetic rat aorta. Endocrine Regulations 2014 48 103108. (doi:10.4149/endo_2014_02_103)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 49

    Hezova R, Slaby O, Faltejskova P, Mikulkova Z, Buresova I, Raja KR, Hodek J, Ovesna J & Michalek J. microRNA-342, microRNA-191 and microRNA-510 are differentially expressed in T regulatory cells of type 1 diabetic patients. Cellular Immunology 2010 260 7074. (doi:10.1016/j.cellimm.2009.10.012)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 50

    Kato M, Wang L, Putta S, Wang M, Yuan H, Sun G, Lanting L, Todorov I, Rossi JJ & Natarajan R. Post-transcriptional up-regulation of Tsc-22 by Ybx1, a target of miR-216a, mediates TGF-beta-induced collagen expression in kidney cells. Journal of Biological Chemistry 2010 285 3400434015. (doi:10.1074/jbc.M110.165027)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 51

    Kovacs B, Lumayag S, Cowan C & Xu S. MicroRNAs in early diabetic retinopathy in streptozotocin-induced diabetic rats. Investigative Ophthalmology and Visual Science 2011 52 44024409. (doi:10.1167/iovs.10-6879)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 52

    Osipova J, Fischer DC, Dangwal S, Volkmann I, Widera C, Schwarz K, Lorenzen JM, Schreiver C, Jacoby U, Heimhalt M et al.. Diabetes-associated microRNAs in pediatric patients with type 1 diabetes mellitus: a cross-sectional cohort study. Journal of Clinical Endocrinology and Metabolism 2014 99 E1661E1665. (doi:10.1210/jc.2013-3868)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 53

    Silva VA, Polesskaya A, Sousa TA, Correa VM, Andre ND, Reis RI, Kettelhut IC, Harel-Bellan A & De Lucca FL. Expression and cellular localization of microRNA-29b and RAX, an activator of the RNA-dependent protein kinase (PKR), in the retina of streptozotocin-induced diabetic rats. Molecular Vision 2011 17 22282240.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 54

    Wang Q, Bozack SN, Yan Y, Boulton ME, Grant MB & Busik JV. Regulation of retinal inflammation by rhythmic expression of MiR-146a in diabetic retina. Investigative Ophthalmology and Visual Science 2014 55 39863994. (doi:10.1167/iovs.13-13076)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 55

    Yousefzadeh N, Alipour MR & Soufi FG. Deregulation of NF-small ka, CyrillicB-miR-146a negative feedback loop may be involved in the pathogenesis of diabetic neuropathy. Journal of Physiology and Biochemistry 2015 71 5158. (doi:10.1007/s13105-014-0378-4)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 56

    Qing S, Yuan S, Yun C, Hui H, Mao P, Wen F, Ding Y & Liu Q. Serum miRNA biomarkers serve as a fingerprint for proliferative diabetic retinopathy. Cellular Physiology and Biochemistry 2014 34 17331740. (doi:10.1159/000366374)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 57

    Xiong F, Du X, Hu J, Li T, Du S & Wu Q. Altered retinal microRNA expression profiles in early diabetic retinopathy: an in silico analysis. Current Eye Research 2014 39 720729. (doi:10.3109/02713683.2013.872280)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 58

    Alipour MR, Khamaneh AM, Yousefzadeh N, Mohammad-nejad D & Soufi FG. Upregulation of microRNA-146a was not accompanied by downregulation of pro-inflammatory markers in diabetic kidney. Molecular Biology Reports 2013 40 64776483. (doi:10.1007/s11033-013-2763-4)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 59

    Bacon S, Engelbrecht B, Schmid J, Pfeiffer S, Gallagher R, McCarthy A, Burke M, Concannon C, Prehn JH & Byrne MM. MicroRNA-224 is readily detectable in urine of individuals with diabetes mellitus and is a potential indicator of beta-cell demise. Genes 2015 6 399416. (doi:10.3390/genes6020399)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 60

    Tian C, Ouyang X, Lv Q, Zhang Y & Xie W. Cross-talks between microRNAs and mRNAs in pancreatic tissues of streptozotocin-induced type 1 diabetic mice. Biomedical Reports 2015 3 333342.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 61

