Next Article in Journal
Investigating Autism-Related Symptoms in Children with Prader-Willi Syndrome: A Case Study
Previous Article in Journal
Short-Term Local Adaptation of Historical Common Bean (Phaseolus vulgaris L.) Varieties and Implications for In Situ Management of Bean Diversity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

A Systematic Study of Dysregulated MicroRNA in Type 2 Diabetes Mellitus

1
Institute of Medical Systems Biology, Guangdong Medical University, Dongguan 523808, China
2
Department of Epidemiology and Medical Statistics, Guangdong Medical University, Dongguan 523808, China
3
Institute for Systems Biology, Seattle, WC 98109, USA
4
Department of endocrinology, Zhongshan Chen Xinghai Hospital Affiliated to Guangdong Medical University, Zhongshan 528415, China
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2017, 18(3), 456; https://doi.org/10.3390/ijms18030456
Submission received: 26 December 2016 / Revised: 9 February 2017 / Accepted: 14 February 2017 / Published: 28 February 2017
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)

Abstract

:
MicroRNAs (miRNAs) are small noncoding RNAs that modulate the cellular transcriptome at the post-transcriptional level. miRNA plays important roles in different disease manifestation, including type 2 diabetes mellitus (T2DM). Many studies have characterized the changes of miRNAs in T2DM, a complex systematic disease; however, few studies have integrated these findings and explored the functional effects of the dysregulated miRNAs identified. To investigate the involvement of miRNAs in T2DM, we obtained and analyzed all relevant studies published prior to 18 October 2016 from various literature databases. From 59 independent studies that met the inclusion criteria, we identified 158 dysregulated miRNAs in seven different major sample types. To understand the functional impact of these deregulated miRNAs, we performed targets prediction and pathway enrichment analysis. Results from our analysis suggested that the altered miRNAs are involved in the core processes associated with T2DM, such as carbohydrate and lipid metabolisms, insulin signaling pathway and the adipocytokine signaling pathway. This systematic survey of dysregulated miRNAs provides molecular insights on the effect of deregulated miRNAs in different tissues during the development of diabetes. Some of these miRNAs and their mRNA targets may have diagnostic and/or therapeutic utilities in T2DM.

1. Introduction

Diabetes mellitus has become a global epidemic. It affects roughly 8.3% of the population, with 90% of these patients suffering from type 2 diabetes mellitus (T2DM) [1]. T2DM is a complex disease caused by both genetic and environmental factors [2]. It often occurs in middle-aged adults with chronic hyperglycemia. Lifestyle changes are often used to prevent or manage the progression of the disease. However, the pancreatic β-cell dysfunction usually progresses and results in insulin resistance and/or relative insulin insufficiency in key metabolic organs, such as skeletal muscle, liver and adipose tissues [3]. Long-term de-regulation of energy metabolism in T2DM leads to systemic complications, such as CAD (coronary artery disease), stroke, peripheral arterial disease [4], CKD (chronic kidney disease) [5], microvascular diseases (retinopathy) [6] and various other conditions, such as cancers [7,8,9,10,11].
MicroRNAs (miRNAs) are 17–21 nt regulatory RNAs that suppress the translation and stability of mRNA through imperfect base pairing in the 3′ untranslated region of its mRNA targets [12]. There are over 2500 human miRNAs deposited in the latest release of miRBase (available online: www.mirbase.org). It has been estimated that more than 60% of protein coding transcripts are regulated by one or more miRNAs [13]. A number of dysregulated miRNAs from insulin-sensitive organs, including skeletal muscle, white adipose tissue and insulin-producing pancreatic β-cells, has been linked to processes related to diabetes, such as insulin secretion, pancreatic β-cells and adipocyte differentiation [14]. Systematic studies on the involvement of these aberrant miRNAs in T2DM will enhance our understanding of the molecular processes involved in the development of T2DM and facilitate the development of new disease management tools, therapeutic targets, diagnostic markers and preventive measures for T2DM.
Many studies have characterized the changes and involvement of miRNA in different cell types and organs during the development of T2DM [15,16,17,18,19,20,21,22]. Due to the complexity of T2DM pathology, a thorough systematic comparison, integration and investigation on the functional implications of these dysregulated miRNAs is still needed. We searched all reports related to miRNA and T2DM, downloaded and analyzed the data that met our inclusion criteria and identified a set of dysregulated miRNAs from 59 independent studies. Based on their predicted and validated targets, we conducted pathway enrichment analysis and built mRNA-miRNA interaction networks that help our understanding of the effects of these aberrantly expressed miRNAs in T2DM. The identified miRNA meta-signature and perturbed networks associated with T2DM will facilitate our understanding of the underlying biological processes in the development of T2DM.

2. Results

2.1. Study Characteristics

The schema of data identification and selection is shown in Figure 1. According to the search criteria outlined, a total of 7329 publications were identified through initial searches of various databases using different combinations of terms as described in the Materials and Methods section. During the data extraction step, 2794 duplicated records were removed, and an additional 2664 studies were also excluded due to either review, meta-analysis, letter, news or editorial articles (n = 405); thesis or conference reports/abstracts (n = 268); unrelated studies (n = 1973); non-English or Chinese publications (n = 18). After detailed evaluation, another 71 studies were removed due to not using human samples (n = 52), being T1DM studies (n = 16) and studies without specify T1 or T2DM (n = 3). At the end, the results from 59 published studies were retrieved from public databases, which include a total of 2671 T2DM patients and 2573 healthy controls (Figure 1).
The main characteristics extracted from various datasets are summarized in Table 1, which includes the number of patients investigated, the measurement platform, type of sample used and the identity of miRNAs in the study [23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81]. Comparing results from multiple tissues provides an overall view of the impact of miRNAs in T2DM pathology. Before analysis, we updated the miRNA name according to the latest nomenclature provided by miRBase. Three miRNAs, miR-463, miR-768 and miR-801, were removed from analysis due to these miRNAs no longer being recognized as miRNAs. Several major sample types were involved in the 59 studies, including adipose tissue, islet, skeletal muscle, whole blood, PBMC (peripheral blood mononuclear cell), serum and plasma. Some miRNAs showed inconsistent changes among different studies within the same sample type, and these miRNAs are indicated in Table 2 and Table 3. For studies that included profiling, as well as validation components, we selected validated miRNAs for further study; for example in the study by Karolina et al. [26], eight qRT-PCR validated differentially expressed miRNAs (miR-29a, -144, -150, -192, -320a, -30d, -146a and miR-182) identified by microarray profiling results were included in our analysis (Table 1).

2.2. Quality Assessment

The quality assessment scores listed in Table 1 were conducted in accordance with the QUADOMICS tool. None of the studies were classified as “low quality”, fulfilling fewer than 12 of the 16 criteria. Since none of the studies were conducted as blind tests (without knowledge of the reference standard and patient sample), all studies failed Criteria 12 and 13 of the QUADOMICS tool.

2.3. Alterations of Specific miRNAs’ Levels among Different Tissues in T2DM

From 59 independent reports, a total of 158 dysregulated miRNAs were identified (Table 1 and Table 2). According to the sample type used, results from most of the studies can be divided into seven major groups (adipose, islet, skeletal muscle, whole blood, PBMC, plasma and serum).
Skeletal muscle had the most number of affected miRNAs: 29 with decreased concentration and 31 with increased concentration in skeletal muscle samples from T2DM patients compared to healthy controls (Table 2). As expected, there are more aberrantly-expressed miRNAs in common among whole blood, PBMC, serum and plasma (Table 3). This is especially true between serum and plasma, since these are prepared from a common source. Between serum and plasma, there are 16 affected miRNAs in common; however, the concentration changes between the two may be different. For example, the levels of miR-191-5p and miR-192-5p are decreased in both serum and plasma in T2DM patients compared to healthy controls, but the concentration changes of miR-29b-3p and miR-320a are opposite between serum and plasma (Table 2 and Table 3). The concentration of miR-375, a highly enriched miRNA in pancreatic tissue, is increased in the islet of T2DM patients, as well as in whole blood, serum and plasma. There are also some common changes between skeletal muscle and whole blood, for example miR-100-5p, miR-126-3p and miR-144-3p. This suggests some of the deregulated circulating cell-free miRNAs might reflect T2DM-associated pathologies in different tissues.

2.4. Perturbed Pathways Mediated by Dysregulated miRNAs

Using the list of dysregulated miRNAs in each sample type identified from the literature, we performed pathway enrichment analysis based on validated and predicted miRNA targets (Table S1). Some of these pathways are known to be involved in T2DM; for example, various pathways associated with metabolic processes (carbohydrate and lipid metabolism), cell-cell communications (focal adhesion, tight junction), cell growth and death (apoptosis and cell cycle), signal transduction (JAK-STAT, MAPK, TGF-β, Wnt, cytokine-cytokine receptor interaction and neurotrophin signaling), immune response (leukocyte transendothelial migration, T-cell receptor signaling, Nod-like receptor signaling, Toll-like receptor signaling and chemokine signaling), insulin signaling and type 2 diabetes signaling.
Based on the KEGG (Kyoto Encyclopedia of Genes and Genomes) pathway map and affected miRNAs from each sample type, we constructed miRNA-mRNA interaction networks for three key T2DM-associated pathways, T2DM signaling, insulin signaling and adipocytokine pathways in pancreatic islet (Figure 2) and adipose tissues (Figure 3), to better illustrate the complex interactions between mRNAs and affected miRNAs in key tissues associated with the manifestation of T2DM. Since adipose tissue is one of the key organs involved in the development of T2DM, we included the expression level changes of protein coding mRNAs involved in the pathways from a visceral adipose tissue expression profiling dataset obtained from the public domain (GSE16415, available online: https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE16415). The dataset compared adipose tissues gene expression differences between women with type 2 diabetes with age and BMI matched normal women. The omental biopsies samples were from five diabetic and five control females undergoing cholecystectomy. All subjects used in the study were older than 50 years with BMI values greater than 30. In both pancreatic islet and adipose tissues, the dysregulated miRNAs target several well-known T2DM-associated genes. For example, the adiponectin receptor 2 (ADIPOR2) in the adipocytokine pathway is targeted by miR-375 and miR-136-5p in pancreatic islet and miR-146a in adipose tissues. The key enzymes in lipid biosynthesis and fatty acid degradation, acyl-coA synthetase long-chain family members (ACSL1, 2, 3 and 4), are also targeted by multiple miRNAs, including miR-17-5p, -130b-3p, -134-5p and -181a-5p in adipose tissue and miR-7-5p, -369-3p, -495-3p and -655-3p in islets.

