Circulating microRNAs associated with gestational diabetes mellitus: useful biomarkers?

in Journal of Endocrinology
Authors:
Sofie Dinesen Department of Science and Environment, Roskilde University, Universitetsvej 1, Roskilde, Denmark

Search for other papers by Sofie Dinesen in
Current site
Google Scholar
PubMed
Close
https://orcid.org/0000-0002-3718-5008
,
Alisar El-Faitarouni Department of Science and Environment, Roskilde University, Universitetsvej 1, Roskilde, Denmark

Search for other papers by Alisar El-Faitarouni in
Current site
Google Scholar
PubMed
Close
https://orcid.org/0000-0002-0688-9053
, and
Louise T Dalgaard Department of Science and Environment, Roskilde University, Universitetsvej 1, Roskilde, Denmark

Search for other papers by Louise T Dalgaard in
Current site
Google Scholar
PubMed
Close
https://orcid.org/0000-0002-3598-2775

Correspondence should be addressed to L T Dalgaard: ltd@ruc.dk
Free access

Sign up for journal news

Different types of small non-coding RNAs, especially miRNAs, may be found in the circulation, either protein-bound or enclosed in extracellular vesicles. During gestation, and particularly during gestational diabetes mellitus (GDM), the levels of several miRNAs are altered. Worldwide the incidence of GDM is increasing, in part driven by the current obesity epidemic. This is a point of public health concern because offspring of women with GDM frequently suffer from short- and long-term complications of maternal GDM. This has prompted the investigation of whether levels of specific miRNA species, detected early in gestation, may be used as diagnostic or prognostic markers for the development of GDM. Here, we summarize the mechanisms of RNA secretion and review circulating miRNAs associated with GDM. Several miRNAs are associated with GDM: miR-29a-3p and miR-29b-3p are generally upregulated in GDM pregnancies, also when measured prior to the development of GDM, while miR-16-5p is consistently upregulated in GDM pregnancies, especially in late gestation. miR-330-3p in circulation is increased in late gestation GDM women, especially in those with poor insulin secretion. miR-17-5p, miR-19a/b-3p, miR-223-3p, miR-155-5p, miR-125-a/b-5p, miR-210-3p and miR-132 are also associated with GDM, but less so and with more contradictory results reported. There could be a publication bias as miRNAs identified early are investigated the most, suggesting that it is likely that additional, more recently detected miRNAs could also be associated with GDM. Thus, circulating miRNAs show potential as biomarkers of GDM diagnosis or prognosis, especially multiple miRNAs containing prediction algorithms show promise, but further studies are needed.

Abstract

Different types of small non-coding RNAs, especially miRNAs, may be found in the circulation, either protein-bound or enclosed in extracellular vesicles. During gestation, and particularly during gestational diabetes mellitus (GDM), the levels of several miRNAs are altered. Worldwide the incidence of GDM is increasing, in part driven by the current obesity epidemic. This is a point of public health concern because offspring of women with GDM frequently suffer from short- and long-term complications of maternal GDM. This has prompted the investigation of whether levels of specific miRNA species, detected early in gestation, may be used as diagnostic or prognostic markers for the development of GDM. Here, we summarize the mechanisms of RNA secretion and review circulating miRNAs associated with GDM. Several miRNAs are associated with GDM: miR-29a-3p and miR-29b-3p are generally upregulated in GDM pregnancies, also when measured prior to the development of GDM, while miR-16-5p is consistently upregulated in GDM pregnancies, especially in late gestation. miR-330-3p in circulation is increased in late gestation GDM women, especially in those with poor insulin secretion. miR-17-5p, miR-19a/b-3p, miR-223-3p, miR-155-5p, miR-125-a/b-5p, miR-210-3p and miR-132 are also associated with GDM, but less so and with more contradictory results reported. There could be a publication bias as miRNAs identified early are investigated the most, suggesting that it is likely that additional, more recently detected miRNAs could also be associated with GDM. Thus, circulating miRNAs show potential as biomarkers of GDM diagnosis or prognosis, especially multiple miRNAs containing prediction algorithms show promise, but further studies are needed.

Introduction

The current worldwide obesity epidemic drives an increased incidence and prevalence of gestational diabetes mellitus (GDM) as well as type 2 diabetes (T2D) in pregnancy (Sun et al. 2022). In 2019, according to the International Diabetes Federation (IDF), 16% of viable children were affected by hyperglycemia in pregnancy, corresponding to 20 million live births of which GDM accounts for 75–90% (Sun et al. 2022). GDM is defined as diabetes that is diagnosed for the first time during pregnancy, at any time during pregnancy. While GDM usually resolves following delivery, women who develop GDM during pregnancy are at high risk of developing T2D later in life or have recurrent GDM in future pregnancies. Of importance, GDM carries a risk of a number of complications for both mother and child (Damm et al. 2016). There is an increased morbidity for both during pregnancy and around birth. For the mother, there is an increased risk of preeclampsia and increased mortality around the time of birth, while GDM fetuses are also at risk of macrosomia, premature birth, neonatal icterus and perinatal hypoglycemia (Teh et al. 2011). Women developing GDM have a higher degree of insulin resistance compared with women who remain normoglycemic in pregnancy (Kampmann et al. 2019), and late sequelae include a higher risk of developing diabetes, particularly T2DM, and metabolic diseases later in life (Kelstrup et al. 2013).

In efforts to prevent GDM, several risk factors have been identified that may contribute to the onset of GDM in women. These include obesity, smoking, family history of diabetes, birth of a child with macrosomia, ethnicity (all ethnicities except Anglo-European) and maternal age (Teh et al. 2011).

Therefore, the IDF and IADPSG recommend screening for GDM (International Association of Diabetes in Pregnancy Study Group 2015), preferably via a measurement of plasma glucose levels (HbA1c, random or fasting plasma glucose values) in all or high-risk women at the first antenatal visit, followed by screening using oral glucose tolerance test (OGTT) at gestational weeks 24–28. This is a very comprehensive screening program and some countries, such as Denmark, rely on screening based on risk factors such as elevated BMI or family history of diabetes or previous birth of a large child. Screening is usually performed from gestational weeks 24 to 28, as insulin resistance increases in the second trimester and blood glucose levels rise. However, at that time point, the unborn child may already have been affected long enough to develop metabolic adaptations that may cause complications later in life, such as the increased risk of T2D and cardiovascular disease (Damm et al. 2016, Rani & Begum, 2016). Therefore, there is a search for better biomarkers to allow diagnosis of GDM at an earlier time point or give more specific prognostic estimates of the risk of GDM. To this end, circulating levels of small non-coding RNA molecules, especially miRNA, have been investigated and suggested as a novel category of biomarkers potentially serving to improve the diagnosis and prediction of disease development (Condrat et al. 2020). Identifying potential biomarkers for early prediction of GDM before week 20 of pregnancy will help identify and treat incident GDM in pregnant women. The current review aims at describing the current status of circulating small RNAs, primarily miRNAs, associated with gestational diabetes, and further evaluate the potential of such to act as prognostic biomarkers for GDM.

miRNA biogenesis

miRNAs are small non-coding single-stranded RNA molecules of about 22 nucleotides in length, found in plants, animals, viruses, human tissues and blood. More than 2000 different miRNAs have been identified in the human genome (Kozomara et al. 2019). miRNAs act via RNA silencing to regulate, in a posttranscriptional manner, the degradation of mRNA, thereby adjusting protein levels, and about 30% of protein-coding genes are predicted to be targeted by one or more miRNAs.

miRNAs are transcribed in the nucleus to form a primary transcript, which is cut into a precursor miRNA by the nuclease DROSHA forming a hairpin structure. Next, the pre-miRNA is transported to the cytoplasm by EXPORTIN-5, where it is further cleaved by DICER to form a duplex short RNA with imperfect base pairing (Fig. 1). The mature miRNA duplex is unwound and the mature single-stranded miRNA is assembled into the RNA-induced silencing complex (RISC) (consisting of DICER, TRBP and AGO2) to then induce translational inhibition or transcript degradation of mRNAs to which it can base pair (Fig. 1) (Bartel & Chen 2004, Chendrimada et al. 2005, Chu et al. 2010, Guo et al. 2010, Eichhorn et al. 2014). The miRNA–RISC complex then identifies possible target mRNAs through sequence complementary of the miRNA seed sequence to the 3′-UTR of the target mRNA. The seed sequence consists of 6–8 nucleotides at the 5′-end of the miRNA, where perfect or near perfect complementary base pairing between the miRNA and mRNA results in a rapid degradation of the transcript, while partial complementary between the miRNA–mRNA complex prevents the protein translation process. However, miRNA-dependent repression also results in mRNA decay, which has been shown to account for most miRNA-dependent repression (Mathys et al. 2014, Filipowicz & Sonenberg 2015).

Figure 1
Figure 1

miRNA and EV biogenesis. The canonical miRNA biogenesis starts as a primary miRNA transcript is formed in the nucleus by RNA polymerase II. Primary miRNA transcripts contain partially complementary hairpin structures. miRNA genes may be located as isolated transcriptional units or be located in exons or introns of protein-coding genes. DROSHA, a nuclease, removes the sequences outside the hairpin to form the precursor miRNA, in combination with the partner DGCR8. Subsequently, the miRNA precursor is exported to the cytoplasm by EXPORTIN 5 using RAN-GTP. Then, the nuclease DICER with TRBP removes the hairpin turn and the resulting duplex mature miRNA is unwound and inserts itself into the RISC complex. The miRNA–RISC complex then degrades or halts the transcription of mRNAs which are recognized by the miRNA. The RISC complex may dock to the ER membrane to load miRNAs into exosomes and multivesicular bodies. Extracellular RNA may also be released from cells in microvesicles that bud from the plasma membrane or may be bound by RNA-binding proteins, both of which will protect the miRNA in circulation from degradation. The figure was partly generated using Servier Medical Art, provided by Servier, licensed under a Creative Commons Attribution 3.0 unported license.

Citation: Journal of Endocrinology 256, 1; 10.1530/JOE-22-0170

Circulating miRNA as biomarkers

In addition to playing a major role inside the cell, miRNA also plays an important role outside the cell, in body fluids and elsewhere as circulating miRNAs. Circulating RNAs may be found in blood, saliva, breast milk and urine, as well as other fluids, for example, in the case of tissue damage but also as the result of controlled excretion. Approximately 10% of circulating miRNAs are secreted in exosomes, which are a specific type of extracellular vesicles (EVs) (Chevillet et al. 2014, Albanese et al. 2021). The remaining 90% of the miRNAs are encased in other EVs, such as microvesicles or apoptotic bodies, or form complexes with proteins such as Ago2 or with HDL particles (Arroyo et al. 2011, Vickers et al. 2011, Boon & Vickers 2013).

Enclosing the miRNA in vesicles or binding it to protein complexes prevents the miRNA from being digested and thus it remains stable in body fluids protected from ribonucleases. Exosomes derived from multivesicular bodies, a specialized subset of endosomes, are loaded with miRNAs at the endoplasmic reticulum (ER) surface and which involves the ER membrane protein VAP-A and the ceramide transfer protein CERT (Barman et al. 2022). This is interesting because it has also been demonstrated that mRNA–miRNA interactions via Ago2 loading take place at the ER membrane (Barman & Bhattacharyya 2015). The secretion of miRNAs from cells into exosomes depends on the enzyme neutral sphingomyelinase 2, which is known as a rate-limiting enzyme of ceramide biosynthesis (Kosaka et al. 2010). Exosomal miRNA transfer appears to be a selective process because the miRNA content within the exosomes is quite dissimilar to the miRNA composition of the parent cell (Guduric-Fuchs et al. 2012, Villarroya-Beltri et al. 2013, Squadrito et al. 2014). Microvesicles, formed by outward budding of the plasma membrane, have miRNAs delivered by the ADP ribosylation factor ARF6, which binds Exportin 5 (Clancy et al. 2019). Thus, the loading of both small and large EVs appears to be under regulatory control. For example, miRNAs, miR-155, miR-210 and miR-23 are selectively loaded into exosomes massively increasing their release from cells during inflammation facilitated by the RNA-binding protein FMR1 and a common A-A/U-U/A-GC motif in these miRNAs (Wozniak et al. 2020).

During pregnancy, the number of EVs increases in circulation, especially EVs derived from the placenta (Salomon et al. 2018, Menon et al. 2019). The stability and ease of detection by quantitative reverse transcription PCR (qRT-PCR) has fostered an intense scientific interest in the possible use of miRNAs as circulating biomarkers for a large variety of pathological conditions. Studies measuring miRNAs in circulation make use of different experimental strategies for quantifying miRNA levels, the most commonly used techniques being qRT-PCR, small RNA-seq and arrays. Quantitative RT-PCR is advantageous when few miRNAs are measured in many clinical samples and assays can be made with spike-ins during RNA isolation and during cDNA synthesis, enabling control for variation in RNA isolation and cDNA synthesis efficiency. On the other hand, small RNA-seq has the advantage of measuring all small RNAs in a sample but is rarely made with spike-ins and the cost usually prohibits measuring large cohorts. Small RNA arrays can be standardized with spike-ins to enable quantitative measurements, but they suffer from lower sensitivity and therefore require the use of higher sample volumes for RNA extraction (Table 1).

Table 1

Commonly used methods for miRNA biomarker studies.

Measurement method Principle Advantages Limitations
Quantitative reverse-transcription polymerase chain reaction (qRT-PCR) In qRT-PCR, cDNA is synthesized using a reverse transcriptase enzyme and the target is amplified by PCR.

The quantification is done using fluorescent detection during the PCR amplification and relies on the first detectable cycle of product formation (named Cq or Ct).