    Ma H, Zhang S, Shi D, Mao Y & Cui J. MicroRNA-26a promotes regulatory T cells and suppresses autoimmune diabetes in mice. Inflammation 2016 39 19. (doi:10.1007/s10753-015-0215-0)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 62

    Nabih ES & Andrawes NG. The association between circulating levels of miRNA-181a and pancreatic beta cells dysfunction via SMAD7 in type 1 diabetic children and adolescents. Journal of Clinical Laboratory Analysis 2016 30 727731. (doi:10.1002/jcla.21928)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 63

    Erener S, Mojibian M, Fox JK, Denroche HC & Kieffer TJ. Circulating miR-375 as a biomarker of beta-cell death and diabetes in mice. Endocrinology 2013 154 603608. (doi:10.1210/en.2012-1744)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 64

    Garcia de la Torre N, Fernandez-Durango R, Gomez R, Fuentes M, Roldan-Pallares M, Donate J, Barabash A, Alonso B, Runkle I, Duran A et al.. Expression of angiogenic microRNAs in endothelial progenitor cells from type 1 diabetic patients with and without diabetic retinopathy. Investigative Ophthalmology and Visual Science 2015 56 40904098. (doi:10.1167/iovs.15-16498)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 65

    Estrella S, Garcia-Diaz DF, Codner E, Camacho-Guillen P & Perez-Bravo F. Expression of miR-22 and miR-150 in type 1 diabetes mellitus: possible relationship with autoimmunity and clinical characteristics. Medicina Clinica 2016 147 245247. (doi:10.1016/j.medcli.2016.05.016)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 66

    Marchand L, Jalabert A, Meugnier E, Van den Hende K, Fabien N, Nicolino M, Madec AM, Thivolet C & Rome S. miRNA-375 a sensor of glucotoxicity is altered in the serum of children with newly diagnosed type 1 diabetes. Journal of Diabetes Research 2016 2016 1869082.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 67

    Li S, Chen X, Zhang H, Liang X, Xiang Y, Yu C, Zen K, Li Y & Zhang CY. Differential expression of microRNAs in mouse liver under aberrant energy metabolic status. Journal of Lipid Research 2009 50 17561765. (doi:10.1194/jlr.M800509-JLR200)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 68

    Sebastiani G, Spagnuolo I, Patti A, Grieco FA, Cataldo D, Ferretti E, Tiberti C & Dotta F. MicroRNA expression fingerprint in serum of type 1 diabetic patients. Diabetologia 2012 55 S48.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 69

    Perez-Bravo F, Matthews DR, Haahr HL & Syed F. Differential apoptosis in lymphocytes of patients with type 1 diabetes associated with relative expression of microRNA-146a. Diabetologia 2014 57 (Supplement 1) S144 abstract 332. (doi:10.1007/s00125-014-3355-0)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 70

    Seyhan AA, Nunez Lopez YO, Xie H, Yi F, Mathews C, Pasarica M & Pratley RE. Pancreas-enriched miRNAs are altered in the circulation of subjects with diabetes: a pilot cross-sectional study. Scientific Reports 2016 6 31479.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 71

    Erener S, Marwaha A, Tan R, Panagiotopoulos C & Kieffer TJ. Profiling of circulating microRNAs in children with recent onset of type 1 diabetes. JCI Insight 2017 2 e89656.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 72

    Garcia-Contreras M, Shah SH, Tamayo A, Robbins PD, Golberg RB, Mendez AJ & Ricordi C. Plasma-derived exosome characterization reveals a distinct microRNA signature in long duration Type 1 diabetes. Scientific Reports 2017 7 5998. (doi:10.1038/s41598-017-05787-y)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 73

    Wang G, Gu Y, Xu N, Zhang M & Yang T. Decreased expression of miR-150, miR146a and miR424 in type 1 diabetic patients: association with ongoing islet autoimmunity. Biochemical and Biophysical Research Communications 2017 [in press]. (doi:10.1016/j.bbrc.2017.06.196)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 74

    Sebastiani G, Ventriglia G, Stabilini A, Socci C, Morsiani C, Laurenzi A, Nigi L, Formichi C, Mfarrej B, Petrelli A et al.. Regulatory T-cells from pancreatic lymphnodes of patients with type-1 diabetes express increased levels of microRNA miR-125a-5p that limits CCR2 expression. Scientific Reports 2017 7 6897. (doi:10.1038/s41598-017-07172-1)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 75