3. Discussion

Even though the etiology of T2DM is yet to be fully understood, insulin resistance and pancreatic β-cell dysfunction are the two major causes for various T2DM-associated clinical phenotypes [3], and these conditions usually occur years before the clinical diagnosis of T2DM [82]. Many studies have suggested that miRNA is one of the key players in the pathogenesis of T2DM and associated complications, such as cardiovascular conditions and kidney dysfunction [83,84,85]. The current analysis identified 158 T2DM-associated dysregulated miRNAs in seven different sample types. Pathway enrichment analysis revealed that a number of T2DM relevant pathways is targeted by these dysregulated miRNAs. For example, the disturbed insulin signaling pathway might be one of the key reasons for the glucose and lipid metabolism impairments, which cause insulin insufficiency in skeletal muscle, liver and adipose tissues [86]. The other interesting finding is the observation that some kidney and cardiovascular function-related pathways were also affected by these dysregulated miRNAs (Table S1). For example, the vasopressin-regulated water reabsorption pathway is enriched in genes that are targeted by dysregulated miRNAs in adipose tissues. The other example is the association between various cardiovascular disease processes and dysregulated miRNAs in serum.
It is well-known that hyperglycemia, hyperlipidemia and islet β cell dysfunction are associated with the onset of T2DM [82]. Our analysis also shows that a number of aberrant miRNAs are involved in pathways associated with energy metabolism, insulin signaling, type 2 diabetes signaling and adipocytokine signaling (Table S1). Among the 19 dysregulated miRNAs in pancreatic islets, miR-375 is probably the best characterized miRNA that has been shown to be involved in both insulin secretion, release and glucose homeostasis [39,87]. Increased miR-375 levels in the islets inhibit PDPK1 (3-phosphoinositide-dependent protein kinase 1) expression [31], an important component of the PI3k/protein kinase B signal cascade (Figure 2B, insulin signaling pathway). Loss of PDPK1 in β cells results in progressive hyperglycemia due to reductions in the number and size of β cells, as well as defective β cell function [88]. In addition to miR-375, other dysregulated miRNAs in the islets (such as miR-7-5p, -369-5p, -129-3p,-136-5p, -187-3p, -589-5p, -224-5p, -655-3p, -495-3p) affect the expression of IRS1, IRS2, AKT1, PPARA, MAPK9, MAPK10, STAT3, PPKAG2, ACSL3 and ACSL4, which are important genes involved in insulin signaling and type 2 diabetes pathways [89,90] (Figure 2A–C), and have been implicated in various events associated with islet β cell development and glucose-stimulated insulin secretion (GSIS) [23,36,90,91].
Adipose tissues play a key role in maintaining the homeostasis of the body’s energy metabolism by releasing an array of hormones and cytokines: adipocytokines. The current study revealed that the dysregulated miRNAs play important roles in glucose, as well as lipid metabolism (Table S1) [29,50,92]. These miRNAs, including miR-17-5p, -155-5p, -125b-5p, -30e-5p, -27a-5p, -221-3p, -199a-5p, -130b-3p, -181a-5p, -29a, -29b, interact with multiple transcription factors, such as PPARs (peroxisome proliferator activated receptors), including PPARG, also known as PPARγ, and adipocyte-enriched genes (GLUT4 (also known as SLC2A4), SOCS1, SOCS3, GRB2, INSR and PPARG), to regulate many aspects of the lipid and glucose metabolisms [90,93]. Some of these miRNAs (miR-130b-3p, -140-5p, -147a, -199a-5p, -27b, -221-3p and -30e-5p) have also been implicated in the regulation of adipogenesis through PPARG [94,95]. miR-181a-5p, -132-3p, -199a-5p, -24-3p and -126-3p also contribute to an increased glucose uptake by affecting PI3K/AKT activity, where the PI3K/ATK signaling is essential for SLC2A4 (GLUT4) translocation to the cell membrane. In addition, miR-130b-3p, -132-3p, -181a-5p, -199a-5p and -221-3p can directly target PPKAs (AMP-activated protein kinase) to affect energy metabolism in various signal transduction pathways, including adipocytokine and insulin signaling pathways [90,93] (Figure 3A–C). Dysfunction of adipose tissues may induce inflammation and insulin resistance by releasing pro-inflammatory cytokines, such as TNFA (tumor necrosis factor alpha) and IL-6 (interleukin 6), as well as energy regulating hormones including LEP (leptin), ADIPOQ (adiponectin), RETN (resistin) and ITLN1 (intelectin 1) [96]. The aberrant expression of these genes/proteins will further the manifestation of T2DM.
Skeletal muscle is one of the main tissues involved in glucose uptake and metabolism. Insulin resistance in skeletal muscle is an early sign of T2DM and is a risk factor for cardiovascular diseases common among patients with T2DM [49,86]. Studies have indicated that several miRNAs (miR-17-5p, -24, -126, -125b-5p, -130b-3p, -132-3p, -181a-3p, -181a-5p, -197-3p and -221-3p) can target and affect MAPK protein (mitogen-activated protein kinases) levels under high glucose condition [49,97,98]. MAPKs can then affect the level of MEF2 (myocyte enhancer factor 2), which is a key muscle-specific transcription factor to regulate the transcription of the insulin-responsive SLC2As (solute carrier family 2 transporters, which facilitate glucose transport), including GLUT4 (glucose transporter 4, SLC2A4) [49,93].
There are a number of studies investigating the relationship of miRNA and diabetes based on animal models and cell line systems [99,100,101,102,103]. These studies provide valuable information, but in the current analysis, we did not include these studies because the model systems, either in vivo or in vitro, can only reflect very limited aspects of the T2DM pathology. The physiopathological difference between human and model organisms will also generate a very different set of dysregulated genes and miRNAs [104,105]. Moreover, even the same miRNA may interact with very different sets of targets due to a difference in both mRNA and miRNA sequences between human and animal models. For example, miR-375 has an identical mature sequence between human (hsa-miR-375) and mouse (mmu-miR-375), but the peroxisome proliferative activated receptor, gamma, coactivator 1 alpha (PPARGC1A), a transcriptional coactivator, interacts with PPARgamma (PPARG) to regulate genes involved in energy metabolism and is a target of mouse miR-375, but not human. Therefore, in the current study, we only selected studies from human patient samples in order to investigate the functional involvement of miRNAs in human T2DM pathology. These findings may provide a more accurate and more realistic view of the effect of miRNAs in diabetes.
Some of the miRNA-mRNA interactions in Figure 2 and Figure 3 are based on results from various miRNA target prediction programs. We used miRSystem, an integrated miRNA function prediction website based on multiple miRNA target prediction algorithms, to facilitate the summarization of miRNA-mRNA interaction [106]. There are other similar websites, such as miRGator (available online: www.mirgator.kobic.re.kr/) [107] and miRPathDB (available online: mpd.bioinf.uni-sb.de/) [108]. All of these tools provide a similar function and deliver comparable results.
In the past few years, there have been a number of reports revealing disease-associated concentration changes of specific miRNA in circulation, such as miR-375 in patients with T2DM (Table 3) [31,34,39,75]. However, the idea of using circulating miRNA to reflect pathological changes in specific tissues and/or as a reliable biomarker for disease diagnosis faces numerous challenges. These difficulties are due to many issues, including the accuracy and consistency of different miRNA measurement platforms, low concentration of circulating RNA in samples and the high sequence similarity of miRNA family members. However, the biggest challenge of circulating RNA-based biomarker discovery probably is due to the fact that the spectrum of circulating RNA is the sum of many different tissues and cell types in the body. How to identify changes from specific tissues or disease stages from very complex and noisy data is difficult. Using tissue or disease-enriched RNAs (miRNAs or mRNAs), like miR-375, which is enriched in pancreatic tissue, may provide a useful approach for circulating RNA-based biomarker discovery. In addition, using animal models that allow frequent and detailed monitoring of disease-associated RNA profile changes in tissue and body fluid samples may also provide insights on the relationship of RNA between the two compartments.
In conclusion, we present a systematic analysis by integrating various T2DM-related miRNA datasets from the literature. These dysregulated miRNAs affect various metabolic and signal transduction pathways associated with T2DM and may play important roles in the development of diabetes. The pathway analysis results also provide possible links to kidney and cardiovascular complications associated the T2DM. However, many other factors, both endogenous and exogenous, may influence the spectrum of miRNA in tissues and circulation. Recent findings suggest that diet, a key component in T2DM disease management, affects the spectra of miRNA in various tissues [109,110]. For example, the consumption of milk resulted in significant increases of miRNA levels in plasma and PBMCs [111]. These findings suggested that individual’s diet and nutritional status can influence miRNA in tissues and circulation. Therefore, more studies with larger and well-defined populations are clearly needed to resolve this complex miRNA-mediated regulatory network. The dysregulated miRNAs and their interacting mRNA targets may provide new insights to the T2DM pathology and provide new disease monitoring and management tools.

4. Materials and Methods

4.1. Search Strategy and Eligibility of Relevant Studies

We scanned all relevant studies published prior to 18 October 2016 through a comprehensive search of records in PubMed, The Cochrane Library, the Chinese National Knowledge Infrastructure (CNKI) database (available online: http://www.cnki.net), Wanfang database (available online: http://www.wanfangdata.com), VIP database (VIP information/Chinese Scientific Journals database, available online: http://www.cqvip.com) and Google Scholar. The search was conducted by using combinations of the following key words: “miRNA” or “microRNA” and “diabetes” or “type 2 diabetes”. The online search was accompanied by checking cited references from the articles for potentially eligible reports. References of identified studies that met the inclusion criteria have been assessed by two independent reviewers. To ensure that relevant studies were not missed, searches in Gene Expression Omnibus (GEO, available online: www.ncbi.nlm.nih.gov/geo/) and Array Express (available online: www.ebi.ac.uk/arrayexpress) repositories were also performed.

4.2. Inclusion and Exclusion Criteria

All miRNA studies associated with T2DM were included in the current meta-analysis if they met all of the following criteria: (1) full text of original experimental articles based on miRNA expression profiling between diabetic patients and normal individuals; (2) studies based on human samples; and (3) original articles published in a language of either English or Chinese. The main exclusion criteria were as follows: (1) meta-analyses, series, abstract, commentary, review, letters and editorial; (2) duplicate data; (3) studies that compared different stages of diabetes, but did not include normal samples; and (4) studies based on cell lines or animal models.

4.3. Data Extraction

Data were extracted by two independent reviewers from all publications that met the inclusion criteria. The lists of miRNAs with statistically-significant expression changes were extracted from the publications. All miRNA names were standardized according to miRBase Version 21 (Table 1 and Table 2). Any discrepancies were resolved by consensus or in consultation with a third reviewer. Information extracted from eligible publications includes: first author’s last name, year of publication, country of origin, population ethnicity, disease type, measurement platform, source of cases and control groups, number of samples and number of miRNAs measured. All relevant information was compiled in Table 1.