Often performed on one miRNA target at a time, although qPCR arrays can measure up to 384 wells at a time.
Highly quantitative

Very sensitive

Spike-ins can be added during RNA-isolation and cDNA synthesis, which are then quantified during the qPCR to enable absolute quantification.

Low cost enables the measurement of larger cohorts.
Is a targeted approach, hence only candidate miRNAs are investigated.

No general and consensus data analysis strategy exists.
Small RNA-sequencing (smRNA-seq) In smRNA-seq, RNA linkers are ligated to the RNA and used to generate an smRNA library, which is amplified, size-selected and sequenced using massive parallel sequencing methods (i.e. using Illumina or Ion Torrent sequencing). Allows the detection of all small RNAs in a sample.

Partially quantitative: Although spike-ins can be added during library synthesis, this is rarely implemented.

Very sensitive.
Per sample higher cost.

Highly abundant miRNAs constitute a large fraction of the reads and require higher sequence depth to increase costs.

No general and consensus data analysis strategy exists.
Small RNA arrays All small RNAs in a sample are labeled and hybridized to an oligonucleotide array. To increase hybridization strengths to the short miRNAs, oligos are often modified using locked nucleic acid analogs. Allows the detection of all known miRNAs.

RNAs in a given sample.

Not sensitive

Per sample cost is low, given the number of miRNAs measured.
Has a limited dynamic range.

Data analysis strategies are more uniform.

Not all circulating miRNAs have the potential as diagnostic or prognostic biomarkers. miRNA, as a biomarker, must be able to meet certain criteria to be considered as a biomarker candidate; it must be readably detectable and measurable. Moreover, its specificity and its sensitivity for the condition under investigation are important criteria to be considered and it must have clinical relevance. Since circulating miRNA is a relatively new research area, for most miRNAs in circulation, the influence of different parameters such as age, gender and disease/health on their levels, as well as population variability is not known. Moreover, since thrombocytes also contain miRNAs, the levels of specific miRNAs, such as miR-451 and miR-223, are released into the serum following platelet aggregation. Therefore, such miRNAs have different levels in serum and plasma and it is important to make note of the sample material reported in clinical studies of circulating miRNAs.

Literature search methods

The databases PubMed, Science Direct and Scopus were searched using the string: (Gestational diabetes OR pregnancy-induced diabetes) AND (microRNA OR miRNA OR microribonucl*) AND human. Further literature was identified through searches on bioRxiv.org and medRxiv.org and via the literature references of identified articles.

Circulating RNAs associated with gestational diabetes: what has been found?

Based on our literature searches, we identified 22 original studies (Table 2) investigating circulating levels of more than 130 different miRNA species in relation to GDM (Fig. 2). However, the majority of the different miRNA types were only investigated in one or two studies (Fig. 2). Only 33 different miRNA species were investigated in more than 1 original study, with miR-29a-3p and miR-16-5p being the most studied miRNAs, investigated in 6 (Zhao et al. 2011, Wander et al. 2017, Tagoma et al. 2018, Gillet et al. 2019, Martinez-Ibarra et al. 2019, Sorensen et al. 2021, 2022) and 6 (Zhu et al. 2015, Cao et al. 2017, Tagoma et al. 2018, Hocaoglu et al. 2019, Martinez-Ibarra et al. 2019, Sorensen et al. 2021, 2022) original studies, respectively. miR-223-3p (Wander et al. 2017, Tagoma et al. 2018, Yoffe et al. 2019, Sorensen et al. 2021), miR-330-3p (Martinez-Ibarra et al. 2019, Pfeiffer et al. 2020, Xiao et al. 2020, Sorensen et al. 2021) and miR-132-3p (Zhao et al. 2011, Tagoma et al. 2018, Gillet et al. 2019, Zhou et al. 2019) were each investigated in four studies, while miR-155-5p (Wander et al. 2017, Tagoma et al. 2018, Hocaoglu et al. 2019), miR-210-3p (Wander et al. 2017, Tagoma et al. 2018, Gillet et al. 2019), miR-19a/b-3p (Cao et al. 2017, Stirm et al. 2018, Tagoma et al. 2018), miR-17-5p (Cao et al. 2017, Lamadrid-Romero et al. 2018, Tagoma et al. 2018) and miR-125a/b-5p (Zhao et al. 2011, Lamadrid-Romero et al. 2018, Nair et al. 2018) were each studied in three original studies (Table 1, Fig. 2). Thus, it is clear that the majority of reported circulating miRNAs associated with GDM have only been identified in one study. Hence, we should assume that it is likely that additional miRNAs to those covered below are also associated with GDM, but these have not yet been sufficiently analyzed by the research community to be covered in this review.

Figure 2
Figure 2

Distribution of miRNAs examined in relation to GDM. The majority of original studies examining miRNAs for association with GDM were only examined in one published study, while miR-16-5p and miR-29a-3p were investigated in six studies each. Thus, the majority of reported circulating miRNAs associated with GDM were only identified in one study.

Citation: Journal of Endocrinology 256, 1; 10.1530/JOE-22-0170

Table 2

Studies of circulating miRNAs associated with GDM included in the narrative review.

Reference n GDM n NGT Trimester investigated Sample material Quantification methods
(Cao et al. 2017) 85 72 Not given Plasma RT-qPCR
(Filardi et al. 2022) 12 12 T3 Plasma Taqman array → RT-qPCR
(Gillet et al. 2019) 23 46 T1 + T2 Exosomes RT-qPCR
(Hocaoglu et al. 2019) 19 28 T3 Serum RT-qPCR
(Lamadrid-Romero et al. 2018) 12/24/16 13/24/20 T1/T2/T3 Serum RT-qPCR
(Martinez-Ibarra et al. 2019) 18 22 T2 Serum RT-qPCR
(Nair et al. 2018) 12 12 At birth (T3) Exosomes RT-qPCR
(Peng et al. 2018) 11 12 T3 Plasma RT-qPCR
(Pfeiffer et al. 2020) 31 29 T2 + T3 Serum RT-qPCR
(Qi & Wang 2019) 108/48 100 T3 Serum RT-qPCR
(Sebastiani et al. 2017) 21 10 T2/T3 Plasma Taqman array → RT-qPCR
(Stirm et al. 2018) 38 38 T2 + T3 Whole blood RT-qPCR
(Sorensen et al. 2021) 82 41 T1 and T2/3 GDM Serum RT-qPCR
(Sorensen et al. 2022) 82 41 T1 + T2 +T3 GDM Serum RT-qPCR
(Tagoma et al. 2018) 13 9 T2 + T3 Plasma RT-qPCR
(Wander et al. 2017) 36 80 T2 Plasma RT-qPCR
(Xiao et al. 2020) 30 10 T2 + T3 Serum RT-qPCR
(Yoffe et al. 2019) 23 20 T1 Plasma RT-qPCR
(Zhang & Chen 2020) 30 30 Not given Serum RT-qPCR
(Zhang et al. 2021) 61 57 T2 Exosomes RT-qPCR
(Zhao et al. 2011) 24 24 T2 + T3 Serum RT-qPCR
(Zhou et al. 2019) 108 50 T2 + T3 Serum RT-qPCR
(Zhu et al. 2015) 10 10 T2 Plasma RT-qPCR

Gives an overview of the studies included in the review. T1, trimester 1; T2, trimester 2; T3: trimester 3.

The miR-29 family members miR-29a-3p and miR-29b-3p are generally upregulated in GDM pregnancies

The miR-29 family consists of seven miRNAs, of which miR-29a-3p and miR-29b-3p are the two major isoforms. The miR-29a-3p, but also the other miR-29 members, have been found to be increased by obesity or prediabetes in metabolically relevant tissues, such as β-cells, adipose tissue, skeletal muscle and in the liver. Moreover, antisense inhibition of miR-29a-3p improves liver insulin resistance and improves glycemic control, altogether suggesting that the miR-29 family has an essential function in intermediate metabolism (Hung et al. 2019, Dalgaard et al. 2022). Sørensen et al. (Sorensen et al. 2021) examined miR-29a-3p in relation to GDM diagnosed early in pregnancy (before week 20) and late in pregnancy (weeks 24–28) and found significantly increased expression of miR-29a-3p in late-diagnosed GDM compared to the NGT group, which is supported by findings of Martínez-Ibarra et al. (Martinez-Ibarra et al. 2019) also identifying significantly increased miR-29a-3p in second trimester diagnosed GDM cases. Further support of a general upregulation of circulating miR-29a-3p, and possibly also miR-29b-3p in GDM pregnancies can be found in a study by Tagoma et al. (Tagoma et al. 2018), which found these 2 miRNAs upregulated more than 3-fold, although not reaching statistical significance, possibly as a cause of lower power due to a low number of investigated subjects. However, the published literature does not uniformly agree on the association of circulating miR-29a/b with GDM, as Wander et al. (Wander et al. 2017) and Zhao et al. (Zhao et al. 2011) found no or negative association, respectively, with GDM, although Wander et al. (Wander et al. 2017) observed increased miR-29a-3p in the circulation of GDM pregnancies, when carrying a male fetus. Thus, the majority of studies support that miR-29a-3p, and possibly also miR-29b-3p, are upregulated in GDM pregnancies in the second trimester compared with age and gestational age-matched control women with NGT. The reason for the different regulation of miR-29 in Zhao et al. (Zhao et al. 2011) is not known but may be due to the ethnicity of the sample population as this study was the only to investigate miR-29a-3p in Chinese women. Alternatively, different sample handling may explain the differing results, as Zhao et al. (Zhao et al. 2011) report blood sample processing within 4 h, whereas the studies by Wander et al. (Wander et al. 2017), Gillet et al. (Gillet et al. 2019), Martínez-Ibarra et al. (Martinez-Ibarra et al. 2019) and Sorensen et al. (Sorensen et al. 2021) processed samples within 1 h. Although miRNA is regarded as being stable in circulation, the difference in the processing of the samples could possibly result in relatively higher decay of the miRNA.

Altogether, these studies support that circulating miR-29a-3p, and to some degree miR-29b-3p, levels are also increased during GDM and may have biomarker potential as a diagnostic marker. However, prognostic biomarkers for GDM would be much more desirable. While Sorensen et al. (Sorensen et al. 2022) demonstrate a significant increase in the level of miR-29a-3p in serum already early in pregnancy (average week 16), which is confirmed by Gillet et al. (Gillet et al. 2019), although measured in EVs, the studies by Wander et al. (Wander et al. 2017) and Martínez-Ibarra et al. (Martinez-Ibarra et al. 2019) do not investigate early pregnancy samples (before week 20). Therefore, it is difficult to fully assess the ability of miR-29a-3p as a prognostic biomarker and to assess the possibility of using miR-29a-3p levels as a prognostic biomarker. Thus, more prospective studies are needed investigating early pregnancies. Interestingly, increased miR-29a-3p was found associated with T2D in a systematic review (Villard et al. 2015), and it is therefore conceivable that pregestational increased levels of miR-29a-3p may be carried forward into gestation.

miR-16-5p is upregulated especially in late GDM pregnancy

miR-16 is reported to affect insulin sensitivity in human and rodent tissue, but also in models of T2D. Insulin-resistant tissues have lower expression of miR-16 and muscle-specific miR-16 KO mice displayed impaired insulin sensitivity and protein turnover (Lim et al. 2022). Several studies have examined miR-16-5p and found it significantly increased in late-diagnosed GDM (>24 weeks of gestation) (Zhu et al. 2015, Cao et al. 2017, Martinez-Ibarra et al. 2019, Sorensen et al. 2021). Overall, studies report very similar increases: An approximate 2.5-fold increase in miR-16-5p in second and third trimester diagnosed GDM women compared with NGT pregnant women, with levels increasing throughout gestation (Cao et al. 2017, Sorensen et al. 2022). However, a smaller study was unable to detect any difference in circulating miR-16-5p (Hocaoglu et al. 2019), but this could be due to the low number of included subjects. Moreover, miR-16-5p was also found to be significantly increased in the first trimester (Cao et al. 2017, Sorensen et al. 2021, 2022), which suggests that miR-16-5p could potentially be used as a diagnostic as well as a prognostic biomarker of GDM. This is emphasized by the finding that miR-16-5p in combination with miR-29a-3p and the also GDM-associated miR-134-5p forms a superior diagnostic measure compared with 2 h plasma glucose following an OGTT (Sorensen et al. 2021), which is one of the recommended diagnostic measures of the IADPSG (International Association of Diabetes and Pregnancy Study Groups 2010).

Circulating miR-17-5p is increased throughout gestation

Downregulation of miR-17-5p was associated with a decrease in the size of the pancreatic islets, elevated levels of blood glucose and loss of glucose tolerance. Furthermore, exogenous miR-17-5p inhibited TXNIP and NLRP3 inflammasome activation and decreased streptozotocin-induced β-cell death (Liu et al. 2021). In a longitudinal study, miR-17-5p was measured during each trimester (T1, T2 and T3) corresponding to gestational weeks 16–20, 20–24 and 24–28, and it was found to be significantly increased in GDM women at all 3 measurement periods (Cao et al. 2017). This suggests a general upregulation of miR-17-5p in GDM independently of gestational age. Moreover, this study identified a positive correlation between the level of miR-17-5p and insulin resistance and an interaction with GDM, as the insulin resistance associated with pregnancy may be more pronounced in GDM pregnancies (Kampmann et al. 2019). Tagoma et al. (Tagoma et al. 2018) measured the level of miR-17-5p at weeks 23–31 of pregnancy and found a 10.9-fold increase in miR-17-5p and a 2.6-fold increase in circulating miR-17-5p levels. However, due to large variability, these findings were not statistically significant, although the observations were congruent with Cao et al. (Cao et al. 2017). Zhu et al. (Zhu et al. 2015) reported a 2-fold increase in miR-17-5p in week 24–18, similar to a study by Lamadrid-Romero et al. (Lamadrid-Romero et al. 2018), which showed tendencies of increased levels of miR-17-5p in GDM pregnancies, in T1 and T2, while they were unable to show differences in miR-17 levels between GDM and control women in the third trimester. Altogether these findings indicate a generally increased level of miR-17-5p in pregnancy, especially those complicated by GDM, despite the observation that downregulation of miR-17-5p in β-cells is associated with the development of diabetes (Liu et al. 2021).