    Simpson LJ & Ansel KM. MicroRNA regulation of lymphocyte tolerance and autoimmunity. Journal of Clinical Investigation 2015 125 22422249. (doi:10.1172/JCI78090)

  • 76

    Wang L, He L, Zhang R, Liu X, Ren Y, Liu Z, Zhang X, Cheng W & Hua ZC. Regulation of T lymphocyte activation by microRNA-21. Molecular Immunology 2014 59 163171. (doi:10.1016/j.molimm.2014.02.004)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 77

    Lu TX, Hartner J, Lim EJ, Fabry V, Mingler MK, Cole ET, Orkin SH, Aronow BJ & Rothenberg ME. MicroRNA-21 limits in vivo immune response-mediated activation of the IL-12/IFN-gamma pathway, Th1 polarization, and the severity of delayed-type hypersensitivity. Journal of Immunology 2011 187 33623373. (doi:10.4049/jimmunol.1101235)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 78

    Rouas R, Fayyad-Kazan H, El Zein N, Lewalle P, Rothe F, Simion A, Akl H, Mourtada M, El Rifai M, Burny A et al.. Human natural Treg microRNA signature: role of microRNA-31 and microRNA-21 in FOXP3 expression. European Journal of Immunology 2009 39 16081618. (doi:10.1002/eji.200838509)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 79

    Sheedy FJ. Turning 21: induction of miR-21 as a key switch in the inflammatory response. Frontiers in Immunology 2015 6 19.

  • 80

    Sheedy FJ, Palsson-McDermott E, Hennessy EJ, Martin C, O'Leary JJ, Ruan Q, Johnson DS, Chen Y & O'Neill LA. Negative regulation of TLR4 via targeting of the proinflammatory tumor suppressor PDCD4 by the microRNA miR-21. Nature Immunology 2010 11 141147. (doi:10.1038/ni.1828)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 81

    Pan W, Zhu S, Yuan M, Cui H, Wang L, Luo X, Li J, Zhou H, Tang Y & Shen N. MicroRNA-21 and microRNA-148a contribute to DNA hypomethylation in lupus CD4+ T cells by directly and indirectly targeting DNA methyltransferase 1. Open Journal of Immunology 2010 184 67736781. (doi:10.4049/jimmunol.0904060)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 82

    Churov AV, Oleinik EK & Knip M. MicroRNAs in rheumatoid arthritis: altered expression and diagnostic potential. Autoimmunity Reviews 2015 14 10291037. (doi:10.1016/j.autrev.2015.07.005)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 83

    Ventriglia G, Nigi L, Sebastiani G & Dotta F. MicroRNAs: novel players in the dialogue between pancreatic islets and immune system in autoimmune diabetes. BioMed Research International 2015 2015 749734.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 84

    Ruan Q, Wang T, Kameswaran V, Wei Q, Johnson DS, Matschinsky F, Shi W & Chen YH. The microRNA-21-PDCD4 axis prevents type 1 diabetes by blocking pancreatic beta cell death. PNAS 2011 108 1203012035. (doi:10.1073/pnas.1101450108)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 85

    Backe MB, Novotny GW, Christensen DP, Grunnet LG & Mandrup-Poulsen T. Altering beta-cell number through stable alteration of miR-21 and miR-34a expression. Islets 2014 6 e27754. (doi:10.4161/isl.27754)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 86

    Gonzalez-Martin A, Adams BD, Lai M, Shepherd J, Salvador-Bernaldez M, Salvador JM, Lu J, Nemazee D & Xiao C. The microRNA miR-148a functions as a critical regulator of B cell tolerance and autoimmunity. Nature Immunology 2016 17 433440. (doi:10.1038/ni.3385)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 87

    Melkman-Zehavi T, Oren R, Kredo-Russo S, Shapira T, Mandelbaum AD, Rivkin N, Nir T, Lennox KA, Behlke MA, Dor Y et al.. miRNAs control insulin content in pancreatic beta-cells via downregulation of transcriptional repressors. EMBO Journal 2011 30 835845. (doi:10.1038/emboj.2010.361)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 88