4.4. Quality Assessment and Statistical Analysis

We assessed the quality of the data using the QUADAS2 (Quality Assessment of Diagnostic Accuracy Studies 2) checklist. The categories in the scoring system used for assessing study quality are summarized in Table 1. Quality scores ranged from 0–10, and studies were scored as “good” if the score was 8–10, “fair” if the score was 5–7 and “poor” if the score was <4. The dysregulated miRNAs were grouped into seven groups based on the different sample sources used in the studies (adipose, islet, skeletal muscle, whole blood, PBMC, plasma, serum).

4.5. miRNA Target Prediction and Pathway Enrichment Analysis

To identify the functional involvement of miRNAs in the progression of T2DM, we used miRNA target prediction algorithms and information from validated miRNA target databases to generate the list of putative miRNA targets. The list of putative interacting targets for the miRNAs was generated using the miRSystem (available online: http://mirsystem.cgm.ntu.edu.tw/index.php) web server, which provides information from seven miRNA target prediction algorithms, DIANA-microT, miRanda, mirBridge, PicTar, PITA, RNA22 and TargetScan, and two experimentally-validated miRNA target databases, TarBase and miRecords [106]. To increase the reliability of target prediction, the targets that have been predicted by more than 5 different algorithms were then used for pathway enrichment analysis (available online: http://david.abcc.ncifcrf.gov/home.jsp). Cytoscape (available online: http://www.cytoscape.org/) and KEGG pathway maps (available online: http://www.genome.jp/kegg/pathway.html) were used to generate miRNA-mRNA interaction networks. We also included the mRNA expression level changes from visceral adipose tissue expression profiling data from the public domain (GSE16415).

Supplementary Materials

Supplementary materials can be found at www.mdpi.com/1422-0067/18/2/456/s1.

Acknowledgments

The work is supported by the Talents Recruitment Grant of “Yangfan Plan” of Guangdong Province, China (No. 201433005) (Yuqing He), the Science and Technology Program of Guangdong (2013B051000080) (Yuqing He), the International Science & Technology Cooperation Program of Dongguan (2013508152005) (Yuqing He), the Science & Technology Program of Dongguan (2014108101055) (Yuqing He), the Innovation Project from the Education Department of Guangdong Province (2015KTSCX048y) (Haibing Yu), the National Science Foundation of China (81273166) (Yuanlin Ding) and the International Science & Technology Cooperation Program of Dongguan (20135081520017) (Yuanlin Ding), as well as support by the NIH (U01HL126496-02) (Kai Wang).

Author Contributions

All authors reviewed and approved the final version of the manuscript. Yuqing He, Kai Wang and Yuanlin Ding conducted the initial literature review, contributed to the study design and drafted the article. Juanjuan Lin, Biyu Liang, Hanwei Hang and Haibing Yu analyzed the literature. Taek-Kyun Kim analyzed and prepared the network.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

GSISglucose-stimulated insulin secretion
PDPK13-phosphoinositide dependent protein kinase 1
PPARGperoxisome proliferator-activated receptor gamma
MAPKmitogen-activated protein kinase
ADIPOR2adiponectin receptor 2
ACSLacyl-coA synthetase long-chain family members
TNFAtumor necrosis factor alpha
SLC2Assolute carrier family 2, which facilitates glucose transport
PPKAsAMP-activated protein kinase
MEF2myocyte enhancer factor 2
T2DMtype 2 diabetes mellitus
miRNAmicroRNA