Circulating miR-19a-3p and miR-19b-3p are only slightly increased in GDM

miR-19a and miR-19b are both part of the miR-17-92 (miR-17) locus and are excised from the same primary transcript. The direct role of miR-19 has not been thoroughly described, yet it might share some characteristics with miR-17 in supporting a healthy β-cell function. Elevated levels of miR-19 were observed in replicating β-cells compared with non-replicating β-cells (Mandelbaum et al. 2019). Moreover, in skeletal muscle, an inverse relation between miR-19a-3p and citrate synthase, a target of miR-19a-3p, expression was identified, suggesting that miR-19a-3p might regulate the mitochondrial capacity of skeletal muscles (Pinto et al. 2017). Cao et al. (Cao et al. 2017) examined miR-19a/b-3p and found no significant difference in the expression of either miRNA isoforms in their study in relation to GDM pregnancy. Both isoforms were measured several times during pregnancy at weeks 16–20, 20–24 and 24–28, with no significant difference observed in circulating levels of miR-19a/b-3p at any point. In contrast to this, the studies by Zhu et al. (2015) and Stirm et al. (2018) demonstrated a significant difference in the expression of both miR-19a-3p and miR-19b-3p in their screening group during pregnancy in weeks 24–28. miR-19a-3p displayed a fold change of 2.2 with an associated P-value of 0.002, while miR-19b-3p showed a fold change of 1.6 with an associated P-value of 0.005. Therefore, they chose to include both miRNAs in their validation group. However, in the validation group, no significant difference was detected with a P-value of 0.3 for both isoforms. The difference in results between the screening and validation groups may be due to the population sizes. In the screening group, a small population was used causing the result to be non-representative and therefore significant. When the miRNAs were examined in a larger population, the difference was no longer significant. In another study, miR-19a/b-3p was upregulated approximately 9-fold in GDM pregnancies, although this failed to reach statistical significance (Tagoma et al. 2018). Thus, based on these three studies, miR-19a/b-3p is not a possible diagnostic biomarker for GDM, as none of the studies found a difference in the expression of miR-19 in relation to GDM, although all three studies consistently showed an increase in circulating miR-19a/b-3p in GDM.

miR-223-3p could be increased early in gestation in GDM

In β-cells, miR-223 is upregulated by diabetes, and β-cell overexpression of miR-223 enhances proliferation, while deficiency of miR-223 increases apoptosis, ultimately reducing the functional β-cell mass (Li et al. 2019). Moreover, miR-223-3p is associated with impaired insulin sensitivity in adipose tissue (Sanchez-Ceinos et al. 2021). Several studies investigated the correlation between miR-223-3p and GDM: Yoffe et al. (Yoffe et al. 2019) showed a large, 9-fold, significant increase in expression of miR-223-3p between control and GDM groups with an adjusted P-value of 1.4 × 10−7, performed on samples from the first trimester, i.e. up to and including week 12. However, the study by Sorensen et al. (2021) did not identify any difference between GDM and NGT in early gestation, and thus, it was concluded that miR-223-3p could not be used as a prognostic biomarker. However, it may be difficult to compare these 2 studies, as the early GDM group in Sorensen et al. (2021) was defined as less than gestational week 20 with an average of gestational week 15.3. Furthermore, it is unknown whether the upregulated miR-223-3p in the first trimester, in the study by Yoffe et al. (2019), was maintained or decreased again in the second trimester. In another study, levels of miR-223-3p in circulation decreased through gestation (Sorensen et al. 2022). In samples measured during the second trimester, Wander et al. (2017) found miR-223-3p significantly increased in GDM women, if the data were corrected for the sex of the fetus. The study concluded that the miRNA was upregulated only in women, who were pregnant with boys. But based on the lack of significant difference without correction for fetal sex, the results from this study agree well with the conclusion of Sørensen et al. (Sorensen et al. 2021, 2022). Tagoma et al. (2018) examined the expression of miR-223-3p during pregnancy in weeks 23–31, and although the study showed a fold increase of 11 for miR-223-3p, this was not significant (P = 0.24). The studies that examined miR-223-3p, therefore, do not uniformly agree on the potential for use as a diagnostic biomarker for GDM. The studies are also not comparable due to different periods of examination; however, there is a general trend that miR-223-3p is increased in early gestation of GDM women, although larger studies early in pregnancy are needed to conclusively determine this.

Circulating miR-330-3p levels are associated with GDM in late gestation

The miR-330-3p gene is located within an intron of a longer non-coding RNA named EML2 (EMAP like 2 (Genome Browser: genome.ucsc.edu), with wide-spread expression pattern (Ludwig et al. 2016). It was reported upregulated in second-trimester plasma samples from GDM women and high levels of miR-330-3p were associated with increased risk of cesarean section and decreased β-cell function (Sebastiani et al. 2017). Similarly, two other studies found miR-330-3p increased in GDM pregnancies in the third trimester (Martinez-Ibarra et al. 2019, Pfeiffer et al. 2020), while early GDM had lower levels of miR-330-3p compared with matched glucose-tolerant control women (Sorensen et al. 2022). Interestingly, increased levels of miR-330-3p led to decreased levels of glucokinase in insulin-secreting cells (INS-1 cells) (Xiao et al. 2020), in concordance with findings of lower insulin secretion among GDM women with high levels of miR-330-3p in circulation (Sebastiani et al. 2017). In correlation to this, miR-330-5p might also play a role in the function of adipose tissue through actions on the macrophages, as an inverse correlation between the miR-330-5p and Tim-3 (T cell immunoglobulin domain, mucin domain) protein levels was observed in macrophages of mice, resulting in insulin resistance (Sun et al. 2018).

miR-155-5p in circulation is not consistently associated with GDM

miR-155-5p is an inflammation-regulated miRNA, involved in multiple inflammatory diseases (Worm et al. 2009, Ma et al. 2011, Moura et al. 2019). As hyperglycemia induces inflammatory signaling (Kelstrup et al. 2012), it is a credible hypothesis that miR-155-5p should be upregulated in GDM. miR-155 participates in the regulation of insulin sensitivity in adipose tissue, liver and skeletal muscle. Impaired glucose tolerance and decreased insulin sensitivity have been found in mice lacking miR-155, although these mice had an unaltered insulin production capacity. Dysregulated expression of miR-155 has also been shown to predict the development of some late complications of diabetes mellitus – such as retinopathy, neuropathy and nephropathy (Jankauskas et al. 2021).

We identified three studies, in which circulating levels of miR-155-5p were examined in relation to GDM, all of them in mid-to-late pregnancy. Wander et al. (Wander et al. 2017) examined miR-155-5p during the second trimester and detected an increased level in GDM women, when corrected for gestational age and fetal sex. The second study, by Tagoma et al. (2018), found miR-155-5p increased 4.6-fold, but this did not reach statistical significance. The third study by Hocaoglu et al. (2019) examined miR-155-5p in the third trimester but did not find any significant difference in their GDM group compared to their control group, although miR-155-5p was found to be decreased in pre-eclampsia. However, the study examined the level of the miRNA in leukocytes and not whole blood or serum. This creates a bias when compared with other studies, which examined plasma or serum. Thus, it appears that there is little to no biomarker potential for measuring miR-155-5p in GDM.

miR-125a-5p and miR-125b-5p appear differentially regulated during gestation

miR-125a/b are upregulated by obesity in adipose tissue (Herrera et al. 2009). Overexpression of miR-125a in mice increased insulin sensitivity, while knock-down reduced insulin sensitivity. Furthermore, miR-125a knock-down has been shown capable to attenuate lipid accumulation in hepatocytes (Liu et al. 2020). For the miR-125b-2 isomer, congruent observations were made, as deletion of miR-125b-2 increases liver and adipose tissue, and causes an accumulation of fat, reduced glucose utilization and decreased insulin sensitivity (Wei et al. 2020). These findings suggest that miR-125a and miR-125b play an important role in the regulation of insulin resistance and lipogenesis. Moreover, miR-125b is a negative regulator of insulin secretion, possibly via control intracellular lysosomes (Cheung et al. 2022), altogether suggesting that upregulation of miR-125 species may contribute to the development of T2D.

Lamadrid-Romero et al. (2018) investigated the expression of miR-125b-5p during all three trimesters of pregnancy (T1, T2 and T3) and showed increased circulating levels of miR-125b-5p in T2 and T3 in women with NGT during pregnancy, while levels in GDM complicated pregnant women were regulated differently showing increased levels of miR-125b-5p during T1, but decreased levels in during T2 and T3. These observations indicate that the timing of sampling could be important. Zhao et al. (2011) examined the amount of miR-125b-5p at weeks 16–19 but showed no significant changes in the expression of miR-125b-5p, while Nair et al. (2018) showed a significant increase (P = 0.05) in the expression of the miR-125a-5p isoform when measuring on circulating exosomes in plasma. However, as the exosome measurements made by Nair et al. (2018) were performed after birth, when the rise in miR-125a-5p containing plasma exosomes occurred is of uncertain cause. However, Zhang et al. (2021) found significantly decreased levels of miR-125b-5p in isolated exosomes from week 24 to 28. In the second and third trimesters, Tagoma et al. (2018) observed a 3-fold increase in circulating miR-125b-5p; however, this did not reach statistical significance (P = 0.14). Thus, studies regarding miR-125a/b-5p are inconsistent, as studies report an increase in early gestation (Lamadrid-Romero et al. 2018) or no difference (Zhao et al. 2011), while in mid-to-late gestation levels were either insignificantly increased (Tagoma et al. 2018) or significantly decreased (Lamadrid-Romero et al. 2018). Based on these observations, miR-125a/b-5p cannot be used as a biomarker for GDM, but studies investigating miR-125 species are generally small and have low power. Therefore, new studies for this miRNA should focus on investigating a larger number of subjects.

The hypoxamiR, miR-210-3p, in relation to GDM

miR-210-3p is known as a hypoxamiR because it is upregulated by hypoxia in most cell types and participates in the coordinated response to hypoxia (Zaccagnini et al. 2022). Decreased levels of miR-210-3p in β-cells result in β-cell functional impairment and apoptosis, which could contribute to the development of β-cell dysfunction and death during T2D (Nesca et al. 2013). Three studies studied circulating levels of miR-210-3p in GDM; Gillet et al. (2019) observed a significant increase of 1.3-fold in EVs from GDM women, examined between 6–15 weeks of gestation, while Wander et al. (2017) observed a 1.5-fold increase measured in the second trimester, but this was not significant. In another study by Tagoma et al. (2018), no significant difference between NGT and GDM was detected at gestational weeks 23–31, although average levels were increased in GDM. Thus, miR-210-3p was upregulated in EVs in early gestation, but when measured in plasma in second trimester, no significant changes were observed. No studies examined EVs in second trimester. Thus, although increases in miR-210-3p are reported, the magnitude of the increases is small and data appear variable. Based on this, miR-210-3p is not suitable as a diagnostic or prognostic marker of GDM. Possibly, miR-210-3p as a hypoxamiR would be a more relevant marker of intrauterine growth retardation due to insufficient placental function.

miR-132-3p is decreased in circulation in second trimester GDM patients

miR-132 downregulation correlates with a minor, but significant, inhibition of cell proliferation, whereas upregulated miR-132 expression was detected in different T2D models. Inhibition of the miR-132 expression in pancreatic β-cells has been associated with an increase in the cleavage of caspase-9, while upregulation of miR-132 resulted in reduced levels of caspase-9, indicating a necessary role of miR-132 in the protection against apoptosis (Mziaut et al. 2020). Moreover, in vivo whole-body silencing of miR-132 using an antisense oligonucleotide reduces blood glucose levels and increases insulin secretion (Bijkerk et al. 2019). Therefore, miR-132 might have a role in protecting against the development of diabetes by promoting β-cell expansion and decreasing β-cell death (Eliasson & Esguerra 2020).

We identified four studies investigating miR-132-3p in relation to GDM. In early gestation EVs miR-132-3p was increased 1.65-fold (Gillet et al. 2019), and, insignificantly, in plasma form second-trimester GDM patients (2.3-fold, P = 0.14) (Tagoma et al. 2018). However, two other studies found miR-132-3p decreased in GDM pregnancies (Zhao et al. 2011, Zhou et al. 2019); Zhao et al. (2011) observed a 32% reduction of miR-132-3p plasma of second trimester GDM patients, and Zhou et al. (2019) observed a highly significant 46% reduction of serum miR-132-3p, which gave rise to a receiver operator curve area of under the curve (ROC AUC) of 0.89. The number of studied subjects in the studies by Zhao et al. (2011) and Zhou et al. (2019) is more than twice the numbers studies by Gillet et al. (2019) and by Tagoma et al. (2018), and it seems most likely that miR-132-3p is downregulated in circulation in GDM. The downregulation of miR-132-3p in GDM is interesting given the observation that silencing miR-132-3p using antagomir injections in mice increased β-cell insulin secretion and lowered blood glucose (Bijkerk et al. 2019).