    Seoudi AM, Lashine YA & Abdelaziz AI. MicroRNA-181a - a tale of discrepancies. Expert Reviews in Molecular Medicine 2012 14 e5. (doi:10.1017/S1462399411002122)

  • 89

    Cichocki F, Felices M, McCullar V, Presnell SR, Al-Attar A, Lutz CT & Miller JS. Cutting edge: microRNA-181 promotes human NK cell development by regulating Notch signaling. Journal of Immunology 2011 187 61716175. (doi:10.4049/jimmunol.1100835)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 90

    Li QJ, Chau J, Ebert PJ, Sylvester G, Min H, Liu G, Braich R, Manoharan M, Soutschek J, Skare P et al.. miR-181a is an intrinsic modulator of T cell sensitivity and selection. Cell 2007 129 147161. (doi:10.1016/j.cell.2007.03.008)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 91

    Zhang J, Jima DD, Jacobs C, Fischer R, Gottwein E, Huang G, Lugar PL, Lagoo AS, Rizzieri DA, Friedman DR et al.. Patterns of microRNA expression characterize stages of human B-cell differentiation. Blood 2009 113 45864594. (doi:10.1182/blood-2008-09-178186)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 92

    Xie W, Li Z, Li M, Xu N & Zhang Y. miR-181a and inflammation: miRNA homeostasis response to inflammatory stimuli in vivo. Biochemical and Biophysical Research Communications 2013 430 647652. (doi:10.1016/j.bbrc.2012.11.097)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 93

    Xie W, Li M, Xu N, Lv Q, Huang N, He J & Zhang Y. MiR-181a regulates inflammation responses in monocytes and macrophages. PLoS ONE 2013 8 e58639. (doi:10.1371/journal.pone.0058639)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 94

    Magenta A, Greco S, Gaetano C & Martelli F. Oxidative stress and microRNAs in vascular diseases. International Journal of Molecular Sciences 2013 14 1731917346. (doi:10.3390/ijms140917319)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 95

    Devlin C, Greco S, Martelli F & Ivan M. miR-210: more than a silent player in hypoxia. IUBMB Life 2011 63 94100.

  • 96

    Kulshreshtha R, Ferracin M, Wojcik SE, Garzon R, Alder H, Agosto-Perez FJ, Davuluri R, Liu CG, Croce CM, Negrini M et al.. A microRNA signature of hypoxia. Molecular and Cellular Biology 2007 27 18591867. (doi:10.1128/MCB.01395-06)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 97

    Greco S, Gaetano C & Martelli F. HypoxamiR regulation and function in ischemic cardiovascular diseases. Antioxidants and Redox Signaling 2014 21 12021219. (doi:10.1089/ars.2013.5403)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 98

    Lu J, Xie F, Geng L, Shen W, Sui C & Yang J. Potential role of microRNA-210 as biomarker in human cancers detection: a meta-analysis. BioMed Research International 2015 2015 303987.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 99

    Poy MN, Hausser J, Trajkovski M, Braun M, Collins S, Rorsman P, Zavolan M & Stoffel M. miR-375 maintains normal pancreatic alpha- and beta-cell mass. PNAS 2009 106 58135818. (doi:10.1073/pnas.0810550106)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 100

    Li X. MiR-375, a microRNA related to diabetes. Gene 2014 533 14. (doi:10.1016/j.gene.2013.09.105)

  • 101

    Samandari N, Mirza AH, Nielsen LB, Kaur S, Hougaard P, Fredheim S, Mortensen HB & Pociot F. Circulating microRNA levels predict residual beta cell function and glycaemic control in children with type 1 diabetes mellitus. Diabetologia 2017 60 354363. (doi:10.1007/s00125-016-4156-4)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 102

    Zhu Y, You W, Wang H, Li Y, Qiao N, Shi Y, Zhang C, Bleich D & Han X. MicroRNA-24/MODY gene regulatory pathway mediates pancreatic beta-cell dysfunction. Diabetes 2013 62 31943206. (doi:10.2337/db13-0151)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 103

    Lu LF, Boldin MP, Chaudhry A, Lin LL, Taganov KD, Hanada T, Yoshimura A, Baltimore D & Rudensky AY. Function of miR-146a in controlling Treg cell-mediated regulation of Th1 responses. Cell 2010 142 914929. (doi:10.1016/j.cell.2010.08.012)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 104

    Rusca N & Monticelli S. MiR-146a in immunity and disease. Molecular Biology International 2011 2011 437301.