References

  1. Rogers, E.A.; Fine, S.; Handley, M.A.; Davis, H.; Kass, J.; Schillinger, D. Development and early implementation of the bigger picture, a youth-targeted public health literacy campaign to prevent type 2 diabetes. J. Health Commun. 2014, 19, 144–160. [Google Scholar] [CrossRef] [PubMed]
  2. Jin, W.; Patti, M. Genetic determinants and molecular pathways in the pathogenesis of type 2 diabetes. Clin. Sci. 2009, 116, 99–111. [Google Scholar] [CrossRef] [PubMed]
  3. Ribbing, J.; Hamrén, B.; Svensson, M.K.; Karlsson, M.O. A model for glucose, insulin, and β-cell dynamics in subjects with insulin resistance and patients with type 2 diabetes. J. Clin. Pharmacol. 2010, 50, 861–872. [Google Scholar] [CrossRef] [PubMed]
  4. Pedicino, D.; Liuzzo, G.; Trotta, F.; Giglio, A.F.; Giubilato, S.; Martini, F.; Zaccardi, F.; Scavone, G.; Previtero, M.; Massaro, G. Adaptive immunity, inflammation, and cardiovascular complications in type 1 and type 2 diabetes mellitus. J. Diabetes Res. 2013, 2013, 184258. [Google Scholar] [CrossRef] [PubMed]
  5. Bello, N.A.; Pfeffer, M.A.; Skali, H.; McGill, J.B.; Rossert, J.; Olson, K.A.; Weinrauch, L.; Cooper, M.E.; de Zeeuw, D.; Rossing, P. Retinopathy and clinical outcomes in patients with type 2 diabetes mellitus, chronic kidney disease, and anemia. BMJ Open Diabetes Res. Care 2014, 2, e000011. [Google Scholar] [CrossRef] [PubMed]
  6. Paneni, F.; Beckman, J.A.; Creager, M.A.; Cosentino, F. Diabetes and vascular disease: Pathophysiology, clinical consequences, and medical therapy: Part I. Eur. Heart J. 2013, 34, 2436–2443. [Google Scholar] [CrossRef] [PubMed]
  7. Shlomai, G.; Neel, B.; LeRoith, D.; Gallagher, E.J. Type 2 diabetes mellitus and cancer: The role of pharmacotherapy. J. Clin. Oncol. 2016, 34, 4261–4269. [Google Scholar] [PubMed]
  8. Klil-Drori, A.J.; Azoulay, L.; Pollak, M.N. Cancer, obesity, diabetes, and antidiabetic drugs: Is the fog clearing? Nat. Rev. Clin. Oncol. 2017, 14, 85–99. [Google Scholar] [CrossRef] [PubMed]
  9. Liu, H.T.; Xu, Y.T.; Li, H.Y.; Zhao, J.; Zhai, H.Y.; Chen, Y. Loss of microRNA-145 expression is involved in the development and prognosis of breast cancer complicated by type 2 diabetes mellitus. Int. J. Biol. Markers 2016, 31, e368–e374. [Google Scholar] [CrossRef] [PubMed]
  10. Wang, Y.; Wang, B.; Yan, S.; Shen, F.; Cao, H.; Fan, J.; Zhang, R.; Gu, J. Type 2 diabetes and gender differences in liver cancer by considering different confounding factors: A meta-analysis of cohort studies. Ann. Epidemiol. 2016, 26, 764–772. [Google Scholar] [CrossRef] [PubMed]
  11. Sayyid, R.K.; Fleshner, N.E. Diabetes mellitus type 2: A driving force for urological complications. Trends Endocrinol. Metab. 2016, 27, 249–261. [Google Scholar] [CrossRef] [PubMed]
  12. Esteller, M. Non-coding RNAs in human disease. Nat. Rev. Genet. 2011, 12, 861–874. [Google Scholar] [CrossRef] [PubMed]
  13. Kellis, M.; Wold, B.; Snyder, M.P.; Bernstein, B.E.; Kundaje, A.; Marinov, G.K.; Ward, L.D.; Birney, E.; Crawford, G.E.; Dekker, J. Defining functional DNA elements in the human genome. Proc. Natl. Acad. Sci. USA 2014, 111, 6131–6138. [Google Scholar] [CrossRef] [PubMed]
  14. Eliasson, L.; Esguerra, J.L.S. Role of non-coding RNAs in pancreatic β-cell development and physiology. Acta Physiol. 2014, 211, 273–284. [Google Scholar] [CrossRef] [PubMed]
  15. Lee, M.J.; Park, D.H.; Kang, J.H. Exosomes as the source of biomarkers of metabolic diseases. Ann. Pediatr. Endocrinol. Metab. 2016, 21, 119–125. [Google Scholar] [CrossRef] [PubMed]
  16. Simpson, K.; Wonnacott, A.; Fraser, D.J.; Bowen, T. MicroRNAs in diabetic nephropathy: From biomarkers to therapy. Curr. Diabetes Rep. 2016, 16, 35. [Google Scholar] [CrossRef] [PubMed]
  17. Deiuliis, J.A. MicroRNAs as regulators of metabolic disease: Pathophysiologic significance and emerging role as biomarkers and therapeutics. Int. J. Obes. 2016, 40, 88–101. [Google Scholar] [CrossRef] [PubMed]
  18. Kato, M.; Natarajan, R. MicroRNAs in diabetic nephropathy: Functions, biomarkers, and therapeutic targets. Ann. N. Y. Acad. Sci. 2015, 1353, 72–88. [Google Scholar] [CrossRef] [PubMed]
  19. Price, N.L.; Ramirez, C.M.; Fernandez-Hernando, C. Relevance of microRNA in metabolic diseases. Crit. Rev. Clin. Lab. Sci. 2014, 51, 305–320. [Google Scholar] [CrossRef] [PubMed]
  20. Chen, H.; Lan, H.Y.; Roukos, D.H.; Cho, W.C. Application of microRNAs in diabetes mellitus. J. Endocrinol. 2014, 222, 1–10. [Google Scholar] [CrossRef] [PubMed]
  21. Fernandez-Hernando, C.; Ramirez, C.M.; Goedeke, L.; Suarez, Y. MicroRNAs in metabolic disease. Arterioscler. Thromb. Vasc. Biol. 2013, 33, 178–185. [Google Scholar] [CrossRef] [PubMed]
  22. Guay, C.; Roggli, E.; Nesca, V.; Jacovetti, C.; Regazzi, R. Diabetes mellitus, a microRNA-related disease? Transl. Res. 2011, 157, 253–264. [Google Scholar] [CrossRef] [PubMed]
  23. Kameswaran, V.; Bramswig, N.C.; McKenna, L.B.; Penn, M.; Schug, J.; Hand, N.J.; Chen, Y.; Choi, I.; Vourekas, A.; Won, K.-J. Epigenetic regulation of the DLK1-MEG3 microRNA cluster in human type 2 diabetic islets. Cell Metab. 2014, 19, 135–145. [Google Scholar] [CrossRef] [PubMed]
  24. Balasubramanyam, M.; Aravind, S.; Gokulakrishnan, K.; Prabu, P.; Sathishkumar, C.; Ranjani, H.; Mohan, V. Impaired miR-146a expression links subclinical inflammation and insulin resistance in type 2 diabetes. Mol. Cell. Biochem. 2011, 351, 197–205. [Google Scholar] [CrossRef] [PubMed]
  25. Caporali, A.; Meloni, M.; Vollenkle, C.; Bonci, D.; Sala-Newby, G.B.; Addis, R.; Spinetti, G.; Losa, S.; Masson, R.; Baker, A.H.; et al. Deregulation of microRNA-503 contributes to diabetes mellitus-induced impairment of endothelial function and reparative angiogenesis after limb ischemia. Circulation 2011, 123, 282–291. [Google Scholar] [CrossRef] [PubMed]
  26. Karolina, D.S.; Armugam, A.; Tavintharan, S.; Wong, M.T.; Lim, S.C.; Sum, C.F.; Jeyaseelan, K. MicroRNA 144 impairs insulin signaling by inhibiting the expression of insulin receptor substrate 1 in type 2 diabetes mellitus. PLoS ONE 2011, 6, e22839. [Google Scholar] [CrossRef]
  27. Karolina, D.S.; Tavintharan, S.; Armugam, A.; Sepramaniam, S.; Pek, S.L.; Wong, M.T.; Lim, S.C.; Sum, C.F.; Jeyaseelan, K. Circulating miRNA profiles in patients with metabolic syndrome. J. Clin. Endocrinol. Metab. 2012, 97, E2271–E2276. [Google Scholar] [CrossRef] [PubMed]
  28. Meng, S.; Cao, J.T.; Zhang, B.; Zhou, Q.; Shen, C.X.; Wang, C.Q. Downregulation of microRNA-126 in endothelial progenitor cells from diabetes patients, impairs their functional properties, via target gene Spred-1. J. Mol. Cell. Cardiol. 2012, 53, 64–72. [Google Scholar] [CrossRef] [PubMed]
  29. Ortega, F.J.; Moreno-Navarrete, J.M.; Pardo, G.; Sabater, M.; Hummel, M.; Ferrer, A.; Rodriguez-Hermosa, J.I.; Ruiz, B.; Ricart, W.; Peral, B. MiRNA expression profile of human subcutaneous adipose and during adipocyte differentiation. PLoS ONE 2010, 5, e9022. [Google Scholar] [CrossRef] [PubMed]
  30. Zampetaki, A.; Kiechl, S.; Drozdov, I.; Willeit, P.; Mayr, U.; Prokopi, M.; Mayr, A.; Weger, S.; Oberhollenzer, F.; Bonora, E.; et al. Plasma microRNA profiling reveals loss of endothelial miR-126 and other microRNAs in type 2 diabetes. Circul. Res. 2010, 107, 810–817. [Google Scholar] [CrossRef] [PubMed]
  31. Zhao, H.; Guan, J.; Lee, H.-M.; Sui, Y.; He, L.; Siu, J.J.; Patty, P.; Tong, P.C.; Lai, F.M.; Chan, J.C. Up-regulated pancreatic tissue microRNA-375 associates with human type 2 diabetes through β-cell deficit and islet amyloid deposition. Pancreas 2010, 39, 843–846. [Google Scholar] [CrossRef] [PubMed]
  32. Baldeon Rojas, L.; Weigelt, K.; de Wit, H.; Ozcan, B.; van Oudenaren, A.; Sempertegui, F.; Sijbrands, E.; Grosse, L.; Freire, W.; Drexhage, H.A.; et al. Decreased serum level of miR-146a as sign of chronic inflammation in type 2 diabetic patients. PLoS ONE 2014, 9, e115209. [Google Scholar]
  33. Granjon, A.; Gustin, M.P.; Rieusset, J.; Lefai, E.; Meugnier, E.; Guller, I.; Cerutti, C.; Paultre, C.; Disse, E.; Rabasa-Lhoret, R.; et al. The microRNA signature in response to insulin reveals its implication in the transcriptional action of insulin in human skeletal muscle and the role of a sterol regulatory element-binding protein-1C/myocyte enhancer factor 2C pathway. Diabetes 2009, 58, 2555–2564. [Google Scholar] [CrossRef] [PubMed]
  34. Kong, L.; Zhu, J.; Han, W.; Jiang, X.; Xu, M.; Zhao, Y.; Dong, Q.; Pang, Z.; Guan, Q.; Gao, L.; et al. Significance of serum microRNAs in pre-diabetes and newly diagnosed type 2 diabetes: A clinical study. Acta Diabetol. 2011, 48, 61–69. [Google Scholar] [CrossRef] [PubMed]
  35. Mao, G.; Liu, L. MicroRNA-18a is a genetic marker for the early diagnosis of cerebral injury induced by type 2 diabetes. Exp. Ther. Med. 2014, 8, 1901–1905. [Google Scholar] [CrossRef] [PubMed]