Placenta-derived extracellular vesicle-associated miRNAs as biomarkers of GDM

The amount of EVs in circulation increases during gestation, as well as the number of exosomes derived from the placenta, carrying the placental alkaline phosphatase marker PLAP (Salomon et al. 2016), and numbers of PLAP+ exosomes from GDM patients were more proinflammatory and increased significantly compared with control women during gestation (Salomon et al. 2016, Liu et al. 2018). Physiologically, small (s) EVs from pregnant women play a role in regulating insulin sensitivity, as mice infused with sEVs isolated from pregnant women in second trimester displayed decreased insulin sensitivity, whereas sEVs from GDM patients induced overt glucose intolerance in mice (James-Allan et al. 2020). Moreover, placental exosomes control insulin sensitivity, possibly via their content of miRNAs (Nair et al. 2018). miR-92a-3p was suggested as a mediator of these responses (Nair et al. 2021). Of note, miR-92a-5p is part of the same miRNA cluster (miR17HG) also containing miR-17-5p and miR-19a/b-3p (https://genome.ucsc.edu) and share regulatory sequences. It is possible that the entire miRNA cluster could be relevant for GDM diagnosis or prognosis either measured in plasma or in isolated EVs from the placenta.

Conclusions and perspectives

In this review, we aimed to collect and discuss the published literature for circulating miRNAs being associated with GDM to determine if miRNAs, by currently available information, could potentially be biomarkers for early detection of GDM. Although we initially searched for studies investigating all types of small RNAs, identified original studies were generally focused on miRNAs, which therefore became the focus of this review; however, other types of small RNAs, such as piwi-associated RNAs and tRNA fragments are also present in the circulation and could also be possible biomarker candidates. There are relatively few miRNAs for which levels in circulation are consistently associated with GDM: miR-29a/b-3p and miR-16-5p, whereas other reported miRNAs are not consistently associated with GDM. miR-29a/b-3p and miR-16-5p generally have good discriminatory power to detect GDM, and they may also have predictive capabilities for GDM when measured early in gestation.

It is clear from the literature that there is large heterogeneity between studies with regard to miRNAs being associated with GDM, with only few miRNAs being convincingly and reliably associated with GDM. Both methodological and clinical factors are likely to contribute to the diversity in findings. Methodological factors can be divided into pre- and post-analytical factors. Differences between studies with regard to pre-analytical factors such as sample type (serum, plasma or full blood), sample collection and processing (i.e. centrifugation times), RNA-extraction methods and quality control, as well as the qRT-PCR-assay can all result in variability between studies. Moreover, the main challenge of post-analytical level is data normalization as qRT-PCR relative quantification is often used (de Gonzalo-Calvo et al. 2022). For measurements of circulating miRNAs, no standardized protocols exist. Consensus guidelines for the development of assays for circulating RNAs underscore the importance of standardized assays and emphasize the necessity of validation at all steps of an RNA measurement from how the sample is treated, the RNA extracted and measured (i.e. using qRT-PCR) as well as to how the data are analyzed and reported (Acuna-Alonzo et al. 2010).

Clinical factors also contribute to the disparity between studies because cohorts may be recruited from different underlying patient populations, through different inclusion and exclusion criteria, and specifically for GDM, diagnostic criteria differ between countries creating a lack of comparability between studies. Furthermore, as miRNA levels change during gestation, the gestational sampling time point also offers a source of heterogeneity between studies (Sorensen et al. 2022). Furthermore, due to the discovery nature of many biomarker studies, a general flaw is the risk of them being underpowered, which promotes publication bias and lack of reproducibility (Ioannidis et al. 2014). Thus, to improve the likelihood of circulating miRNAs to be used in a clinically validated test in relation to GDM, increased transparency of study reporting, standardized workflows, assays and rigorous data analytical pipelines are needed (de Gonzalo-Calvo et al. 2022). Moreover, there is a need for systematic reviews and meta-analyses on circulating miRNAs in GDM in order to extract data from the existing literature in a systematic and unbiased manner.

In order to proceed with these miRNAs as diagnostic or prognostic markers for GDM, further studies are necessary, in larger cohorts and in patients with other pregnancy-associated morbidities such as pre-eclampsia. In order for an miRNA to be developed into a biomarker, it is also necessary to identify a population baseline to establish if the miRNA is specific for GDM. Moreover, it seems likely that miRNAs may be used as GDM biomarkers in combination with each other because the combination of several miRNAs will yield a more specific algorithm with a higher ROC AUC.

There are indications that circulating RNA markers are entering the clinic, i.e. there is a marketed prediction algorithm (NIS4, offered by Genfit) for the development of NASH or NASH-related fibrosis that involves miRNA-34a in combination with alpha-2 macroglobulin, YKL-40, and glycated hemoglobin (Harrison et al. 2020). Moreover, within the cancer field, the Thyramir miRNA 11 miRNA-based classifier for thyroid nodules is offered by Interpace Diagnostics and shows superior performance (Finkelstein et al. 2022). However, there are no clinically validated and marketed commercial tests based on miRNAs for predicting future GDM, based on an early sample, although such a test would be very valuable in the clinical management of women at risk of GDM.

As GDM share many features with T2D, it would also be of interest to investigate levels of the GDM-associated miRNAs in subjects with impaired glucose tolerance to determine if these would be able to predict later progression to overt T2D. Moreover, by detecting the risk of GDM earlier in pregnancy, it will be possible to initiate preventive measures and treatment earlier to prevent exposure of the fetus to extended periods of hyperglycemia. The use of miRNA as a diagnostic biomarker from a patient perspective could be beneficial because miRNA is easy to measure, as it can be done with an ordinary blood test, which for most would be preferable to an OGTT.

Another interesting question is from which tissue differentially regulated RNAs originate. If we could pinpoint the tissue or cell type of origin of the RNAs differentially regulated in GDM, this would provide important pathophysiological and novel information about the RNA secretion patterns in pregnancy and in GDM.

Declaration of interest

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

Funding

There is no funding to declare.

Author contribution statement

LTD conceived the study, researched the data and wrote the manuscript. SD and AE-F researched the data and wrote the manuscript. All authors approved the final version.