  • 105

    Taganov KD, Boldin MP, Chang KJ & Baltimore D. NF-kappaB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. PNAS 2006 103 1248112486. (doi:10.1073/pnas.0605298103)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 106

    Motawi TK, Rizk SM, Ibrahim TM & Ibrahim IA. Circulating microRNAs, miR-92a, miR-100 and miR-143, as non-invasive biomarkers for bladder cancer diagnosis. Cell Biochemistry and Function 2016 34 142148. (doi:10.1002/cbf.3171)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 107

    Wang M, Ren D, Guo W, Wang Z, Huang S, Du H, Song L & Peng X. Loss of miR-100 enhances migration, invasion, epithelial-mesenchymal transition and stemness properties in prostate cancer cells through targeting Argonaute 2. International Journal of Oncology 2014 45 362372. (doi:10.3892/ijo.2014.2413)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 108

    Gong Y, He T, Yang L, Yang G, Chen Y & Zhang X. The role of miR-100 in regulating apoptosis of breast cancer cells. Scientific Reports 2015 5 11650. (doi:10.1038/srep11650)

  • 109

    Sylvius N, Bonne G, Straatman K, Reddy T, Gant TW & Shackleton S. MicroRNA expression profiling in patients with lamin A/C-associated muscular dystrophy. FASEB Journal 2011 25 39663978. (doi:10.1096/fj.11-182915)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 110

    Wang L, Su W, Du W, Xu Y, Wang L, Kong D, Han Z, Zheng G & Li Z. Gene and microRNA profiling of human induced pluripotent stem cell-derived endothelial cells. Stem Cell Reviews 2015 11 219227. (doi:10.1007/s12015-014-9582-4)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 111

    Ortega FJ, Moreno-Navarrete JM, Pardo G, Sabater M, Hummel M, Ferrer A, Rodriguez-Hermosa JI, Ruiz B, Ricart W, Peral B et al.. MiRNA expression profile of human subcutaneous adipose and during adipocyte differentiation. PLoS ONE 2010 5 e9022. (doi:10.1371/journal.pone.0009022)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 112

    Pek SL, Sum CF, Lin MX, Cheng AK, Wong MT, Lim SC & Tavintharan S. Circulating and visceral adipose miR-100 is down-regulated in patients with obesity and Type 2 diabetes. Molecular and Cellular Endocrinology 2016 427 112123. (doi:10.1016/j.mce.2016.03.010)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 113

    Fayyad-Kazan H, Bitar N, Najar M, Lewalle P, Fayyad-Kazan M, Badran R, Hamade E, Daher A, Hussein N, ElDirani R et al.. Circulating miR-150 and miR-342 in plasma are novel potential biomarkers for acute myeloid leukemia. Journal of Translational Medicine 2013 11 31. (doi:10.1186/1479-5876-11-31)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 114

    Van der Auwera I, Limame R, van Dam P, Vermeulen PB, Dirix LY & Van Laere SJ. Integrated miRNA and mRNA expression profiling of the inflammatory breast cancer subtype. British Journal of Cancer 2010 103 532541. (doi:10.1038/sj.bjc.6605787)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 115

    Ronchetti D, Lionetti M, Mosca L, Agnelli L, Andronache A, Fabris S, Deliliers GL & Neri A. An integrative genomic approach reveals coordinated expression of intronic miR-335, miR-342, and miR-561 with deregulated host genes in multiple myeloma. BMC Medical Genomics 2008 1 37.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 116

    Oger F, Gheeraert C, Mogilenko D, Benomar Y, Molendi-Coste O, Bouchaert E, Caron S, Dombrowicz D, Pattou F, Duez H et al.. Cell-specific dysregulation of microRNA expression in obese white adipose tissue. Journal of Clinical Endocrinology and Metabolism 2014 99 28212833. (doi:10.1210/jc.2013-4259)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 117

    Wang L, Xu L, Xu M, Liu G, Xing J, Sun C & Ding H. Obesity-associated miR-342-3p promotes adipogenesis of mesenchymal stem cells by suppressing ctbp2 and releasing C/EBPalpha from CtBP2 binding. Cellular Physiology and Biochemistry 2015 35 22852298. (doi:10.1159/000374032)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 118