  36. Locke, J.; da Silva Xavier, G.; Dawe, H.; Rutter, G.; Harries, L. Increased expression of miR-187 in human islets from individuals with type 2 diabetes is associated with reduced glucose-stimulated insulin secretion. Diabetologia 2014, 57, 122–128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Ren, Y.H.; Shi, X.X.; Yan, D.D.; Li, X.L. The change of expression level of circulating miRNA-126 in patients with type 2 diabetes and its relative factors. Clin. J. Diabetes 2014, 22, 633–636. [Google Scholar]
  38. Santovito, D.; De Nardis, V.; Marcantonio, P.; Mandolini, C.; Paganelli, C.; Vitale, E.; Buttitta, F.; Bucci, M.; Mezzetti, A.; Consoli, A.; et al. Plasma exosome microRNA profiling unravels a new potential modulator of adiponectin pathway in diabetes: Effect of glycemic control. J. Clin. Endocrinol. Metab. 2014, 99, 1681–1685. [Google Scholar] [CrossRef] [PubMed]
  39. Sun, K.; Chang, X.; Yin, L.; Li, J.; Zhou, T.; Zhang, C.; Chen, X. Expression and DNA methylation status of microRNA-375 in patients with type 2 diabetes mellitus. Mol. Med. Rep. 2014, 9, 967–972. [Google Scholar] [CrossRef] [PubMed]
  40. Yang, Z.; Chen, H.; Si, H.; Li, X.; Ding, X.; Sheng, Q.; Chen, P.; Zhang, H. Serum miR-23a, a potential biomarker for diagnosis of pre-diabetes and type 2 diabetes. Acta Diabetol. 2014, 51, 823–831. [Google Scholar] [CrossRef] [PubMed]
  41. Zhang, L.Q.; Han, Z.P. Expression and clinical significance of peripheral hsa-miR-29b during the type 2 diabetes. J. Mod. Lab. Med. 2014, 29, 113–116. [Google Scholar]
  42. Agarwal, P.; Srivastava, R.; Srivastava, A.K.; Ali, S.; Datta, M. MiR-135a targets IRS2 and regulates insulin signaling and glucose uptake in the diabetic gastrocnemius skeletal muscle. Biochim. Biophys. Acta 2013, 1832, 1294–1303. [Google Scholar] [CrossRef] [PubMed]
  43. Corral-Fernandez, N.E.; Salgado-Bustamante, M.; Martinez-Leija, M.E.; Cortez-Espinosa, N.; Garcia-Hernandez, M.H.; Reynaga-Hernandez, E.; Quezada-Calvillo, R.; Portales-Perez, D.P. Dysregulated miR-155 expression in peripheral blood mononuclear cells from patients with type 2 diabetes. Exp. Clin. Endocrinol. Diabetes 2013, 121, 347–353. [Google Scholar] [CrossRef] [PubMed]
  44. Jiang, S.Y.; Jiao, J.; Zhang, T.T.; Xu, Y.P. Pharmacokinetics study of recombinant hirudin in the plasma of rats using chromogenic substrate, ELISA, and radioisotope assays. PLoS ONE 2013, 8, e64336. [Google Scholar] [CrossRef] [PubMed]
  45. Pescador, N.; Perez-Barba, M.; Ibarra, J.M.; Corbaton, A.; Martinez-Larrad, M.T.; Serrano-Rios, M. Serum circulating microRNA profiling for identification of potential type 2 diabetes and obesity biomarkers. PLoS ONE 2013, 8, e77251. [Google Scholar] [CrossRef] [PubMed]
  46. Rong, Y.; Bao, W.; Shan, Z.; Liu, J.; Yu, X.; Xia, S.; Gao, H.; Wang, X.; Yao, P.; Hu, F.B.; et al. Increased microRNA-146a levels in plasma of patients with newly diagnosed type 2 diabetes mellitus. PLoS ONE 2013, 8, e73272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Zhou, J.; Peng, R.; Li, T.; Luo, X.; Peng, H.; Zha, H.; Yin, P.; Wen, L.; Zhang, Z. A potentially functional polymorphism in the regulatory region of let-7a-2 is associated with an increased risk for diabetic nephropathy. Gene 2013, 527, 456–461. [Google Scholar] [CrossRef] [PubMed]
  48. Liu, Y.; Gao, G.; Yang, C.; Zhou, K.; Shen, B.; Liang, H.; Jiang, X. The role of circulating microRNA-126 (miR-126): A novel biomarker for screening prediabetes and newly diagnosed type 2 diabetes mellitus. Int. J. Mol. Sci. 2014, 15, 10567–10577. [Google Scholar] [CrossRef] [PubMed]
  49. Gallagher, I.J.; Scheele, C.; Keller, P.; Nielsen, A.R.; Remenyi, J.; Fischer, C.P.; Roder, K.; Babraj, J.; Wahlestedt, C.; Hutvagner, G. Integration of microRNA changes in vivo identifies novel molecular features of muscle insulin resistance in type 2 diabetes. Genome Med. 2010, 2, 9. [Google Scholar] [CrossRef] [PubMed]
  50. Klöting, N.; Berthold, S.; Kovacs, P.; Schön, M.R.; Fasshauer, M.; Ruschke, K.; Stumvoll, M.; Blüher, M. MicroRNA expression in human omental and subcutaneous adipose tissue. PLoS ONE 2009, 4, e4699. [Google Scholar] [CrossRef] [PubMed]
  51. Pan, J.; Chen, H. Expression of miR-155 and miR-146a in patients with type 2 diabetes mellitus and its significance. Zhejiang Clin. Med. 2014, 16, 1042–1043. [Google Scholar]
  52. Yang, S.; Zhao, J.; Chen, Y.; Lei, M. Biomarkers associated with ischemic stroke in diabetes mellitus patients. Cardiovasc. Toxicol. 2016, 16, 213–222. [Google Scholar] [CrossRef] [PubMed]
  53. Yan, S.; Wang, T.; Huang, S.; Di, Y.; Huang, Y.; Liu, X.; Luo, Z.; Han, W.; An, B. Differential expression of microRNAs in plasma of patients with prediabetes and newly diagnosed type 2 diabetes. Acta Diabetol. 2016, 53, 693–702. [Google Scholar] [CrossRef] [PubMed]
  54. Wang, Y.; Li, J.Y.; Du, W.P.; Jin, W.B. Association analysis between plasma leptin, insulin levels and their microRNA expression in patients with type 2 diabetes mellitus. Chin. J. Diabetes 2016, 24, 28–31. [Google Scholar]
  55. Wang, C.; Wan, S.; Yang, T.; Niu, D.; Zhang, A.; Yang, C.; Cai, J.; Wu, J.; Song, J.; Zhang, C.Y.; et al. Increased serum microRNAs are closely associated with the presence of microvascular complications in type 2 diabetes mellitus. Sci. Rep. 2016, 6, 20032. [Google Scholar] [CrossRef] [PubMed]
  56. Seyhan, A.A.; Nunez Lopez, Y.O.; Xie, H.; Yi, F.; Mathews, C.; Pasarica, M.; Pratley, R.E. Pancreas-enriched miRNAs are altered in the circulation of subjects with diabetes: A pilot cross-sectional study. Sci. Rep. 2016, 6, 31479. [Google Scholar] [CrossRef] [PubMed]
  57. Rezk, N.A.; Sabbah, N.A.; Saad, M.S. Role of microRNA 126 in screening, diagnosis, and prognosis of diabetic patients in Egypt. IUBMB Life 2016, 68, 452–458. [Google Scholar] [CrossRef] [PubMed]
  58. Pek, S.L.; Sum, C.F.; Lin, M.X.; Cheng, A.K.; Wong, M.T.; Lim, S.C.; Tavintharan, S. Circulating and visceral adipose miR-100 is down-regulated in patients with obesity and type 2 diabetes. Mol. Cell. Endocrinol. 2016, 427, 112–123. [Google Scholar] [CrossRef] [PubMed]
  59. Li, M.Y.; Pan, S.R.; Qiu, A.Y. Roles of microRNA-221/222 in type 2 diabetic patients with post-menopausal breast cancer. Genet. Mol. Res. 2016. [Google Scholar] [CrossRef] [PubMed]
  60. Latouche, C.; Natoli, A.; Reddy-Luthmoodoo, M.; Heywood, S.E.; Armitage, J.A.; Kingwell, B.A. MicroRNA-194 modulates glucose metabolism and its skeletal muscle expression is reduced in diabetes. PLoS ONE 2016, 11, e0155108. [Google Scholar] [CrossRef] [PubMed]
  61. Jansen, F.; Wang, H.; Przybilla, D.; Franklin, B.S.; Dolf, A.; Pfeifer, P.; Schmitz, T.; Flender, A.; Endl, E.; Nickenig, G.; et al. Vascular endothelial microparticles-incorporated microRNAs are altered in patients with diabetes mellitus. Cardiovasc. Diabetol. 2016, 15, 49. [Google Scholar] [CrossRef] [PubMed]
  62. Hou, X.; Wu, W.; Yin, B.; Liu, X.; Ren, F. MicroRNA-463–3p/ABCG4: A new axis in glucose-stimulated insulin secretion. Obesity 2016, 24, 2368–2376. [Google Scholar] [CrossRef] [PubMed]
  63. Ding, L.; Ai, D.; Wu, R.; Zhang, T.; Jing, L.; Lu, J.; Zhong, L. Identification of the differential expression of serum microRNA in type 2 diabetes. Biosci. Biotechnol. Biochem. 2016, 80, 461–465. [Google Scholar] [CrossRef] [PubMed]
  64. Baldeon Rojas, L.; Weigelt, K.; de Wit, H.; Ozcan, B.; van Oudenaren, A.; Sempertegui, F.; Sijbrands, E.; Grosse, L.; van Zonneveld, A.J.; Drexhage, H.A.; et al. Study on inflammation-related genes and microRNAs, with special emphasis on the vascular repair factor HGF and miR-574-3p, in monocytes and serum of patients with T2D. Diabetol. Metab. Syndr. 2016, 8, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Sebastiani, G.; Po, A.; Miele, E.; Ventriglia, G.; Ceccarelli, E.; Bugliani, M.; Marselli, L.; Marchetti, P.; Gulino, A.; Ferretti, E.; et al. MicroRNA-124a is hyperexpressed in type 2 diabetic human pancreatic islets and negatively regulates insulin secretion. Acta Diabetol. 2015, 52, 523–530. [Google Scholar] [CrossRef] [PubMed]
  66. Olivieri, F.; Spazzafumo, L.; Bonafe, M.; Recchioni, R.; Prattichizzo, F.; Marcheselli, F.; Micolucci, L.; Mensa, E.; Giuliani, A.; Santini, G.; et al. MiR-21-5p and miR-126a-3p levels in plasma and circulating angiogenic cells: Relationship with type 2 diabetes complications. Oncotarget 2015, 6, 35372–35382. [Google Scholar] [PubMed]
  67. Long, Y.; Zhan, Q.; Yuan, M.; Duan, X.; Zhou, J.; Lu, J.; Li, Z.; Yu, F.; Zhou, X.; Yang, Q.; et al. The expression of microRNA-223 and FAM5C in cerebral infarction patients with diabetes mellitus. Cardiovasc. Toxicol. 2017, 17, 42–48. [Google Scholar] [CrossRef] [PubMed]
  68. Lenin, R.; Sankaramoorthy, A.; Mohan, V.; Balasubramanyam, M. Altered immunometabolism at the interface of increased endoplasmic reticulum (ER) stress in patients with type 2 diabetes. J. Leukoc. Biol. 2015, 98, 615–622. [Google Scholar] [CrossRef] [PubMed]