References

  • Acuna-Alonzo V, Flores-Dorantes T, Kruit JK, Villarreal-Molina T, Arellano-Campos O, Hunemeier T, Moreno-Estrada A, Ortiz-Lopez MG, Villamil-Ramirez H & Leon-Mimila P et al.2010 A functional ABCA1 gene variant is associated with low HDL-cholesterol levels and shows evidence of positive selection in Native Americans. Human Molecular Genetics 19 28772885. (https://doi.org/10.1093/hmg/ddq173)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Albanese M, Chen YA, Huls C, Gartner K, Tagawa T, Mejias-Perez E, Keppler OT, Gobel C, Zeidler R & Shein M et al.2021 MicroRNAs are minor constituents of extracellular vesicles that are rarely delivered to target cells. PLOS Genetics 17 e1009951. (https://doi.org/10.1371/journal.pgen.1009951)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Arroyo JD, Chevillet JR, Kroh EM, Ruf IK, Pritchard CC, Gibson DF, Mitchell PS, Bennett CF, Pogosova-Agadjanyan EL & Stirewalt DL et al.2011 Argonaute2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma. Proceedings of the National Academy of Sciences of the United States of America 108 50035008. (https://doi.org/10.1073/pnas.1019055108)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Barman B & Bhattacharyya SN 2015 mRNA targeting to endoplasmic reticulum precedes ago protein interaction and microRNA (miRNA)-mediated translation repression in mammalian cells. Journal of Biological Chemistry 290 2465024656. (https://doi.org/10.1074/jbc.C115.661868)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Barman B, Sung BH, Krystofiak E, Ping J, Ramirez M, Millis B, Allen R, Prasad N, Chetyrkin S & Calcutt MW et al.2022 VAP-A and its binding partner CERT drive biogenesis of RNA-containing extracellular vesicles at ER membrane contact sites. Developmental Cell 57 974–994.e8. (https://doi.org/10.1016/j.devcel.2022.03.012)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bartel DP & Chen CZ 2004 Micromanagers of gene expression: the potentially widespread influence of metazoan microRNAs. Nature Reviews. Genetics 5 396400. (https://doi.org/10.1038/nrg1328)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bijkerk R, Esguerra JLS, Ellenbroek JH, Au YW, Hanegraaf Maj, De Koning EJ, Eliasson L & Van Zonneveld AJ 2019 In vivo silencing of MicroRNA-132 reduces blood glucose and improves insulin secretion. Nucleic Acid Therapeutics 29 6772. (https://doi.org/10.1089/nat.2018.0763)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Boon RA & Vickers KC 2013 Intercellular transport of microRNAs. Arteriosclerosis, Thrombosis, and Vascular Biology 33 186192. (https://doi.org/10.1161/ATVBAHA.112.300139)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cao YL, Jia YJ, Xing BH, Shi DD & Dong XJ 2017 Plasma microRNA-16-5p, -17-5p and -20a-5p: novel diagnostic biomarkers for gestational diabetes mellitus. Journal of Obstetrics and Gynaecology Research 43 974981. (https://doi.org/10.1111/jog.13317)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chendrimada TP, Gregory RI, Kumaraswamy E, Norman J, Cooch N, Nishikura K & Shiekhattar R 2005 TRBP recruits the dicer complex to Ago2 for microRNA processing and gene silencing. Nature 436 740744. (https://doi.org/10.1038/nature03868)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cheung R, Pizza G, Chabosseau P, Rolando D, Tomas A, Burgoyne T, Wu Z, Salowka A, Tapa A & Macklin A et al.2022 Glucose-dependent miR-125b is a negative regulator of beta-cell function. Diabetes 71 15251545. (https://doi.org/10.2337/db21-0803)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chevillet JR, Kang Q, Ruf IK, Briggs HA, Vojtech LN, Hughes SM, Cheng HH, Arroyo JD, Meredith EK & Gallichotte EN et al.2014 Quantitative and stoichiometric analysis of the microRNA content of exosomes. Proceedings of the National Academy of Sciences of the United States of America 111 1488814893. (https://doi.org/10.1073/pnas.1408301111)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chu Y, Yue X, Younger ST, Janowski BA & Corey DR 2010 Involvement of argonaute proteins in gene silencing and activation by RNAs complementary to a non-coding transcript at the progesterone receptor promoter. Nucleic Acids Research 38 77367748. (https://doi.org/10.1093/nar/gkq648)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Clancy JW, Zhang Y, Sheehan C & D'souza-Schorey C 2019 An ARF6-Exportin-5 axis delivers pre-miRNA cargo to tumour microvesicles. Nature Cell Biology 21 856866. (https://doi.org/10.1038/s41556-019-0345-y)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Condrat CE, Thompson DC, Barbu MG, Bugnar OL, Boboc A, Cretoiu D, Suciu N, Cretoiu SM & Voinea SC 2020 miRNAs as biomarkers in disease: latest findings regarding their role in diagnosis and prognosis. Cells 9 276. (https://doi.org/10.3390/cells9020276)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dalgaard LT, Sorensen AE, Hardikar AA & Joglekar MV 2022 The microRNA-29 family: role in metabolism and metabolic disease. American Journal of Physiology. Cell Physiology 323 C367C377. (https://doi.org/10.1152/ajpcell.00051.2022)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Damm P, Houshmand-Oeregaard A, Kelstrup L, Lauenborg J, Mathiesen ER & Clausen TD 2016 Gestational diabetes mellitus and long-term consequences for mother and offspring: a view from Denmark. Diabetologia 59 13961399. (https://doi.org/10.1007/s00125-016-3985-5)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • de Gonzalo-Calvo D, Marchese M, Hellemans J, Betsou F, Skov Frisk NL, Dalgaard LT, Lakkisto P, Foy C, Scherer A & Garcia Bermejo ML et al.2022 Consensus guidelines for the validation of qRT-PCR assays in clinical research by the CardioRNA consortium. Molecular Therapy. Methods and Clinical Development 24 171180. (https://doi.org/10.1016/j.omtm.2021.12.007)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Eichhorn SW, Guo H, Mcgeary SE, Rodriguez-Mias RA, Shin C, Baek D, Hsu SH, Ghoshal K, Villen J & Bartel DP 2014 mRNA destabilization is the dominant effect of mammalian microRNAs by the time substantial repression ensues. Molecular Cell 56 104115. (https://doi.org/10.1016/j.molcel.2014.08.028)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Eliasson L & Esguerra JLS 2020 MicroRNA networks in pancreatic islet cells: normal function and Type 2 diabetes. Diabetes 69 804812. (https://doi.org/10.2337/dbi19-0016)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Filipowicz W & Sonenberg N 2015 The long unfinished march towards understanding microRNA-mediated repression. RNA 21 519524. (https://doi.org/10.1261/rna.051219.115)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Filardi T, Catanzaro G, Grieco GE, Splendiani E, Trocchianesi S, Santangelo C, Brunelli R, Guarino E, Sebastiani G & Dotta F et al. 2022 Identification and validation of miR-222-3p and miR-409-3p as plasma biomarkers in gestational diabetes mellitus sharing validated target genes involved in metabolic homeostasis. International Journal of Molecular Science 23 4276. (https://doi.org/10.3390/ijms23084276)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Finkelstein SD, Sistrunk JW, Malchoff C, Thompson DV, Kumar G, Timmaraju VA, Repko B, Mireskandari A, Evoy-Goodman LA & Massoll NA et al.2022 A retrospective evaluation of the diagnostic performance of an interdependent pairwise microRNA expression analysis with a mutation panel in indeterminate thyroid nodules. Thyroid 32 13621371. (https://doi.org/10.1089/thy.2022.0124)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gillet V, Ouellet A, Stepanov Y, Rodosthenous RS, Croft EK, Brennan K, Abdelouahab N, Baccarelli A & Takser L 2019 miRNA profiles in extracellular vesicles from serum early in pregnancies complicated by gestational diabetes mellitus. Journal of Clinical Endocrinology and Metabolism 104 51575169. (https://doi.org/10.1210/jc.2018-02693)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Guduric-Fuchs J, O'connor A, Camp B, O'neill CL, Medina RJ & Simpson DA 2012 Selective extracellular vesicle-mediated export of an overlapping set of microRNAs from multiple cell types. BMC Genomics 13 357. (https://doi.org/10.1186/1471-2164-13-357)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Guo H, Ingolia NT, Weissman JS & Bartel DP 2010 Mammalian microRNAs predominantly act to decrease target mRNA levels. Nature 466 835840. (https://doi.org/10.1038/nature09267)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Harrison SA, Ratziu V, Boursier J, Francque S, Bedossa P, Majd Z, Cordonnier G, Sudrik FB, Darteil R & Liebe R et al.2020 A blood-based biomarker panel (NIS4) for non-invasive diagnosis of non-alcoholic steatohepatitis and liver fibrosis: a prospective derivation and global validation study. Lancet. Gastroenterology and Hepatology 5 970985. (https://doi.org/10.1016/S2468-1253(2030252-1)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Herrera BM, Lockstone HE, Taylor JM, Wills QF, Kaisaki PJ, Barrett A, Camps C, Fernandez C, Ragoussis J & Gauguier D et al.2009 MicroRNA-125a is over-expressed in insulin target tissues in a spontaneous rat model of Type 2 Diabetes. BMC Medical Genomics 2 54. (https://doi.org/10.1186/1755-8794-2-54)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hocaoglu M, Demirer S, Senturk H, Turgut A & Komurcu-Bayrak E 2019 Differential expression of candidate circulating microRNAs in maternal blood leukocytes of the patients with preeclampsia and gestational diabetes mellitus. Pregnancy Hypertension 17 511. (https://doi.org/10.1016/j.preghy.2019.04.004)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hung YH, Kanke M, Kurtz CL, Cubitt R, Bunaciu RP, Miao J, Zhou L, Graham JL, Hussain MM & Havel P et al.2019 Acute suppression of insulin resistance-associated hepatic miR-29 in vivo improves glycemic control in adult mice. Physiological Genomics 51 379389. (https://doi.org/10.1152/physiolgenomics.00037.2019)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • International Association of Diabetes and Pregnancy Study Groups, Metzger BE, Gabbe SG, Persson B, Buchanan TA, Catalano PA, Damm P, Dyer AR & Leiva A et al.2010 International Association of Diabetes and Pregnancy Study Groups recommendations on the diagnosis and classification of hyperglycemia in pregnancy. Diabetes Care 33 6 7 66 8 2. (https://doi.org/10.2337/dc09-1848)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • International Association of Diabetes in Pregnancy Study Group (IADPSG) Working Group on Outcome Definitions, Feig DS, Corcoy R, Jensen DM, Kautzky-Willer A, Nolan CJ, Oats JJ, Sacks DA, Caimari F & McIntyre HD 2015 Diabetes in pregnancy outcomes: a systematic review and proposed codification of definitions. Diabetes/Metabolism Research and Reviews 31 680690. (https://doi.org/10.1002/dmrr.2640)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ioannidis JP, Greenland S, Hlatky MA, Khoury MJ, Macleod MR, Moher D, Schulz KF & Tibshirani R 2014 Increasing value and reducing waste in research design, conduct, and analysis. Lancet 383 166175. (https://doi.org/10.1016/S0140-6736(1362227-8)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • James-Allan LB, Rosario FJ, Barner K, Lai A, Guanzon D, Mcintyre HD, Lappas M, Powell TL, Salomon C & Jansson T 2020 Regulation of glucose homeostasis by small extracellular vesicles in normal pregnancy and in gestational diabetes. FASEB Journal 34 57245739. (https://doi.org/10.1096/fj.201902522RR)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jankauskas SS, Gambardella J, Sardu C, Lombardi A & Santulli G 2021 Functional role of miR-155 in the pathogenesis of diabetes mellitus and its complications. Non-Coding RNA 7 39. (https://doi.org/10.3390/ncrna7030039)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kampmann U, Knorr S, Fuglsang J & Ovesen P 2019 Determinants of maternal insulin resistance during pregnancy: an updated overview. Journal of Diabetes Research 2019 5320156. (https://doi.org/10.1155/2019/5320156)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kelstrup L, Clausen TD, Mathiesen ER, Hansen T & Damm P 2012 Low-grade inflammation in young adults exposed to intrauterine hyperglycemia. Diabetes Research and Clinical Practice 97 322330. (https://doi.org/10.1016/j.diabres.2012.04.023)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kelstrup L, Damm P, Mathiesen ER, Hansen T, Vaag AA, Pedersen O & Clausen TD 2013 Insulin resistance and impaired pancreatic beta-cell function in adult offspring of women with diabetes in pregnancy. Journal of Clinical Endocrinology and Metabolism 98 37933801. (https://doi.org/10.1210/jc.2013-1536)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kosaka N, Iguchi H, Yoshioka Y, Takeshita F, Matsuki Y & Ochiya T 2010 Secretory mechanisms and intercellular transfer of microRNAs in living cells. Journal of Biological Chemistry 285 1744217452. (https://doi.org/10.1074/jbc.M110.107821)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kozomara A, Birgaoanu M & Griffiths-Jones S 2019 miRBase: from microRNA sequences to function. Nucleic Acids Research 47 D155D162. (https://doi.org/10.1093/nar/gky1141)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lamadrid-Romero M, Solis KH, Cruz-Resendiz MS, Perez JE, Diaz NF, Flores-Herrera H, Garcia-Lopez G, Perichart O, Reyes-Munoz E & Arenas-Huertero F et al.2018 Central nervous system development-related microRNAs levels increase in the serum of gestational diabetic women during the first trimester of pregnancy. Neuroscience Research 130 822. (https://doi.org/10.1016/j.neures.2017.08.003)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li Y, Deng S, Peng J, Wang X, Essandoh K, Mu X, Peng T, Meng ZX & Fan GC 2019 MicroRNA-223 is essential for maintaining functional beta-cell mass during diabetes through inhibiting both FOXO1 and SOX6 pathways. Journal of Biological Chemistry 294 1043810448. (https://doi.org/10.1074/jbc.RA119.007755)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lim S, Deaver JW, Rosa-Caldwell ME, Lee DE, Morena Da Silva F, Cabrera AR, Schrems ER, Saling LW, Washington Ta & Fluckey JD et al.2022 Muscle miR-16 deletion results in impaired insulin sensitivity and contractile function in a sex-dependent manner. American Journal of Physiology. Endocrinology and Metabolism 322 E278E292. (https://doi.org/10.1152/ajpendo.00333.2021)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu J, Wang SZ, Wang QL, Du JG & Wang BB 2018 Gestational diabetes mellitus is associated with changes in the concentration and bioactivity of placental exosomes in the maternal circulation across gestation. European Review for Medical and Pharmacological Sciences 22 20362043. (https://doi.org/10.26355/eurrev_201804_14733)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu R, Wang M, Li E, Yang Y, Li J, Chen S, Shen WJ, Azhar S, Guo Z & Hu Z 2020 Dysregulation of microRNA-125a contributes to obesity-associated insulin resistance and dysregulates lipid metabolism in mice. Biochimica et Biophysica Acta. Molecular and Cell Biology of Lipids 1865 158640. (https://doi.org/10.1016/j.bbalip.2020.158640)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu S, Tang G, Duan F, Zeng C, Gong J, Chen Y & Tan H 2021 MiR-17-5p inhibits TXNIP/NLRP3 inflammasome pathway and suppresses pancreatic beta-cell pyroptosis in diabetic mice. Frontiers in Cardiovascular Medicine 8 768029. (https://doi.org/10.3389/fcvm.2021.768029)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ludwig N, Leidinger P, Becker K, Backes C, Fehlmann T, Pallasch C, Rheinheimer S, Meder B, Stahler C & Meese E et al.2016 Distribution of miRNA expression across human tissues. Nucleic Acids Research 44 38653877. (https://doi.org/10.1093/nar/gkw116)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ma X, Becker Buscaglia LE, Barker JR & Li Y 2011 MicroRNAs in NF-kappaB signaling. Journal of Molecular Cell Biology 3 159166. (https://doi.org/10.1093/jmcb/mjr007)