    Eissa S, Matboli M & Bekhet MM. Clinical verification of a novel urinary microRNA panal: 133b, -342 and -30 as biomarkers for diabetic nephropathy identified by bioinformatics analysis. Biomedicine and Pharmacotherapy 2016 83 9299. (doi:10.1016/j.biopha.2016.06.018)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 119

    Collares CV, Evangelista AF, Xavier DJ, Rassi DM, Arns T, Foss-Freitas MC, Foss MC, Puthier D, Sakamoto-Hojo ET, Passos GA et al.. Identifying common and specific microRNAs expressed in peripheral blood mononuclear cell of type 1, type 2, and gestational diabetes mellitus patients. BMC Research Notes 2013 6 491.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 120

    Kaviani M, Azarpira N, Karimi MH & Al-Abdullah I. The role of microRNAs in islet beta-cell development. Cell Biology International 2016 40 12481255. (doi:10.1002/cbin.10691)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 121

    Schmidt WM, Spiel AO, Jilma B, Wolzt M & Muller M. In vivo profile of the human leukocyte microRNA response to endotoxemia. Biochemical and Biophysical Research Communications 2009 380 437441. (doi:10.1016/j.bbrc.2008.12.190)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 122

    Debernardi S, Massat NJ, Radon TP, Sangaralingam A, Banissi A, Ennis DP, Dowe T, Chelala C, Pereira SP, Kocher HM et al.. Noninvasive urinary miRNA biomarkers for early detection of pancreatic adenocarcinoma. American Journal of Cancer Research 2015 5 34553466.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 123

    Yan B & Wang Z. Long noncoding RNA: its physiological and pathological roles. DNA and Cell Biology 2012 31 (Supplement 1) S34S41.

  • 124

    Fawzy IO, Hamza MT, Hosny KA, Esmat G, El Tayebi HM & Abdelaziz AI. miR-1275: a single microRNA that targets the three IGF2-mRNA-binding proteins hindering tumor growth in hepatocellular carcinoma. FEBS Letters 2015 589 22572265. (doi:10.1016/j.febslet.2015.06.038)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 125

    Pena-Chilet M, Martinez MT, Perez-Fidalgo JA, Peiro-Chova L, Oltra SS, Tormo E, Alonso-Yuste E, Martinez-Delgado B, Eroles P, Climent J et al.. MicroRNA profile in very young women with breast cancer. BMC Cancer 2014 14 529.

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 126

    Bezman NA, Chakraborty T, Bender T & Lanier LL. miR-150 regulates the development of NK and iNKT cells. Journal of Experimental Medicine 2011 208 27172731. (doi:10.1084/jem.20111386)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • 127

    Hong G, Zhang W, Li H, Shen X & Guo Z. Separate enrichment analysis of pathways for up- and downregulated genes. Journal of the Royal Society Interface 2014 11 20130950.

    • PubMed
    • Search Google Scholar
    • Export Citation

 

  • Collapse
  • Expand
  • Flowchart illustrating the search strategy used to identify association studies of miRNAs expression with type 1 diabetes for inclusion in the systematic review.

  • miRNA expression analyzed in at least two studies considering their expression profile in all tissues analyzed independently of the species (A). miRNA expression profile between the species (B). miRNA expression profile in tissues related to T1DM pathogenesis (C).

  • KEGG pathway functional annotation of the differentially miRNAs expressed in PBMCs (A), serum/plasma (B) and pancreas (C). Enrichment scores corresponding to each pathway are displayed as number of targets.

  • Schematic diagram of the selected miRNA-mRNA interaction networks involved in T cell receptor signaling pathway. The network was built based on KEGG pathway map (KEGG: hsa04660). The miRNAs are indicated by rhombus. The predicted miRNA-mRNA interactions are indicated by dotted lines and the thick lines indicate validated miRNA-mRNA interactions.

  • Schematic diagram of the selected miRNA-mRNA interaction networks involved in Jak-STAT signaling pathway. The network was built based on KEGG pathway map (KEGG: hsa04630). The miRNAs are indicated by rhombus. The predicted miRNA-mRNA interactions are indicated by dotted lines and the thick lines indicate validated miRNA-mRNA interactions.