  69. Jiao, Y.; Zhu, M.; Mao, X.; Long, M.; Du, X.; Wu, Y.; Abudureyimu, K.; Zhang, C.; Wang, Y.; Tao, Y.; et al. MicroRNA-130a expression is decreased in Xinjiang Uygur patients with type 2 diabetes mellitus. Am. J. Transl. Res. 2015, 7, 1984–1991. [Google Scholar] [PubMed]
  70. Higuchi, C.; Nakatsuka, A.; Eguchi, J.; Teshigawara, S.; Kanzaki, M.; Katayama, A.; Yamaguchi, S.; Takahashi, N.; Murakami, K.; Ogawa, D.; et al. Identification of circulating miR-101, miR-375 and miR-802 as biomarkers for type 2 diabetes. Metabolism 2015, 64, 489–497. [Google Scholar] [CrossRef] [PubMed]
  71. Bao, L.; Fu, X.; Si, M.; Wang, Y.; Ma, R.; Ren, X.; Lv, H. MicroRNA-185 targets SOCS3 to inhibit β-cell dysfunction in diabetes. PLoS ONE 2015, 10, e0116067. [Google Scholar] [CrossRef] [PubMed]
  72. Baldeon Rojas, L.; Weigelt, K.; de Wit, H.; Ozcan, B.; van Oudenaren, A.; Sempertegui, F.; Sijbrands, E.; Grosse, L.; van Zonneveld, A.J.; Drexhage, H.A.; et al. Type 2 diabetes monocyte microRNA and mRNA expression: Dyslipidemia associates with increased differentiation-related genes but not inflammatory activation. PLoS ONE 2015, 10, e0129421. [Google Scholar]
  73. Al-Kafaji, G.; Al-Mahroos, G.; Alsayed, N.A.; Hasan, Z.A.; Nawaz, S.; Bakhiet, M. Peripheral blood microRNA-15a is a potential biomarker for type 2 diabetes mellitus and pre-diabetes. Mol. Med. Rep. 2015, 12, 7485–7490. [Google Scholar] [CrossRef] [PubMed]
  74. Kong, L.; Han, W.X.; Jiang, X.Y.; AXu, J.; Zhang, X.; Li, Q.; Guan, Q.B.; Gao, L.; Zhao, J.J. Expression and clinical significance of peripheral miR-34a during the onset of type 2 diabetes. J. Shandong Univ. 2010, 48, 1–3. [Google Scholar]
  75. Liang, G.W.; Song, Y.; Shao, D.H.; Xu, X.; He, M.L. The change of serum miR-375 and miR-29a and their correlation with glycemic control and lipid profile in patients with newly diagnosed type 2 diabete. Chin. J. Lab. Diagn. 2013, 17, 475–478. [Google Scholar]
  76. Lu, S.Y.; Lu, Z.H.; Tan, L.; Wan, S. Association of Plasma miR-375 and miR-126 in patients with type 2 diabetes mellitus. J. Mod. Lab. Med. 2014, 29, 18–21. [Google Scholar]
  77. Ortega, F.J.; Mercader, J.M.; Moreno-Navarrete, J.M.; Rovira, O.; Guerra, E.; Esteve, E.; Xifra, G.; Martinez, C.; Ricart, W.; Rieusset, J.; et al. Profiling of circulating microRNAs reveals common microRNAs linked to type 2 diabetes that change with insulin sensitization. Diabetes Care 2014, 37, 1375–1383. [Google Scholar] [CrossRef] [PubMed]
  78. Wang, X.; Sundquist, J.; Zoller, B.; Memon, A.A.; Palmer, K.; Sundquist, K.; Bennet, L. Determination of 14 circulating microRNAs in Swedes and Iraqis with and without diabetes mellitus type 2. PLoS ONE 2014, 9, e86792. [Google Scholar] [CrossRef] [PubMed]
  79. Wu, B.; Liu, W.-W.; HE, Y.; Hu, Y.S.; Zhang, Z.L. Analysis on related metabolic influencing factors for miR-21 expression levels in type 2 diabetic patients. Mod. Prev. Med. 2015, 42, 2218–2222. [Google Scholar]
  80. Yan, S.T.; Li, C.L.; Tian, H.; Li, J.; Pei, Y.; Liu, Y.; Gong, Y.P.; Fang, F.S.; Sun, B.R. MiR-199a is overexpressed in plasma of type 2 diabetes patients which contributes to type 2 diabetes by targeting GLUT4. Mol. Cell. Biochem. 2014, 397, 45–51. [Google Scholar] [CrossRef] [PubMed]
  81. Zhou, B.; Li, C.; Qi, W.; Zhang, Y.; Zhang, F.; Wu, J.X.; Hu, Y.N.; Wu, D.M.; Liu, Y.; Yan, T.T.; et al. Downregulation of miR-181a upregulates sirtuin-1 (SIRT1) and improves hepatic insulin sensitivity. Diabetologia 2012, 55, 2032–2043. [Google Scholar] [CrossRef] [PubMed]
  82. Berends, L.; Ozanne, S. Early determinants of type-2 diabetes. Best Pract. Res. Clin. Endocrinol. Metabol. 2012, 26, 569–580. [Google Scholar] [CrossRef] [PubMed]
  83. Lorenzen, J.; Kumarswamy, R.; Dangwal, S.; Thum, T. MicroRNAs in diabetes and diabetes-associated complications. RNA Biol. 2012, 9, 820–827. [Google Scholar] [CrossRef] [PubMed]
  84. Dehwah, M.A.S.; Xu, A.; Huang, Q. MicroRNAs and type 2 diabetes/obesity. J. Genet. Genom. 2012, 39, 11–18. [Google Scholar] [CrossRef] [PubMed]
  85. Rome, S. Are extracellular microRNAs involved in type 2 diabetes and related pathologies? Clin. Biochem. 2013, 46, 937–945. [Google Scholar] [CrossRef] [PubMed]
  86. Pansuria, M.; Xi, H.; Li, L.; Yang, X.-F.; Wang, H. Insulin resistance, metabolic stress, and atherosclerosis. Front. Biosci. 2012, 4, 916–931. [Google Scholar]
  87. Poy, M.N.; Eliasson, L.; Krutzfeldt, J.; Kuwajima, S.; Ma, X.; MacDonald, P.E.; Pfeffer, S.; Tuschl, T.; Rajewsky, N.; Rorsman, P. A pancreatic islet-specific microRNA regulates insulin secretion. Nature 2004, 432, 226–230. [Google Scholar] [CrossRef] [PubMed]
  88. Hashimoto, N.; Kido, Y.; Uchida, T.; Asahara, S.-I.; Shigeyama, Y.; Matsuda, T.; Takeda, A.; Tsuchihashi, D.; Nishizawa, A.; Ogawa, W. Ablation of PDK1 in pancreatic β-cells induces diabetes as a result of loss of β-cell mass. Nat. Genet. 2006, 38, 589–593. [Google Scholar] [CrossRef] [PubMed]
  89. Gilbert, E.R.; Liu, D. Epigenetics: The missing link to understanding β-cell dysfunction in the pathogenesis of type 2 diabetes. Epigenetics 2012, 7, 841–852. [Google Scholar] [CrossRef] [PubMed]
  90. Plaisance, V.; Waeber, G.; Regazzi, R.; Abderrahmani, A. Role of microRNAs in Islet β-cell compensation and failure during diabetes. J. Diabetes Res. 2014, 2014, 618652. [Google Scholar] [CrossRef] [PubMed]
  91. Nesca, V.; Guay, C.; Jacovetti, C.; Menoud, V.; Peyot, M.-L.; Laybutt, D.R.; Prentki, M.; Regazzi, R. Identification of particular groups of microRNAs that positively or negatively impact on β-cell function in obese models of type 2 diabetes. Diabetologia 2013, 56, 2203–2212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Sacco, J.; Adeli, K. MicroRNAs: Emerging roles in lipid and lipoprotein metabolism. Curr. Opin. Lipidol. 2012, 23, 220–225. [Google Scholar] [CrossRef] [PubMed]
  93. Williams, M.D.; Mitchell, G.M. MicroRNAs in insulin resistance and obesity. Exp. Diabetes Res. 2012, 2012, 484696. [Google Scholar] [CrossRef] [PubMed]
  94. Karbiener, M.; Fischer, C.; Nowitsch, S.; Opriessnig, P.; Papak, C.; Ailhaud, G.; Dani, C.; Amri, E.-Z.; Scheideler, M. MicroRNA miR-27b impairs human adipocyte differentiation and targets PPARγ. Biochem. Biophys. Res. Commun. 2009, 390, 247–251. [Google Scholar] [CrossRef] [PubMed]
  95. Peng, Y.; Yu, S.; Li, H.; Xiang, H.; Peng, J.; Jiang, S. MicroRNAs: Emerging roles in adipogenesis and obesity. Cell. Signal. 2014, 26, 1888–1896. [Google Scholar] [CrossRef] [PubMed]
  96. Hamar, P. Role of regulatory microRNAs in type 2 diabetes mellitus-related inflammation. Nucleic Acid Ther. 2012, 22, 289–294. [Google Scholar] [PubMed]
  97. Keren, A.; Tamir, Y.; Bengal, E. The p38 MAPK signaling pathway: A major regulator of skeletal muscle development. Mol. Cell. Endocrinol. 2006, 252, 224–230. [Google Scholar] [CrossRef] [PubMed]
  98. Song, M.-Y.; Pan, K.-F.; Su, H.-J.; Zhang, L.; Ma, J.-L.; Li, J.-Y.; Yuasa, Y.; Kang, D.; Kim, Y.S.; You, W.-C. Identification of serum microRNAs as novel non-invasive biomarkers for early detection of gastric cancer. PLoS ONE 2012, 7, e33608. [Google Scholar] [CrossRef] [PubMed]
  99. Pan, F.; You, J.; Liu, Y.; Qiu, X.; Yu, W.; Ma, J.; Pan, L.; Zhang, A.; Zhang, Q. Differentially expressed microRNAs in the corpus cavernosum from a murine model with type 2 diabetes mellitus-associated erectile dysfunction. Mol. Genet. Genom. 2016, 291, 2215–2224. [Google Scholar] [CrossRef] [PubMed]
  100. He, J.S.; Lian, C.W.; Fang, Y.L.; Wu, J.Z.; Ye, X.L.; Zhu, S.B. Influence and significance of intervening diabetes microRNA expression profile of NOD mice with exendin-4. Eur Rev. Med. Pharmacol. Sci. 2016, 20, 4322–4327. [Google Scholar] [PubMed]
  101. Singh, G.B.; Raut, S.K.; Khanna, S.; Kumar, A.; Sharma, S.; Prasad, R.; Khullar, M. MicroRNA-200c modulates DUSP-1 expression in diabetes-induced cardiac hypertrophy. Mol. Cell. Biochem. 2016, 424, 1–11. [Google Scholar] [CrossRef] [PubMed]
  102. Wei, S.; Zhang, M.; Yu, Y.; Xue, H.; Lan, X.; Liu, S.; Hatch, G.; Chen, L. HNF-4α regulated miR-122 contributes to development of gluconeogenesis and lipid metabolism disorders in type 2 diabetic mice and in palmitate-treated HepG2 cells. Eur. J. Pharmacol. 2016, 791, 254–263. [Google Scholar] [CrossRef] [PubMed]
  103. Chen, H.Y.; Zhong, X.; Huang, X.R.; Meng, X.M.; You, Y.; Chung, A.C.; Lan, H.Y. MicroRNA-29b inhibits diabetic nephropathy in db/db mice. Mol. Ther. 2014, 22, 842–853. [Google Scholar] [CrossRef] [PubMed]
  104. Guay, C.; Regazzi, R. Role of islet microRNAs in diabetes: Which model for which question? Diabetologia 2015, 58, 456–463. [Google Scholar] [CrossRef] [PubMed]
  105. Huang, B.; Qin, W.; Zhao, B.; Shi, Y.; Yao, C.; Li, J.; Xiao, H.; Jin, Y. MicroRNA expression profiling in diabetic GK rat model. Acta Biochim. Biophys. Sin. 2009, 41, 472–477. [Google Scholar] [CrossRef] [PubMed]