  • Mandelbaum AD, Kredo-Russo S, Aronowitz D, Myers N, Yanowski E, Klochendler A, Swisa A, Dor Y & Hornstein E 2019 miR-17-92 and miR-106b-25 clusters regulate beta cell mitotic checkpoint and insulin secretion in mice. Diabetologia 62 16531666. (https://doi.org/10.1007/s00125-019-4916-z)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Martinez-Ibarra A, Martinez-Razo LD, Vazquez-Martinez ER, Martinez-Cruz N, Flores-Ramirez R, Garcia-Gomez E, Lopez-Lopez M, Ortega-Gonzalez C, Camacho-Arroyo I & Cerbon M 2019 Unhealthy levels of phthalates and bisphenol A in Mexican pregnant women with gestational diabetes and its association to altered expression of miRNAs involved with metabolic disease. International Journal of Molecular Sciences 20. (https://doi.org/10.3390/ijms20133343)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mathys H, Basquin J, Ozgur S, Czarnocki-Cieciura M, Bonneau F, Aartse A, Dziembowski A, Nowotny M, Conti E & Filipowicz W 2014 Structural and biochemical insights to the role of the CCR4-NOT complex and DDX6 ATPase in microRNA repression. Molecular Cell 54 751765. (https://doi.org/10.1016/j.molcel.2014.03.036)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Menon R, Debnath C, Lai A, Guanzon D, Bhatnagar S, Kshetrapal PK, Sheller-Miller S, Salomon C & Garbhini Study Team 2019 Circulating exosomal miRNA profile during term and preterm birth pregnancies: a longitudinal study. Endocrinology 160 249275. (https://doi.org/10.1210/en.2018-00836)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Moura J, Sorensen A, Leal EC, Svendsen R, Carvalho L, Willemoes RJ, Jorgensen PT, Jenssen H, Wengel J & Dalgaard LT et al.2019 microRNA-155 inhibition restores fibroblast growth factor 7 expression in diabetic skin and decreases wound inflammation. Scientific Reports 9 5836. (https://doi.org/10.1038/s41598-019-42309-4)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mziaut H, Henniger G, Ganss K, Hempel S, Wolk S, Mcchord J, Chowdhury K, Ravassard P, Knoch KP & Krautz C et al.2020 MiR-132 controls pancreatic beta cell proliferation and survival through Pten/Akt/Foxo3 signaling. Molecular Metabolism 31 150162. (https://doi.org/10.1016/j.molmet.2019.11.012)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Nair S, Guanzon D, Jayabalan N, Lai A, Scholz-Romero K, Kalita de Croft P, Ormazabal V, Palma C, Diaz E & Mccarthy EA et al.2021 Extracellular vesicle-associated miRNAs are an adaptive response to gestational diabetes mellitus. Journal of Translational Medicine 19 360. (https://doi.org/10.1186/s12967-021-02999-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Nair S, Jayabalan N, Guanzon D, Palma C, Scholz-Romero K, Elfeky O, Zuniga F, Ormazabal V, Diaz E & Rice GE et al.2018 Human placental exosomes in gestational diabetes mellitus carry a specific set of miRNAs associated with skeletal muscle insulin sensitivity. Clinical Science 132 24512467. (https://doi.org/10.1042/CS20180487)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Nesca V, Guay C, Jacovetti C, Menoud V, Peyot ML, Laybutt DR, Prentki M & Regazzi R 2013 Identification of particular groups of microRNAs that positively or negatively impact on beta cell function in obese models of type 2 diabetes. Diabetologia 56 22032212. (https://doi.org/10.1007/s00125-013-2993-y)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Peng H-Y, Li H-P & & Li M-Q 2018 High glucose induces dysfunction of human umbilical vein endothelial cells by upregulating miR-137 in gestational diabetes mellitus. Microvascular Research 118 90-100. (https://doi.org/10.1016/j.mvr.2018.03.002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pfeiffer S, Sanchez-Lechuga B, Donovan P, Halang L, Prehn JHM, Campos-Caro A, Byrne MM & Lopez-Tinoco C 2020 Circulating miR-330-3p in late pregnancy is associated with pregnancy outcomes among lean women with GDM. Scientific Reports 10 908. (https://doi.org/10.1038/s41598-020-57838-6)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pinto SK, Lamon S, Stephenson EJ, Kalanon M, Mikovic J, Koch LG, Britton SL, Hawley JA & Camera DM 2017 Expression of microRNAs and target proteins in skeletal muscle of rats selectively bred for high and low running capacity. American Journal of Physiology. Endocrinology and Metabolism 313 E335E343. (https://doi.org/10.1152/ajpendo.00043.2017)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Qi S & & Wang X 2019 Decreased expression of miR-185 in serum and placenta of patients with gestational diabetes mellitus. Clinical Laboratory 65 [epub]. (https://doi.org/10.7754/Clin.Lab.2019.190445)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rani PR & Begum J 2016 Screening and diagnosis of gestational diabetes mellitus, where do we stand. Journal of Clinical and Diagnostic Research 10 QE01QE04. (https://doi.org/10.7860/JCDR/2016/17588.7689)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Salomon C, Nuzhat Z, Dixon CL & Menon R 2018 Placental exosomes during gestation: liquid biopsies carrying signals for the regulation of human parturition. Current Pharmaceutical Design 24 974982. (https://doi.org/10.2174/1381612824666180125164429)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Salomon C, Scholz-Romero K, Sarker S, Sweeney E, Kobayashi M, Correa P, Longo S, Duncombe G, Mitchell MD & Rice GE et al.2016 Gestational diabetes mellitus is associated with changes in the concentration and bioactivity of placenta-derived exosomes in maternal circulation across gestation. Diabetes 65 598609. (https://doi.org/10.2337/db15-0966)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sanchez-Ceinos J, Rangel-Zuniga OA, Clemente-Postigo M, Podadera-Herreros A, Camargo A, Alcala-Diaz JF, Guzman-Ruiz R, Lopez-Miranda J & Malagon MM 2021 miR-223-3p as a potential biomarker and player for adipose tissue dysfunction preceding type 2 diabetes onset. Molecular Therapy. Nucleic Acids 23 10351052. (https://doi.org/10.1016/j.omtn.2021.01.014)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sebastiani G, Guarino E, Grieco GE, Formichi C, Delli Poggi C, Ceccarelli E & Dotta F 2017 Circulating microRNA (miRNA) expression profiling in plasma of patients with gestational diabetes mellitus reveals upregulation of miRNA miR-330-3p. Frontiers in Endocrinology (Lausanne) 8 345. (https://doi.org/10.3389/fendo.2017.00345)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sorensen AE, Van Poppel MNM, Desoye G, Damm P, Simmons D, Jensen DM, Dalgaard LT, The DCI. 2021 The predictive value of miR-16, -29a and -134 for early identification of gestational diabetes: a nested analysis of the DALI cohort. Cells 10 170. (https://doi.org/10.3390/cells10010170)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sorensen AE, van Poppel MNM, Desoye G, Simmons D, Damm P, Jensen DM, Dalgaard LT & The Dali Core Investigator Group 2022 The temporal profile of circulating miRNAs during gestation in overweight and obese women with or without gestational diabetes mellitus. Biomedicines 10 482. (https://doi.org/10.3390/biomedicines10020482)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Squadrito ML, Baer C, Burdet F, Maderna C, Gilfillan GD, Lyle R, Ibberson M & De Palma M 2014 Endogenous RNAs modulate microRNA sorting to exosomes and transfer to acceptor cells. Cell Reports 8 14321446. (https://doi.org/10.1016/j.celrep.2014.07.035)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Stirm L, Huypens P, Sass S, Batra R, Fritsche L, Brucker S, Abele H, Hennige AM, Theis F & Beckers J et al.2018 Maternal whole blood cell miRNA-340 is elevated in gestational diabetes and inversely regulated by glucose and insulin. Scientific Reports 8 1366. (https://doi.org/10.1038/s41598-018-19200-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sun H, Saeedi P, Karuranga S, Pinkepank M, Ogurtsova K, Duncan BB, Stein C, Basit A, Chan JCN & Mbanya JC et al.2022 IDF Diabetes Atlas: global, regional and country-level diabetes prevalence estimates for 2021 and projections for 2045. Diabetes Research and Clinical Practice 183 109119. (https://doi.org/10.1016/j.diabres.2021.109119)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sun J, Huang Q, Li S, Meng F, Li X & Gong X 2018 miR-330-5p/Tim-3 axis regulates macrophage M2 polarization and insulin resistance in diabetes mice. Molecular Immunology 95 107113. (https://doi.org/10.1016/j.molimm.2018.02.006)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Tagoma A, Alnek K, Kirss A, Uibo R & Haller-Kikkatalo K 2018 MicroRNA profiling of second trimester maternal plasma shows upregulation of miR-195-5p in patients with gestational diabetes. Gene 672 137142. (https://doi.org/10.1016/j.gene.2018.06.004)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Teh WT, Teede HJ, Paul E, Harrison CL, Wallace EM & Allan C 2011 Risk factors for gestational diabetes mellitus: implications for the application of screening guidelines. Australian and New Zealand Journal of Obstetrics and Gynaecology 51 2630. (https://doi.org/10.1111/j.1479-828X.2011.01292.x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Vickers KC, Palmisano BT, Shoucri BM, Shamburek RD & Remaley AT 2011 MicroRNAs are transported in plasma and delivered to recipient cells by high-density lipoproteins. Nature Cell Biology 13 423433. (https://doi.org/10.1038/ncb2210)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Villard A, Marchand L, Thivolet C & Rome S 2015 Diagnostic value of cell-free circulating microRNAs for obesity and Type 2 diabetes: a meta-analysis. Journal of Molecular Biomarkers and Diagnosis 6 251. (https://doi.org/10.4172/2155-9929.1000251)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Villarroya-Beltri C, Gutierrez-Vazquez C, Sanchez-Cabo F, Perez-Hernandez D, Vazquez J, Martin-Cofreces N, Martinez-Herrera DJ, Pascual-Montano A, Mittelbrunn M & Sanchez-Madrid F 2013 SUMOylated hnRNPA2B1 controls the sorting of miRNAs into exosomes through binding to specific motifs. Nature Communications 4 2980. (https://doi.org/10.1038/ncomms3980)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wander PL, Boyko EJ, Hevner K, Parikh VJ, Tadesse MG, Sorensen TK, Williams MA & Enquobahrie DA 2017 Circulating early- and mid-pregnancy microRNAs and risk of gestational diabetes. Diabetes Research and Clinical Practice 132 19. (https://doi.org/10.1016/j.diabres.2017.07.024)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wei LM, Sun RP, Dong T, Liu J, Chen T, Zeng B, Wu JH, Luo JY, Sun JJ & Xi QY et al.2020 MiR-125b-2 knockout increases high-fat diet-induced fat accumulation and insulin resistance. Scientific Reports 10 21969. (https://doi.org/10.1038/s41598-020-77714-7)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Worm J, Stenvang J, Petri A, Frederiksen KS, Obad S, Elmen J, Hedtjarn M, Straarup EM, Hansen JB & Kauppinen S 2009 Silencing of microRNA-155 in mice during acute inflammatory response leads to derepression of c/ebp Beta and down-regulation of G-CSF. Nucleic Acids Research 37 57845792. (https://doi.org/10.1093/nar/gkp577)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wozniak AL, Adams A, King KE, Dunn W, Christenson LK, Hung WT & Weinman SA 2020 The RNA binding protein FMR1 controls selective exosomal miRNA cargo loading during inflammation. Journal of Cell Biology 219. (https://doi.org/10.1083/jcb.201912074)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Xiao Y, Ding J, Shi Y, Lin L, Huang W, Shen D & Wang W 2020 MiR-330-3p contributes to INS-1 cell dysfunction by targeting glucokinase in gestational diabetes mellitus. Journal of Obstetrics and Gynaecology Research 46 864875. (https://doi.org/10.1111/jog.14249)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yoffe L, Polsky A, Gilam A, Raff C, Mecacci F, Ognibene A, Crispi F, Gratacos E, Kanety H & Mazaki-Tovi S et al.2019 Early diagnosis of gestational diabetes mellitus using circulating microRNAs. European Journal of Endocrinology 181 565577. (https://doi.org/10.1530/EJE-19-0206)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zaccagnini G, Greco S, Voellenkle C, Gaetano C & Martelli F 2022 miR-210 hypoxamiR in angiogenesis and diabetes. Antioxidants and Redox Signaling 36 685706. (https://doi.org/10.1089/ars.2021.0200)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhang Y-L & Chen X-Q 2020 Dysregulation of microRNA-770-5p influences pancreatic-β-cell function by targeting TP53 regulated inhibitor of apoptosis 1 in gestational diabetes mellitus. European Review for Medical and Pharmacological Sciences 24 793-801. (https://doi.org/10.26355/eurrev_202001_20062)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhang L, Zhang T, Sun D, Cheng G, Ren H, Hong H, Chen L, Jiao X, Du Y & Zou Y et al.2021 Diagnostic value of dysregulated microribonucleic acids in the placenta and circulating exosomes in gestational diabetes mellitus. Journal of Diabetes Investigation 12 14901500. (https://doi.org/10.1111/jdi.13493)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhao C, Dong J, Jiang T, Shi Z, Yu B, Zhu Y, Chen D, Xu J, Huo R & Dai J et al.2011 Early second-trimester serum miRNA profiling predicts gestational diabetes mellitus. PLoS One 6 e23925. (https://doi.org/10.1371/journal.pone.0023925)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhou X, Xiang C & Zheng X 2019 miR-132 serves as a diagnostic biomarker in gestational diabetes mellitus and its regulatory effect on trophoblast cell viability. Diagnostic Pathology 14 119. (https://doi.org/10.1186/s13000-019-0899-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhu Y, Tian F, Li H, Zhou Y, Lu J & Ge Q 2015 Profiling maternal plasma microRNA expression in early pregnancy to predict gestational diabetes mellitus. International Journal of Gynaecology and Obstetrics 130 4953. (https://doi.org/10.1016/j.ijgo.2015.01.010)

    • PubMed
    • Search Google Scholar
    • Export Citation

 

  • Collapse
  • Expand
  • Figure 1

    miRNA and EV biogenesis. The canonical miRNA biogenesis starts as a primary miRNA transcript is formed in the nucleus by RNA polymerase II. Primary miRNA transcripts contain partially complementary hairpin structures. miRNA genes may be located as isolated transcriptional units or be located in exons or introns of protein-coding genes. DROSHA, a nuclease, removes the sequences outside the hairpin to form the precursor miRNA, in combination with the partner DGCR8. Subsequently, the miRNA precursor is exported to the cytoplasm by EXPORTIN 5 using RAN-GTP. Then, the nuclease DICER with TRBP removes the hairpin turn and the resulting duplex mature miRNA is unwound and inserts itself into the RISC complex. The miRNA–RISC complex then degrades or halts the transcription of mRNAs which are recognized by the miRNA. The RISC complex may dock to the ER membrane to load miRNAs into exosomes and multivesicular bodies. Extracellular RNA may also be released from cells in microvesicles that bud from the plasma membrane or may be bound by RNA-binding proteins, both of which will protect the miRNA in circulation from degradation. The figure was partly generated using Servier Medical Art, provided by Servier, licensed under a Creative Commons Attribution 3.0 unported license.

  • Figure 2

    Distribution of miRNAs examined in relation to GDM. The majority of original studies examining miRNAs for association with GDM were only examined in one published study, while miR-16-5p and miR-29a-3p were investigated in six studies each. Thus, the majority of reported circulating miRNAs associated with GDM were only identified in one study.

  • Acuna-Alonzo V, Flores-Dorantes T, Kruit JK, Villarreal-Molina T, Arellano-Campos O, Hunemeier T, Moreno-Estrada A, Ortiz-Lopez MG, Villamil-Ramirez H & Leon-Mimila P et al.2010 A functional ABCA1 gene variant is associated with low HDL-cholesterol levels and shows evidence of positive selection in Native Americans. Human Molecular Genetics 19 28772885. (https://doi.org/10.1093/hmg/ddq173)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Albanese M, Chen YA, Huls C, Gartner K, Tagawa T, Mejias-Perez E, Keppler OT, Gobel C, Zeidler R & Shein M et al.2021 MicroRNAs are minor constituents of extracellular vesicles that are rarely delivered to target cells. PLOS Genetics 17 e1009951. (https://doi.org/10.1371/journal.pgen.1009951)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Arroyo JD, Chevillet JR, Kroh EM, Ruf IK, Pritchard CC, Gibson DF, Mitchell PS, Bennett CF, Pogosova-Agadjanyan EL & Stirewalt DL et al.2011 Argonaute2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma. Proceedings of the National Academy of Sciences of the United States of America 108 50035008. (https://doi.org/10.1073/pnas.1019055108)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Barman B & Bhattacharyya SN 2015 mRNA targeting to endoplasmic reticulum precedes ago protein interaction and microRNA (miRNA)-mediated translation repression in mammalian cells. Journal of Biological Chemistry 290 2465024656. (https://doi.org/10.1074/jbc.C115.661868)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Barman B, Sung BH, Krystofiak E, Ping J, Ramirez M, Millis B, Allen R, Prasad N, Chetyrkin S & Calcutt MW et al.2022 VAP-A and its binding partner CERT drive biogenesis of RNA-containing extracellular vesicles at ER membrane contact sites. Developmental Cell 57 974–994.e8. (https://doi.org/10.1016/j.devcel.2022.03.012)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bartel DP & Chen CZ 2004 Micromanagers of gene expression: the potentially widespread influence of metazoan microRNAs. Nature Reviews. Genetics 5 396400. (https://doi.org/10.1038/nrg1328)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bijkerk R, Esguerra JLS, Ellenbroek JH, Au YW, Hanegraaf Maj, De Koning EJ, Eliasson L & Van Zonneveld AJ 2019 In vivo silencing of MicroRNA-132 reduces blood glucose and improves insulin secretion. Nucleic Acid Therapeutics 29 6772. (https://doi.org/10.1089/nat.2018.0763)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Boon RA & Vickers KC 2013 Intercellular transport of microRNAs. Arteriosclerosis, Thrombosis, and Vascular Biology 33 186192. (https://doi.org/10.1161/ATVBAHA.112.300139)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cao YL, Jia YJ, Xing BH, Shi DD & Dong XJ 2017 Plasma microRNA-16-5p, -17-5p and -20a-5p: novel diagnostic biomarkers for gestational diabetes mellitus. Journal of Obstetrics and Gynaecology Research 43 974981. (https://doi.org/10.1111/jog.13317)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chendrimada TP, Gregory RI, Kumaraswamy E, Norman J, Cooch N, Nishikura K & Shiekhattar R 2005 TRBP recruits the dicer complex to Ago2 for microRNA processing and gene silencing. Nature 436 740744. (https://doi.org/10.1038/nature03868)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cheung R, Pizza G, Chabosseau P, Rolando D, Tomas A, Burgoyne T, Wu Z, Salowka A, Tapa A & Macklin A et al.2022 Glucose-dependent miR-125b is a negative regulator of beta-cell function. Diabetes 71 15251545. (https://doi.org/10.2337/db21-0803)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chevillet JR, Kang Q, Ruf IK, Briggs HA, Vojtech LN, Hughes SM, Cheng HH, Arroyo JD, Meredith EK & Gallichotte EN et al.2014 Quantitative and stoichiometric analysis of the microRNA content of exosomes. Proceedings of the National Academy of Sciences of the United States of America 111 1488814893. (https://doi.org/10.1073/pnas.1408301111)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chu Y, Yue X, Younger ST, Janowski BA & Corey DR 2010 Involvement of argonaute proteins in gene silencing and activation by RNAs complementary to a non-coding transcript at the progesterone receptor promoter. Nucleic Acids Research 38 77367748. (https://doi.org/10.1093/nar/gkq648)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Clancy JW, Zhang Y, Sheehan C & D'souza-Schorey C 2019 An ARF6-Exportin-5 axis delivers pre-miRNA cargo to tumour microvesicles. Nature Cell Biology 21 856866. (https://doi.org/10.1038/s41556-019-0345-y)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Condrat CE, Thompson DC, Barbu MG, Bugnar OL, Boboc A, Cretoiu D, Suciu N, Cretoiu SM & Voinea SC 2020 miRNAs as biomarkers in disease: latest findings regarding their role in diagnosis and prognosis. Cells 9 276. (https://doi.org/10.3390/cells9020276)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dalgaard LT, Sorensen AE, Hardikar AA & Joglekar MV 2022 The microRNA-29 family: role in metabolism and metabolic disease. American Journal of Physiology. Cell Physiology 323 C367C377. (https://doi.org/10.1152/ajpcell.00051.2022)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Damm P, Houshmand-Oeregaard A, Kelstrup L, Lauenborg J, Mathiesen ER & Clausen TD 2016 Gestational diabetes mellitus and long-term consequences for mother and offspring: a view from Denmark. Diabetologia 59 13961399. (https://doi.org/10.1007/s00125-016-3985-5)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • de Gonzalo-Calvo D, Marchese M, Hellemans J, Betsou F, Skov Frisk NL, Dalgaard LT, Lakkisto P, Foy C, Scherer A & Garcia Bermejo ML et al.2022 Consensus guidelines for the validation of qRT-PCR assays in clinical research by the CardioRNA consortium. Molecular Therapy. Methods and Clinical Development 24 171180. (https://doi.org/10.1016/j.omtm.2021.12.007)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Eichhorn SW, Guo H, Mcgeary SE, Rodriguez-Mias RA, Shin C, Baek D, Hsu SH, Ghoshal K, Villen J & Bartel DP 2014 mRNA destabilization is the dominant effect of mammalian microRNAs by the time substantial repression ensues. Molecular Cell 56 104115. (https://doi.org/10.1016/j.molcel.2014.08.028)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Eliasson L & Esguerra JLS 2020 MicroRNA networks in pancreatic islet cells: normal function and Type 2 diabetes. Diabetes 69 804812. (https://doi.org/10.2337/dbi19-0016)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Filipowicz W & Sonenberg N 2015 The long unfinished march towards understanding microRNA-mediated repression. RNA 21 519524. (https://doi.org/10.1261/rna.051219.115)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Filardi T, Catanzaro G, Grieco GE, Splendiani E, Trocchianesi S, Santangelo C, Brunelli R, Guarino E, Sebastiani G & Dotta F et al. 2022 Identification and validation of miR-222-3p and miR-409-3p as plasma biomarkers in gestational diabetes mellitus sharing validated target genes involved in metabolic homeostasis. International Journal of Molecular Science 23 4276. (https://doi.org/10.3390/ijms23084276)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Finkelstein SD, Sistrunk JW, Malchoff C, Thompson DV, Kumar G, Timmaraju VA, Repko B, Mireskandari A, Evoy-Goodman LA & Massoll NA et al.2022 A retrospective evaluation of the diagnostic performance of an interdependent pairwise microRNA expression analysis with a mutation panel in indeterminate thyroid nodules. Thyroid 32 13621371. (https://doi.org/10.1089/thy.2022.0124)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gillet V, Ouellet A, Stepanov Y, Rodosthenous RS, Croft EK, Brennan K, Abdelouahab N, Baccarelli A & Takser L 2019 miRNA profiles in extracellular vesicles from serum early in pregnancies complicated by gestational diabetes mellitus. Journal of Clinical Endocrinology and Metabolism 104 51575169. (https://doi.org/10.1210/jc.2018-02693)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Guduric-Fuchs J, O'connor A, Camp B, O'neill CL, Medina RJ & Simpson DA 2012 Selective extracellular vesicle-mediated export of an overlapping set of microRNAs from multiple cell types. BMC Genomics 13 357. (https://doi.org/10.1186/1471-2164-13-357)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Guo H, Ingolia NT, Weissman JS & Bartel DP 2010 Mammalian microRNAs predominantly act to decrease target mRNA levels. Nature 466 835840. (https://doi.org/10.1038/nature09267)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Harrison SA, Ratziu V, Boursier J, Francque S, Bedossa P, Majd Z, Cordonnier G, Sudrik FB, Darteil R & Liebe R et al.2020 A blood-based biomarker panel (NIS4) for non-invasive diagnosis of non-alcoholic steatohepatitis and liver fibrosis: a prospective derivation and global validation study. Lancet. Gastroenterology and Hepatology 5 970985. (https://doi.org/10.1016/S2468-1253(2030252-1)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Herrera BM, Lockstone HE, Taylor JM, Wills QF, Kaisaki PJ, Barrett A, Camps C, Fernandez C, Ragoussis J & Gauguier D et al.2009 MicroRNA-125a is over-expressed in insulin target tissues in a spontaneous rat model of Type 2 Diabetes. BMC Medical Genomics 2 54. (https://doi.org/10.1186/1755-8794-2-54)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hocaoglu M, Demirer S, Senturk H, Turgut A & Komurcu-Bayrak E 2019 Differential expression of candidate circulating microRNAs in maternal blood leukocytes of the patients with preeclampsia and gestational diabetes mellitus. Pregnancy Hypertension 17 511. (https://doi.org/10.1016/j.preghy.2019.04.004)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hung YH, Kanke M, Kurtz CL, Cubitt R, Bunaciu RP, Miao J, Zhou L, Graham JL, Hussain MM & Havel P et al.2019 Acute suppression of insulin resistance-associated hepatic miR-29 in vivo improves glycemic control in adult mice. Physiological Genomics 51 379389. (https://doi.org/10.1152/physiolgenomics.00037.2019)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • International Association of Diabetes and Pregnancy Study Groups, Metzger BE, Gabbe SG, Persson B, Buchanan TA, Catalano PA, Damm P, Dyer AR & Leiva A et al.2010 International Association of Diabetes and Pregnancy Study Groups recommendations on the diagnosis and classification of hyperglycemia in pregnancy. Diabetes Care 33 6 7 66 8 2. (https://doi.org/10.2337/dc09-1848)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • International Association of Diabetes in Pregnancy Study Group (IADPSG) Working Group on Outcome Definitions, Feig DS, Corcoy R, Jensen DM, Kautzky-Willer A, Nolan CJ, Oats JJ, Sacks DA, Caimari F & McIntyre HD 2015 Diabetes in pregnancy outcomes: a systematic review and proposed codification of definitions. Diabetes/Metabolism Research and Reviews 31 680690. (https://doi.org/10.1002/dmrr.2640)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ioannidis JP, Greenland S, Hlatky MA, Khoury MJ, Macleod MR, Moher D, Schulz KF & Tibshirani R 2014 Increasing value and reducing waste in research design, conduct, and analysis. Lancet 383 166175. (https://doi.org/10.1016/S0140-6736(1362227-8)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • James-Allan LB, Rosario FJ, Barner K, Lai A, Guanzon D, Mcintyre HD, Lappas M, Powell TL, Salomon C & Jansson T 2020 Regulation of glucose homeostasis by small extracellular vesicles in normal pregnancy and in gestational diabetes. FASEB Journal 34 57245739. (https://doi.org/10.1096/fj.201902522RR)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jankauskas SS, Gambardella J, Sardu C, Lombardi A & Santulli G 2021 Functional role of miR-155 in the pathogenesis of diabetes mellitus and its complications. Non-Coding RNA 7 39. (https://doi.org/10.3390/ncrna7030039)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kampmann U, Knorr S, Fuglsang J & Ovesen P 2019 Determinants of maternal insulin resistance during pregnancy: an updated overview. Journal of Diabetes Research 2019 5320156. (https://doi.org/10.1155/2019/5320156)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kelstrup L, Clausen TD, Mathiesen ER, Hansen T & Damm P 2012 Low-grade inflammation in young adults exposed to intrauterine hyperglycemia. Diabetes Research and Clinical Practice 97 322330. (https://doi.org/10.1016/j.diabres.2012.04.023)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kelstrup L, Damm P, Mathiesen ER, Hansen T, Vaag AA, Pedersen O & Clausen TD 2013 Insulin resistance and impaired pancreatic beta-cell function in adult offspring of women with diabetes in pregnancy. Journal of Clinical Endocrinology and Metabolism 98 37933801. (https://doi.org/10.1210/jc.2013-1536)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kosaka N, Iguchi H, Yoshioka Y, Takeshita F, Matsuki Y & Ochiya T 2010 Secretory mechanisms and intercellular transfer of microRNAs in living cells. Journal of Biological Chemistry 285 1744217452. (https://doi.org/10.1074/jbc.M110.107821)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kozomara A, Birgaoanu M & Griffiths-Jones S 2019 miRBase: from microRNA sequences to function. Nucleic Acids Research 47 D155D162. (https://doi.org/10.1093/nar/gky1141)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lamadrid-Romero M, Solis KH, Cruz-Resendiz MS, Perez JE, Diaz NF, Flores-Herrera H, Garcia-Lopez G, Perichart O, Reyes-Munoz E & Arenas-Huertero F et al.2018 Central nervous system development-related microRNAs levels increase in the serum of gestational diabetic women during the first trimester of pregnancy. Neuroscience Research 130 822. (https://doi.org/10.1016/j.neures.2017.08.003)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li Y, Deng S, Peng J, Wang X, Essandoh K, Mu X, Peng T, Meng ZX & Fan GC 2019 MicroRNA-223 is essential for maintaining functional beta-cell mass during diabetes through inhibiting both FOXO1 and SOX6 pathways. Journal of Biological Chemistry 294 1043810448. (https://doi.org/10.1074/jbc.RA119.007755)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lim S, Deaver JW, Rosa-Caldwell ME, Lee DE, Morena Da Silva F, Cabrera AR, Schrems ER, Saling LW, Washington Ta & Fluckey JD et al.2022 Muscle miR-16 deletion results in impaired insulin sensitivity and contractile function in a sex-dependent manner. American Journal of Physiology. Endocrinology and Metabolism 322 E278E292. (https://doi.org/10.1152/ajpendo.00333.2021)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu J, Wang SZ, Wang QL, Du JG & Wang BB 2018 Gestational diabetes mellitus is associated with changes in the concentration and bioactivity of placental exosomes in the maternal circulation across gestation. European Review for Medical and Pharmacological Sciences 22 20362043. (https://doi.org/10.26355/eurrev_201804_14733)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu R, Wang M, Li E, Yang Y, Li J, Chen S, Shen WJ, Azhar S, Guo Z & Hu Z 2020 Dysregulation of microRNA-125a contributes to obesity-associated insulin resistance and dysregulates lipid metabolism in mice. Biochimica et Biophysica Acta. Molecular and Cell Biology of Lipids 1865 158640. (https://doi.org/10.1016/j.bbalip.2020.158640)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu S, Tang G, Duan F, Zeng C, Gong J, Chen Y & Tan H 2021 MiR-17-5p inhibits TXNIP/NLRP3 inflammasome pathway and suppresses pancreatic beta-cell pyroptosis in diabetic mice. Frontiers in Cardiovascular Medicine 8 768029. (https://doi.org/10.3389/fcvm.2021.768029)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ludwig N, Leidinger P, Becker K, Backes C, Fehlmann T, Pallasch C, Rheinheimer S, Meder B, Stahler C & Meese E et al.2016 Distribution of miRNA expression across human tissues. Nucleic Acids Research 44 38653877. (https://doi.org/10.1093/nar/gkw116)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ma X, Becker Buscaglia LE, Barker JR & Li Y 2011 MicroRNAs in NF-kappaB signaling. Journal of Molecular Cell Biology 3 159166. (https://doi.org/10.1093/jmcb/mjr007)