  106. Lu, T.P.; Lee, C.Y.; Tsai, M.H.; Chiu, Y.C.; Hsiao, C.K.; Lai, L.C.; Chuang, E.Y. MiRSystem: An integrated system for characterizing enriched functions and pathways of microRNA targets. PLoS ONE 2012, 7, e42390. [Google Scholar] [CrossRef] [PubMed]
  107. Cho, S.; Jang, I.; Jun, Y.; Yoon, S.; Ko, M.; Kwon, Y.; Choi, I.; Chang, H.; Ryu, D.; Lee, B.; et al. MiRGator v3.0: A microRNA portal for deep sequencing, expression profiling and mRNA targeting. Nucleic Acids Res. 2013, 41, 252–257. [Google Scholar] [CrossRef] [PubMed]
  108. Backes, C.; Kehl, T.; Stockel, D.; Fehlmann, T.; Schneider, L.; Meese, E.; Lenhof, H.P.; Keller, A. MiRPathDB: A new dictionary on microRNAs and target pathways. Nucleic Acids Res. 2017, 45, D90–D96. [Google Scholar] [CrossRef] [PubMed]
  109. Palmer, J.D.; Soule, B.P.; Simone, B.A.; Zaorsky, N.G.; Jin, L.; Simone, N.L. MicroRNA expression altered by diet: Can food be medicinal? Ageing Res. Rev. 2014, 17, 16–24. [Google Scholar] [CrossRef] [PubMed]
  110. Chen, G.Q.; Lian, W.J.; Wang, G.M.; Wang, S.; Yang, Y.Q.; Zhao, Z.W. Altered microRNA expression in skeletal muscle results from high-fat diet-induced insulin resistance in mice. Mol. Med. Rep. 2012, 5, 1362–1368. [Google Scholar] [PubMed]
  111. Baier, S.R.; Nguyen, C.; Xie, F.; Wood, J.R.; Zempleni, J. MicroRNAs are absorbed in biologically meaningful amounts from nutritionally relevant doses of cow milk and affect gene expression in peripheral blood mononuclear cells, HEK-293 kidney cell cultures, and mouse livers. J. Nutr. 2014, 144, 1495–1500. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The flow chart of the data selection and identification process.
Figure 1. The flow chart of the data selection and identification process.
Ijms 18 00456 g001
Figure 2. Schematic diagram of the microRNA-mRNA interaction networks in pancreatic islets. The networks are built based on the KEGG pathway map: T2DM pathway (KEGG hsa04930) (A); adipocytokine pathway (KEGG hsa04920) (B); and insulin signaling pathway (KEGG hsa04910) (C). The genes are indicated by circles and miRNAs by squares. For detailed descriptions of node shape, edge size, edge color and arrow shapes see inserted legend under (A). The predicted miRNA-mRNA interactions are indicated by light blue lines, and the thick blue lines indicate validated miRNA-mRNA interactions. Interactions denoted in KEGG pathways were presented as black solid or dotted lines indicating direct or indirect interactions, respectively. The identity of genes and miRNAs involved in the process are listed, and the colors indicate the expression level changes in T2DM islets compared to the control; red indicates a higher level in patients; and green represents lower levels compared to the control.
Figure 2. Schematic diagram of the microRNA-mRNA interaction networks in pancreatic islets. The networks are built based on the KEGG pathway map: T2DM pathway (KEGG hsa04930) (A); adipocytokine pathway (KEGG hsa04920) (B); and insulin signaling pathway (KEGG hsa04910) (C). The genes are indicated by circles and miRNAs by squares. For detailed descriptions of node shape, edge size, edge color and arrow shapes see inserted legend under (A). The predicted miRNA-mRNA interactions are indicated by light blue lines, and the thick blue lines indicate validated miRNA-mRNA interactions. Interactions denoted in KEGG pathways were presented as black solid or dotted lines indicating direct or indirect interactions, respectively. The identity of genes and miRNAs involved in the process are listed, and the colors indicate the expression level changes in T2DM islets compared to the control; red indicates a higher level in patients; and green represents lower levels compared to the control.
Ijms 18 00456 g002aIjms 18 00456 g002b
Figure 3. Schematic diagram of the microRNA-mRNA interaction networks in adipose tissue. The networks are built based on the KEGG pathway map: T2DM pathway (KEGG hsa04930) (A); adipocytokine pathway (KEGG hsa04920) (B); and insulin signaling pathway (KEGG hsa04910) (C). The genes are indicated by circles and miRNAs by squares. See the legend of Figure 2A for descriptions of node shape, edge size, edge color and arrow shapes. The identity of genes and miRNAs involved in the process are listed, and the colors indicate the relative expression changes in T2DM adipose tissues compared to the control; red indicates higher level in patients; and green represents lower levels compared to the control. The mRNA expression levels were obtained from GSE16415 (available online: https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE16415).
Figure 3. Schematic diagram of the microRNA-mRNA interaction networks in adipose tissue. The networks are built based on the KEGG pathway map: T2DM pathway (KEGG hsa04930) (A); adipocytokine pathway (KEGG hsa04920) (B); and insulin signaling pathway (KEGG hsa04910) (C). The genes are indicated by circles and miRNAs by squares. See the legend of Figure 2A for descriptions of node shape, edge size, edge color and arrow shapes. The identity of genes and miRNAs involved in the process are listed, and the colors indicate the relative expression changes in T2DM adipose tissues compared to the control; red indicates higher level in patients; and green represents lower levels compared to the control. The mRNA expression levels were obtained from GSE16415 (available online: https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE16415).
Ijms 18 00456 g003aIjms 18 00456 g003b
Table 1. Main characteristics of the reports included in the study.
Table 1. Main characteristics of the reports included in the study.
Author, Year (Ref.)CountrySample TypemiRNAExp ChangeAssay MethodNumber of SamplesAvg AgeGender (M/F)QC
Ding et al., 2016 [63]ChinaSerummiR-451a, -4534UpRT-qPCRT2DM(40)/NC(56)61.2159/379
miR-320d, -3960, -572Down
Hou et al., 2016 [62]ChinaPancreatic isletsmiR-463-3pUpRT-qPCRT2DM(34)/NC(34)53 ± 7.535/337
Jansen et al., 2016 [61]GermanyPlasmamiR-126, -26aDownRT-qPCRT2DM(55)/NC(80)66.4 ± 10.945/907
Latouche et al., 2016 [60]AustraliaSMmiR-194DownRT-qPCRT2DM(6)/NC(5)50.72NA8
Li et al., 2016 [59]ChinaSerummiR-221/222UpRT-qPCRT2DM(30)/NC(20)60.28NA7
Pek et al., 2016 [58]China, Malaysia, India and OthersAdipose tissuemiR-100, miR-378, miR-99a, miR-125b, miR-181a, miR-210 and miR-378DownMicroarray RT-qPCRT2DM(42)/NC(73)40.4 ± 11.25NA7
Rezk et al., 2016 [57]EgyptSerummiR-126DownRT-qPCRT2DM(100)/NC(100)46.9595/1059
Seyhan et al., 2016 [56]USAPlasmamiR-30d, -34a, -21, -148aUpRT-qPCRT2DM(31)/NC(27)40.0530/289
Yan et al., 2016 [53]ChinaPlasmamiR-572UpMicroarray RT-qPCRT2DM(50)/NC(50)45.8749/518
miR-1249, -320bDown
Wang et al., 2016 [55]ChinaSerummiR-661, -571, -770-5p, -892b, -1303, -15a, -16, -125b, -221, -320aUpRT-qPCRT2DM(92)/NC(92)48.95114/708
Baldeon et al., 2016 [64]EcuadorSerummiR-574-3p, -146aDownRT-PCRT2DM(64)/NC(44)61 (37–85)37/718
Wang et al., 2016 [54]ChinaPlasmamiR-296, -9DownRT-qPCRT2DM(150)/NC(150)48.6 ± 1.7150/1507
Long et al., 2015 [67]ChinaPBMCmiR-223-3pDownRT-qPCRT2DM(16)/NC(18)5520/146
Olivieri et al., 2015 [66]ItalyPBMCmiR-126-3p, -21-5pDownRT-qPCRT2DM(76)/NC(107)64.7985/989
Yang et al., 2015 [52]ChinaPlasma/plateletsmiR-144UpRT-qPCRT2DM(114)/NC(30)49.8 ± 9.1NA8
miR-223Down
Higuchi et al., 2015 [70]JapanSerummiR-101, -375, -802UpRT-qPCRT2DM(155)/NC(49)62.3 ± 13.2121/837
Al-Kafaji et al., 2015 [73]BahrainWBmiRNA-15aDownRT-qPCRT2DM(24)/NC(24)52 ± 6.023/258
Lenin et al., 2015 [68]IndiaPBMCmiR-146aDownRT-qPCRT2DM(35)/NC(35)47.3 ± 736/346
Sebastiani et al., 2015 [65]ItaliaPancreatic isletsmiR-124aUpRT-qPCRT2DM(5)/NC(10)71.2 ± 9.87/87
Jiao et al., 2015 [69]ChinaPBmiR-130a, -10b, -143DownRT-qPCRT2DM(30)/NC(42)56 ± 10NA8
Bao et al., 2015 [71]ChinaPlasma/SerummiR-185DownRT-qPCRT2DM(34)/NC(30)NANA9
Baldeón et al., 2015 [72]EcuadorPBMCmiR-34c-5p, -576-3pUpMicroarray RT-qPCRT2DM(64)/NC(44)61 (37–85)37/716
Wu et al., 2015 [79]ChinaPBMCmiR-21UpRT-qPCRT2DM(18)/NC(18)53.6 ± 4.618/187
Ortega et al., 2014 [77]SpainPlasmamiR-140-5p, -142-3p, -222UpRT-qPCRT2DM(48)/NC(45)54 ± 1093/08
miR-423-5p, -125b, -192, -195, -130b, -532-5p, -126Down
Yan et al., 2014 [80]ChinaPlasmamiR-199aUpRT-PCRT2DM(64)/NC(64)46–62NA8
Lu et al., 2014 [76]ChinaPlasmamiR-375, miR-126UpRT-qPCRT2DM(30)/NC(30)53.67 ± 8.9242/188
Wang et al., 2014 [78]Swedes IraqisPlasmamiR-15a, -21, -144, -150, -486-5pUpRT-qPCRT2DM(33)/NC(119)45-6583/697
miR-24, -29b, -126, -320aDown
Liu et al., 2014 [35]ChinaSerummiR-126DownqPCRT2DM(160)/NC(138)50.2 ± 6.778/829
Pan et al., 2014 [51]ChinaWBmiR-146a, -155DownFQ-PCRT2DM(36)/NC(32)61.0 ± 7.020/169
Locke et al., 2014 [36]UKIsletmiR-129-3p, -187, -345UpRT-qPCRT2DM(9)/NC(11)537/25
Yang et al., 2014 [40]ChinaSerummiR-23a, let-7i, -486, -96, -186, -191, -192, -146aDownRT-qPCRT2DM(24)/NC(20)50.60 ± 5.12816/89