  • Mandelbaum AD, Kredo-Russo S, Aronowitz D, Myers N, Yanowski E, Klochendler A, Swisa A, Dor Y & Hornstein E 2019 miR-17-92 and miR-106b-25 clusters regulate beta cell mitotic checkpoint and insulin secretion in mice. Diabetologia 62 16531666. (https://doi.org/10.1007/s00125-019-4916-z)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Martinez-Ibarra A, Martinez-Razo LD, Vazquez-Martinez ER, Martinez-Cruz N, Flores-Ramirez R, Garcia-Gomez E, Lopez-Lopez M, Ortega-Gonzalez C, Camacho-Arroyo I & Cerbon M 2019 Unhealthy levels of phthalates and bisphenol A in Mexican pregnant women with gestational diabetes and its association to altered expression of miRNAs involved with metabolic disease. International Journal of Molecular Sciences 20. (https://doi.org/10.3390/ijms20133343)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mathys H, Basquin J, Ozgur S, Czarnocki-Cieciura M, Bonneau F, Aartse A, Dziembowski A, Nowotny M, Conti E & Filipowicz W 2014 Structural and biochemical insights to the role of the CCR4-NOT complex and DDX6 ATPase in microRNA repression. Molecular Cell 54 751765. (https://doi.org/10.1016/j.molcel.2014.03.036)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Menon R, Debnath C, Lai A, Guanzon D, Bhatnagar S, Kshetrapal PK, Sheller-Miller S, Salomon C & Garbhini Study Team 2019 Circulating exosomal miRNA profile during term and preterm birth pregnancies: a longitudinal study. Endocrinology 160 249275. (https://doi.org/10.1210/en.2018-00836)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Moura J, Sorensen A, Leal EC, Svendsen R, Carvalho L, Willemoes RJ, Jorgensen PT, Jenssen H, Wengel J & Dalgaard LT et al.2019 microRNA-155 inhibition restores fibroblast growth factor 7 expression in diabetic skin and decreases wound inflammation. Scientific Reports 9 5836. (https://doi.org/10.1038/s41598-019-42309-4)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mziaut H, Henniger G, Ganss K, Hempel S, Wolk S, Mcchord J, Chowdhury K, Ravassard P, Knoch KP & Krautz C et al.2020 MiR-132 controls pancreatic beta cell proliferation and survival through Pten/Akt/Foxo3 signaling. Molecular Metabolism 31 150162. (https://doi.org/10.1016/j.molmet.2019.11.012)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Nair S, Guanzon D, Jayabalan N, Lai A, Scholz-Romero K, Kalita de Croft P, Ormazabal V, Palma C, Diaz E & Mccarthy EA et al.2021 Extracellular vesicle-associated miRNAs are an adaptive response to gestational diabetes mellitus. Journal of Translational Medicine 19 360. (https://doi.org/10.1186/s12967-021-02999-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Nair S, Jayabalan N, Guanzon D, Palma C, Scholz-Romero K, Elfeky O, Zuniga F, Ormazabal V, Diaz E & Rice GE et al.2018 Human placental exosomes in gestational diabetes mellitus carry a specific set of miRNAs associated with skeletal muscle insulin sensitivity. Clinical Science 132 24512467. (https://doi.org/10.1042/CS20180487)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Nesca V, Guay C, Jacovetti C, Menoud V, Peyot ML, Laybutt DR, Prentki M & Regazzi R 2013 Identification of particular groups of microRNAs that positively or negatively impact on beta cell function in obese models of type 2 diabetes. Diabetologia 56 22032212. (https://doi.org/10.1007/s00125-013-2993-y)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Peng H-Y, Li H-P & & Li M-Q 2018 High glucose induces dysfunction of human umbilical vein endothelial cells by upregulating miR-137 in gestational diabetes mellitus. Microvascular Research 118 90-100. (https://doi.org/10.1016/j.mvr.2018.03.002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pfeiffer S, Sanchez-Lechuga B, Donovan P, Halang L, Prehn JHM, Campos-Caro A, Byrne MM & Lopez-Tinoco C 2020 Circulating miR-330-3p in late pregnancy is associated with pregnancy outcomes among lean women with GDM. Scientific Reports 10 908. (https://doi.org/10.1038/s41598-020-57838-6)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pinto SK, Lamon S, Stephenson EJ, Kalanon M, Mikovic J, Koch LG, Britton SL, Hawley JA & Camera DM 2017 Expression of microRNAs and target proteins in skeletal muscle of rats selectively bred for high and low running capacity. American Journal of Physiology. Endocrinology and Metabolism 313 E335E343. (https://doi.org/10.1152/ajpendo.00043.2017)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Qi S & & Wang X 2019 Decreased expression of miR-185 in serum and placenta of patients with gestational diabetes mellitus. Clinical Laboratory 65 [epub]. (https://doi.org/10.7754/Clin.Lab.2019.190445)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rani PR & Begum J 2016 Screening and diagnosis of gestational diabetes mellitus, where do we stand. Journal of Clinical and Diagnostic Research 10 QE01QE04. (https://doi.org/10.7860/JCDR/2016/17588.7689)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Salomon C, Nuzhat Z, Dixon CL & Menon R 2018 Placental exosomes during gestation: liquid biopsies carrying signals for the regulation of human parturition. Current Pharmaceutical Design 24 974982. (https://doi.org/10.2174/1381612824666180125164429)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Salomon C, Scholz-Romero K, Sarker S, Sweeney E, Kobayashi M, Correa P, Longo S, Duncombe G, Mitchell MD & Rice GE et al.2016 Gestational diabetes mellitus is associated with changes in the concentration and bioactivity of placenta-derived exosomes in maternal circulation across gestation. Diabetes 65 598609. (https://doi.org/10.2337/db15-0966)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sanchez-Ceinos J, Rangel-Zuniga OA, Clemente-Postigo M, Podadera-Herreros A, Camargo A, Alcala-Diaz JF, Guzman-Ruiz R, Lopez-Miranda J & Malagon MM 2021 miR-223-3p as a potential biomarker and player for adipose tissue dysfunction preceding type 2 diabetes onset. Molecular Therapy. Nucleic Acids 23 10351052. (https://doi.org/10.1016/j.omtn.2021.01.014)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sebastiani G, Guarino E, Grieco GE, Formichi C, Delli Poggi C, Ceccarelli E & Dotta F 2017 Circulating microRNA (miRNA) expression profiling in plasma of patients with gestational diabetes mellitus reveals upregulation of miRNA miR-330-3p. Frontiers in Endocrinology (Lausanne) 8 345. (https://doi.org/10.3389/fendo.2017.00345)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sorensen AE, Van Poppel MNM, Desoye G, Damm P, Simmons D, Jensen DM, Dalgaard LT, The DCI. 2021 The predictive value of miR-16, -29a and -134 for early identification of gestational diabetes: a nested analysis of the DALI cohort. Cells 10 170. (https://doi.org/10.3390/cells10010170)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sorensen AE, van Poppel MNM, Desoye G, Simmons D, Damm P, Jensen DM, Dalgaard LT & The Dali Core Investigator Group 2022 The temporal profile of circulating miRNAs during gestation in overweight and obese women with or without gestational diabetes mellitus. Biomedicines 10 482. (https://doi.org/10.3390/biomedicines10020482)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Squadrito ML, Baer C, Burdet F, Maderna C, Gilfillan GD, Lyle R, Ibberson M & De Palma M 2014 Endogenous RNAs modulate microRNA sorting to exosomes and transfer to acceptor cells. Cell Reports 8 14321446. (https://doi.org/10.1016/j.celrep.2014.07.035)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Stirm L, Huypens P, Sass S, Batra R, Fritsche L, Brucker S, Abele H, Hennige AM, Theis F & Beckers J et al.2018 Maternal whole blood cell miRNA-340 is elevated in gestational diabetes and inversely regulated by glucose and insulin. Scientific Reports 8 1366. (https://doi.org/10.1038/s41598-018-19200-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sun H, Saeedi P, Karuranga S, Pinkepank M, Ogurtsova K, Duncan BB, Stein C, Basit A, Chan JCN & Mbanya JC et al.2022 IDF Diabetes Atlas: global, regional and country-level diabetes prevalence estimates for 2021 and projections for 2045. Diabetes Research and Clinical Practice 183 109119. (https://doi.org/10.1016/j.diabres.2021.109119)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sun J, Huang Q, Li S, Meng F, Li X & Gong X 2018 miR-330-5p/Tim-3 axis regulates macrophage M2 polarization and insulin resistance in diabetes mice. Molecular Immunology 95 107113. (https://doi.org/10.1016/j.molimm.2018.02.006)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Tagoma A, Alnek K, Kirss A, Uibo R & Haller-Kikkatalo K 2018 MicroRNA profiling of second trimester maternal plasma shows upregulation of miR-195-5p in patients with gestational diabetes. Gene 672 137142. (https://doi.org/10.1016/j.gene.2018.06.004)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Teh WT, Teede HJ, Paul E, Harrison CL, Wallace EM & Allan C 2011 Risk factors for gestational diabetes mellitus: implications for the application of screening guidelines. Australian and New Zealand Journal of Obstetrics and Gynaecology 51 2630. (https://doi.org/10.1111/j.1479-828X.2011.01292.x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Vickers KC, Palmisano BT, Shoucri BM, Shamburek RD & Remaley AT 2011 MicroRNAs are transported in plasma and delivered to recipient cells by high-density lipoproteins. Nature Cell Biology 13 423433. (https://doi.org/10.1038/ncb2210)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Villard A, Marchand L, Thivolet C & Rome S 2015 Diagnostic value of cell-free circulating microRNAs for obesity and Type 2 diabetes: a meta-analysis. Journal of Molecular Biomarkers and Diagnosis 6 251. (https://doi.org/10.4172/2155-9929.1000251)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Villarroya-Beltri C, Gutierrez-Vazquez C, Sanchez-Cabo F, Perez-Hernandez D, Vazquez J, Martin-Cofreces N, Martinez-Herrera DJ, Pascual-Montano A, Mittelbrunn M & Sanchez-Madrid F 2013 SUMOylated hnRNPA2B1 controls the sorting of miRNAs into exosomes through binding to specific motifs. Nature Communications 4 2980. (https://doi.org/10.1038/ncomms3980)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wander PL, Boyko EJ, Hevner K, Parikh VJ, Tadesse MG, Sorensen TK, Williams MA & Enquobahrie DA 2017 Circulating early- and mid-pregnancy microRNAs and risk of gestational diabetes. Diabetes Research and Clinical Practice 132 19. (https://doi.org/10.1016/j.diabres.2017.07.024)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wei LM, Sun RP, Dong T, Liu J, Chen T, Zeng B, Wu JH, Luo JY, Sun JJ & Xi QY et al.2020 MiR-125b-2 knockout increases high-fat diet-induced fat accumulation and insulin resistance. Scientific Reports 10 21969. (https://doi.org/10.1038/s41598-020-77714-7)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Worm J, Stenvang J, Petri A, Frederiksen KS, Obad S, Elmen J, Hedtjarn M, Straarup EM, Hansen JB & Kauppinen S 2009 Silencing of microRNA-155 in mice during acute inflammatory response leads to derepression of c/ebp Beta and down-regulation of G-CSF. Nucleic Acids Research 37 57845792. (https://doi.org/10.1093/nar/gkp577)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wozniak AL, Adams A, King KE, Dunn W, Christenson LK, Hung WT & Weinman SA 2020 The RNA binding protein FMR1 controls selective exosomal miRNA cargo loading during inflammation. Journal of Cell Biology 219. (https://doi.org/10.1083/jcb.201912074)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Xiao Y, Ding J, Shi Y, Lin L, Huang W, Shen D & Wang W 2020 MiR-330-3p contributes to INS-1 cell dysfunction by targeting glucokinase in gestational diabetes mellitus. Journal of Obstetrics and Gynaecology Research 46 864875. (https://doi.org/10.1111/jog.14249)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yoffe L, Polsky A, Gilam A, Raff C, Mecacci F, Ognibene A, Crispi F, Gratacos E, Kanety H & Mazaki-Tovi S et al.2019 Early diagnosis of gestational diabetes mellitus using circulating microRNAs. European Journal of Endocrinology 181 565577. (https://doi.org/10.1530/EJE-19-0206)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zaccagnini G, Greco S, Voellenkle C, Gaetano C & Martelli F 2022 miR-210 hypoxamiR in angiogenesis and diabetes. Antioxidants and Redox Signaling 36 685706. (https://doi.org/10.1089/ars.2021.0200)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhang Y-L & Chen X-Q 2020 Dysregulation of microRNA-770-5p influences pancreatic-β-cell function by targeting TP53 regulated inhibitor of apoptosis 1 in gestational diabetes mellitus. European Review for Medical and Pharmacological Sciences 24 793-801. (https://doi.org/10.26355/eurrev_202001_20062)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhang L, Zhang T, Sun D, Cheng G, Ren H, Hong H, Chen L, Jiao X, Du Y & Zou Y et al.2021 Diagnostic value of dysregulated microribonucleic acids in the placenta and circulating exosomes in gestational diabetes mellitus. Journal of Diabetes Investigation 12 14901500. (https://doi.org/10.1111/jdi.13493)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhao C, Dong J, Jiang T, Shi Z, Yu B, Zhu Y, Chen D, Xu J, Huo R & Dai J et al.2011 Early second-trimester serum miRNA profiling predicts gestational diabetes mellitus. PLoS One 6 e23925. (https://doi.org/10.1371/journal.pone.0023925)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhou X, Xiang C & Zheng X 2019 miR-132 serves as a diagnostic biomarker in gestational diabetes mellitus and its regulatory effect on trophoblast cell viability. Diagnostic Pathology 14 119. (https://doi.org/10.1186/s13000-019-0899-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhu Y, Tian F, Li H, Zhou Y, Lu J & Ge Q 2015 Profiling maternal plasma microRNA expression in early pregnancy to predict gestational diabetes mellitus. International Journal of Gynaecology and Obstetrics 130 4953. (https://doi.org/10.1016/j.ijgo.2015.01.010)

    • PubMed
    • Search Google Scholar
    • Export Citation