Santovito et al., 2014 [38]GermanyPlasmamiR-326UpRT-qPCRT2DM(18)/NC(12)57.2 ± 9.612/67
miR-let-7a,let-7fDown
Mao et al., 2014 [35]ChinaSerummiR-18aDownqPCRT2DM(33)/NC(33)53.8 (35–72)13/206
Baldeon et al., 2014 [32]NetherlandsSerummiR-146aDownRT-qPCRT2DM(56)/NC(40)62 (38–85)22/348
Sun et al., 2014 [39]ChinaPlasmamiR-375UpqPCRT2DM(100)/NC(100)51.33 ± 11.7554/469
Kameswaran et al., 2014 [23]USAIsletmiR-7, -136, -369, -369-3p, -411, -432, -487a, -487b, -495, -539-3p, -655, -656DownRT-qPCRT2DM(20)/NC(29)51.35 (22–65)14/68
miR-187, -187 *, -224, -589Up
Zhang et al., 2014 [41]ChinaSerummiR-29bUpRT-PCRT2DM(50)/NC(50)35–7030/208
Ren et al., 2014 [37]ChinaPlasmamiR-126DownRT-PCRT2DM(40)/NC(40)43.0 ± 11.024/169
Zhou et al., 2013 [47]ChinaWBlet-7aUpRT-PCRT2DM(104)/NC(62)52.8 ± 10.459/459
Pescador et al., 2013 [45]SpainSerummiR-503DownRT-qPCRT2DM(13)/NC(20)69.40 ± 7.127/68
Zhang et al., 2013 [47]ChinaPlasmamiR-126DownRT-qPCRT2DM(30)/NC(30)63 ± 8.56 (42–73)16/148
Agarwal et al., 2013 [42]IndiaSMmiR-135aUpRT-PCRT2DM(3)/NC(3)65–753/05
Rong et al., 2013 [46]ChinaPlasmamiR-146aUpqPCRT2DM(90)/NC(90)48.50 (42–56)47/439
Corral, et al., 2013 [43]MéxicoPBMCmiR-146a, -155DownRT-PCRT2DM(20)/NC(20)46.2 (35–59)11/96
Liang et al, 2013 [75]ChinaSerummiR-29a, -375UpRT-FQ-PCRT2DM(48)/NC(38)54.9 ± 9.8 (35–72)27/219
Zhou et al., 2012 [81]ChinaSerummiR-181aUpRT-PCRT2DM(20)/NC(20)NANA7
Meng et al., 2012 [28]ChinaPBMCmiR-21, -27a, -27b, -126, -130aDownMicroarray RT-qPCRT2DM(15)/NC(15)67 ± 87/88
Karolina et al., 2012 [27]SingaporeWBmiR-17, -92a, -130a, -195, -197, -509-5p, -652DownMicroarray RT-qPCRT2DM(50)/NC(46)42.02NA9
miR-27a, -150, -192, -320a, -375Up
Balasubramanyam et al., 2011 [24]IndiaPBMCmiR-146aDownRT-qPCRT2DM(20)/NC(20)43.7 ± 5.1NA8
Caporali et al., 2011 [25]U.K.PlasmamiR-503UpRT-PCRT2DM(10)/NC(11)68.09 ± 9.069/17
SMmiR-503
Karolina et al., 2011 [26]SingaporeWBmiR-15a, -17, -17 *, -23a, -23b, -26a, -26b, -27a, -29b, -29c, -99b *, -106b, -125a-5p, -125b, -126, -130a, -130b, -142-3p, -151-3p, -151-5p, -183, -185, -190, -193a-3p, -194, -221, -222, -299-3p, -320b, -320c, -320d, -335, -361-3p, -375, -502-3p, -550, -550 *, -589, -620, -629, -665, -886-5p, -1285, -1301UpMicroarrayT2DM(21)/NC(15)43.2 (21–70)21/09
miR-7, -19a, -20a, -20b, -30c, -30e, -34b, -106a, -129-5p, -146b-5p, -185 *, -186, -340, -342-3p, -362-5p, -374b, -519e, -532-3p, -636, -637, -652, -660, -923, -1184, -1297, let-7b *, let-7d, let-7e, let-7g, let-7iDown
miR-29a, -144, -150, -192, -320aUpRT-qPCR
miR-30d, -146a, -182Down
Kong et al., 2011 [34]ChinaSerummiR-9, -29a, -30d, -34a, -124a, -146a, -375UpRT-qPCRT2DM(18)/NC(19)47.33 ± 2.6179/99
Zhao et al., 2010 [31]Hong KongPancreasmiR-375UpRT-qPCRT2DM(40)/NC(15)69 ± 1317/238
Zampetaki et al., 2010 [30]U.K.PlasmamiR-15a, -20b, -21, -24, -29b, -126, -150, -191, -197, -223, -320, -486DownMicroarray/RT-qPCRT2DM(80)/NC(80)66.3 ± 8.930/505
miR-28-3pUp
Gallagher et al., 2010 [49]U.K.SMmiR-15b, -30b *, -30c-2 *, -32 *, -93, -106b, -138-1 *, -142-3p, -142-5p, -143, -144, -181a-2 *, -185, -193a-5p, -371-5p, -451, -503, -518c *, -589, -597, -600, -634, -658, -665, -668, -765, -921, -923, -937UpMicroarrayT2DM(45)/NC(47)54.8 ± 10.2NA7
miR-10a, -10b, -15a, -27b, -30e, -95, -100, -126 *, -128, -133a, -152, -154, -190, -196a, -199a-3p, -199b-5p, -206, -208a, -331-3p, -342-3p, -362-3p, -374a, -374b, -378 *, -422a, -423-3p, -424, -455-5p, -519d, -768-3p, -768-5p, -801Down
Ortega et al., 2010 [29]SpainAdiposemiR-125b, -199a-5p, -221, -1229UpRT-PCRT2DM(9)/NC(6)45 ± 100/98
miR-30a *, -130b, -484Down
Kong et al., 2010 [74]ChinaSerummiR-34aUpRT-qPCRT2DM(18)/NC(26)47.33 ± 2.6223/21
Klöting et al., 2009 [50]GermanyAdiposemiR-147, -181a, -197UpMicroarrayT2DM(6)/NC(9)67 ± 2.8NA8
miR-17-5p, -27a, -30e, -132, -134, -140, -155, -210Down
Granjon et al., 2009 [33]FranceSMmiR-1, -133aDownRT-qPCRT2DM(5)/NC(15)51 ± 22/37
Abbreviations: T2DM: type 2 diabetes; NC: normal control; NA: not available; PBMC, peripheral blood mononuclear cell; WB: whole blood; SM: skeletal muscle.
Table 2. List of dysregulated miRNAs identified from seven different types of samples used in T2DM studies.
Table 2. List of dysregulated miRNAs identified from seven different types of samples used in T2DM studies.
Increased in T2DM Patient a
miR-101-3p
miR-124-3p
miR-125b-5p
miR-1303
miR-126-3pmiR-146a-5p
miR-140-5pmiR-15a-5p
miR-142-3pmiR-16-5p
miR-144-3pmiR-181a-5p
miR-106b-5p miR-146a-5pmiR-221-3p
miR-135a-5pmiR-451a miR-148a-3pmiR-222-3p
miR-138-1-3pmiR-503-5p miR-15a-5pmiR-29a-3p
miR-142-3pmiR-518c-5p miR-150-5pmiR-29b-3p
miR-142-5pmiR-589-5p miR-199a-5pmiR-30d-5p
miR-143-3pmiR-597-5p miR-21-5pmiR-320a
miR-144-3pmiR-600 miR-222-3pmiR-34a-5p
miR-124-3pmiR-15b-5pmiR-634let-7a-5p miR-28-3pmiR-375
miR-1229-3pmiR-129-3pmiR-181a-2-3pmiR-658miR-144-3p miR-30d-5pmiR-451a
miR-125b-5pmiR-187-3pmiR-185-5pmiR-665miR-150-5p miR-326miR-4534
miR-147amiR-187-5pmiR-193a-5pmiR-668-3pmiR-192-5p miR-34a-5pmiR-571
miR-181a-5pmiR-224-5pmiR-30b-3pmiR-765miR-27a-3p miR-375miR-661
miR-197-3pmiR-345-5pmiR-30c-2-3pmiR-921miR-29a-3pmiR-21-5pmiR-486-5pmiR-770-5p
miR-199a-5pmiR-375miR-32-3pmiR-93-5pmiR-320amiR-34c-5pmiR-503-5pmiR-892b
miR-221-3pmiR-589-5pmiR-371a-5pmiR-937-3pmiR-375miR-576-3pmiR-572miR-9-5p
Adipose bIslet bSkeletal Muscle bWhole Blood bPBMC bPlasma bSerum b
T2DM(72)/NC(103)T2DM(88)/NC(113)T2DM(114)/NC(128)T2DM(378)/NC(312)T2DM(264)/NC(277)T2DM(1058)/NC(1234)T2DM(961)/NC(821)
miR-100-5pmiR-136-5pmiR-100-5pmiR-208a-3pmiR-10b-5pmiR-126-3plet-7a-5plet-7i-5p
miR-125b-5pmiR-369-3pmiR-10a-5pmiR-27b-3pmiR-130a-3pmiR-130a-3plet-7f-5pmiR-126-3p
miR-130b-3pmiR-411-5pmiR-10b-5pmiR-30e-5pmiR-143-3pmiR-146a-5pmiR-1249-3pmiR-146a-5p
miR-132-3pmiR-432-5pmiR-126-5pmiR-331-3pmiR-146a-5pmiR-155-5pmiR-125b-5pmiR-186-5p
miR-134-5pmiR-487a-3pmiR-128-3pmiR-342-3pmiR-155-5pmiR-21-5pmiR-126-3pmiR-18a-5p
miR-140-5pmiR-487b-3pmiR-133a-3pmiR-362-3pmiR-15a-5pmiR-223-3pmiR-130b-3pmiR-191-5p
miR-155-5pmiR-495-3pmiR-1-3pmiR-374a-5pmiR-17-5pmiR-27a-3pmiR-15a-5pmiR-192-5p
miR-17-5pmiR-539-3pmiR-152-3pmiR-374b-5pmiR-182-5pmiR-27b-3pmiR-150-5pmiR-23a-3p
miR-181a-5pmiR-655-3pmiR-154-5pmiR-378a-5pmiR-195-5p miR-191-5pmiR-320d
miR-210-3pmiR-656-3pmiR-15a-5pmiR-422amiR-197-3p miR-192-5pmiR-3960
miR-27a-3pmiR-7-5pmiR-190a-5pmiR-423-3pmiR-30d-5p miR-195-5pmiR-486-5p
miR-30a-5p miR-194-5pmiR-424-5pmiR-509-5p miR-197-3pmiR-503-5p
miR-30e-5p miR-196a-5pmiR-455-5pmiR-652-3p miR-20b-5pmiR-572
miR-378a-3p miR-199a-3pmiR-519d-3pmiR-92a-3p miR-21-5pmiR-574-3p
miR-484 miR-199b-5pmiR-95-3p miR-223-3pmiR-96-5p
miR-99a-5p miR-206 miR-24-3p
miR-26a-5p
miR-296-5p
miR-29b-3p
miR-320a
miR-320b
miR-423-5p
miR-486-5p
miR-532-5p
miR-9-5p
Decreased in T2DM patient a
a miRNAs reported in multiple studies are listed in italic boldface characters, and inconsistent concentration changes between studies from the same sample type are underlined; b the number of samples involved in patients (T2DM) and controls (NC) is indicated in parentheses under each sample type.
Table 3. Common changes of miRNAs in different sample types.
Table 3. Common changes of miRNAs in different sample types.
miRNA IDPlasmaSerumPBMCWhole BloodSkeletal MuscleIsletAdipose
let-7a-5pDown Up
miR-100-5p DownDown
miR-124-3p Up Up
miR-125b-5pDownUp Up
Down
miR-126-3pUpDownDown Down
Down
miR-126-5p Down
miR-130a-3p DownDown
miR-140-5pUp Down
miR-142-3pUp Up
miR-143-3p DownUP
miR-144-3pUp UpUp
miR-146a-5pUpDownDownDown
Up
miR-150-5pUp Up
Down
miR-155-5p DownDown
miR-15a-5pUpUp DownDown
Down
miR-17-5p Down Down
miR-181a-5p Up Up
Down
miR-191-5pDownDown
miR-192-5pDownDown Up
miR-195-5pDown Down
miR-197-3pDown Down Up
miR-199a-5pUp Up
miR-21-5pUp Up
DownDown
miR-221-3p UP Up
miR-222-3pUpUp
miR-223-3pDown Down
miR-27a-3p DownUp
miR-27b-3p Down Down
miR-29a-3p Up Up
miR-29b-3pDownUp
miR-30d-5pUpUp Down
miR-30e-5p Down Down
miR-320aDownUp Up
miR-34a-5pUpUp
miR-375UpUp Up Up
miR-451a Up Up
miR-486-5pUpDown
Down
miR-503-5pUpDown Up
miR-572UpDown
miR-589-5p UpUp
miR-9-5pDownUp

Share and Cite

MDPI and ACS Style

He, Y.; Ding, Y.; Liang, B.; Lin, J.; Kim, T.-K.; Yu, H.; Hang, H.; Wang, K. A Systematic Study of Dysregulated MicroRNA in Type 2 Diabetes Mellitus. Int. J. Mol. Sci. 2017, 18, 456. https://doi.org/10.3390/ijms18030456

AMA Style

He Y, Ding Y, Liang B, Lin J, Kim T-K, Yu H, Hang H, Wang K. A Systematic Study of Dysregulated MicroRNA in Type 2 Diabetes Mellitus. International Journal of Molecular Sciences. 2017; 18(3):456. https://doi.org/10.3390/ijms18030456

Chicago/Turabian Style

He, Yuqing, Yuanlin Ding, Biyu Liang, Juanjuan Lin, Taek-Kyun Kim, Haibing Yu, Hanwei Hang, and Kai Wang. 2017. "A Systematic Study of Dysregulated MicroRNA in Type 2 Diabetes Mellitus" International Journal of Molecular Sciences 18, no. 3: 456. https://doi.org/10.3390/ijms18030456

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