Skip to main content
Endocrine Reviews logoLink to Endocrine Reviews
. 2017 Feb 17;38(2):145–168. doi: 10.1210/er.2016-1122

Emerging Roles for MicroRNAs in Diabetic Microvascular Disease: Novel Targets for Therapy

Yu Zhang 1,2, Xinghui Sun 1,3, Basak Icli 1, Mark W Feinberg 1,
PMCID: PMC5460677  PMID: 28323921

ESSENTIAL POINTS

  • Impaired microvascular function is a hallmark during the development of insulin resistance and in diabetic subjects

  • MicroRNAs, small noncoding RNAs that fine-tune gene expression, have emerged as critical regulators in insulin-responsive tissues and cell types important for maintaining vascular homeostasis and preventing the sequelae of diabetes-induced end organ injury

  • Current pathophysiological paradigms of microRNAs and their target genes demonstrate novel mechanisms and therapeutic targets in regulating the diabetic microvasculature in diabetes-associated complications such as retinopathy, nephropathy, wound healing, and myocardial injury

  • Circulating microRNAs in exosomes or microvesicles may be diagnostically useful in type I or type II diabetes

  • Emerging delivery platforms for manipulating microRNA expression or function may serve as the next frontier in therapeutic intervention to improve diabetes-associated microvascular dysfunction and its attendant clinical consequences

Abstract

Chronic, low-grade systemic inflammation and impaired microvascular function are critical hallmarks in the development of insulin resistance. Accordingly, insulin resistance is a major risk factor for type 2 diabetes and cardiovascular disease. Accumulating studies demonstrate that restoration of impaired function of the diabetic macro- and microvasculature may ameliorate a range of cardiovascular disease states and diabetes-associated complications. In this review, we focus on the emerging role of microRNAs (miRNAs), noncoding RNAs that fine-tune target gene expression and signaling pathways, in insulin-responsive tissues and cell types important for maintaining optimal vascular homeostasis and preventing the sequelae of diabetes-induced end organ injury. We highlight current pathophysiological paradigms of miRNAs and their targets involved in regulating the diabetic microvasculature in a range of diabetes-associated complications such as retinopathy, nephropathy, wound healing, and myocardial injury. We provide an update of the potential use of circulating miRNAs diagnostically in type I or type II diabetes. Finally, we discuss emerging delivery platforms for manipulating miRNA expression or function as the next frontier in therapeutic intervention to improve diabetes-associated microvascular dysfunction and its attendant clinical consequences.

I. Introduction

Diabetic subjects exhibit both accelerated macro- and microvasculature disease, an effect that significantly contributes to cardiovascular disease and diabetic-associated complications such as retinopathy, nephropathy, and neuropathy. Insulin resistance, a major risk factor for both type 2 diabetes and ischemic cardiovascular disease, is a common pathologic state caused by decreased sensitivity to insulin, a decrease in maximal response to insulin, or a combination of both (1, 2). It is central to the pathogenesis of type 2 diabetes and helps to maintain the diabetic state (3–7).

The recent recognition of microRNAs (miRNAs) as important mediators in the diabetic macro- and microvasculature has provided unique molecular and cellular insights into their impact on pathophysiological pathways in insulin resistance, type 2 diabetes, and obesity. Moreover, the appreciation that miRNAs can be detected extracellularly, such as in circulating blood or urine, raises the potential for their use as biomarkers for diagnosis and prognosis or in response to diabetic or cardiovascular therapeutics.

Herein, we focus on the emerging pathophysiological roles of miRNAs, evolutionarily conserved small noncoding RNAs of ∼22 nucleotides that fine-tune target gene expression and signaling pathways in insulin-responsive tissues and cell types important for maintaining optimal vascular homeostasis and preventing the complications of diabetes-induced end organ injury. We highlight current paradigms of miRNAs and their target genes involved in regulating the diabetic microvasculature in a range of diabetes-associated sequelae such as retinopathy, nephropathy, wound healing, and myocardial injury. We provide an update of the potential use of circulating miRNAs diagnostically in type 1 or type 2 diabetes. Finally, we discuss emerging delivery platforms for manipulation of miRNA expression or function as the next therapeutic frontier to improve diabetes-associated microvascular dysfunction and its attendant clinical consequences. We also review the potential challenges and opportunities for miRNAs as therapeutics in diabetic vascular disease.

II. Biogenesis and Function of miRNAs

MiRNAs are one class of small, evolutionarily conserved noncoding RNAs of ∼22 nucleotides in length that are involved in nearly all developmental and pathophysiological processes in animals as they target over one-half of protein-coding transcripts (8, 9). The first miRNA, Caenorhabditis elegans lineage-4 (lin-4), was identified in 1981 (10) and was cloned in 1993 (11), followed by lethal-7 (let-7) (12). miRNAs are initially transcribed by RNA polymerase II in the nucleus to form long RNA transcripts, termed primary-miRNA (pri-miRNA), with the stem-loop structure in which the mature miRNA sequences are embedded (13, 14) (Fig. 1). Pri-miRNAs contain the cap structure and poly(A) tails, which are the unique properties of class II gene transcripts (15). The nuclear ribonuclease (RNase)-III enzymes, Drosha, cleaves pri-miRNA into hairpin-shaped RNA of approximately 70 base pairs precursor-miRNAs (premiRNA), which consist of an imperfect stem-loop structure (16). Drosha is a nonspecific RNase; to mediate the genesis of miRNAs from the pri-miRNA, it forms a protein complex of approximately 600 kDa, termed microprocessor, with the essential cofactor DGCR8, also known as Pasha; a protein containing two double-stranded RNA (dsRNA)-binding domains (17–19). The specific cleavage at the stem-loop of the hairpin RNA structure of pri-miRNA is directed by DGCR8 (19). The microprocessor complex is also crucial for the posttranscriptional cross-regulation between Drosha and DGCR8 (20). DGCR8 stabilizes Drosha through an interaction between the middle domain of Drosha and the conserved C-terminal domain of DGCR8 (20, 21), whereas Drosha decreases DGCR8 mRNA level by cleaving it at a hairpin in the second exon (20, 22) (Fig. 1).

Figure 1.

Figure 1.

The biogenesis of miRNA begins with the transcription of miRNA to pri-miRNA in the nucleus. The pri-miRNA is cleaved by the Drosha/DGCR8 complex to form premiRNA, which can then be exported from the nucleus to the cytoplasm by exportin5. In the cytoplasm, premiRNA is further processed into the mature miRNA duplex by Dicer. One strand (usually the guide strand) of the mature miRNA forms a complex with Dicer and the Argonaute protein, known as the miRNA-containing RISC, where miRNA binds to the 3′ UTR of its target mRNA, resulting in the degradation (if the miRNA:mRNA duplex complementarity is perfect) or suppression of the translation of the target mRNA (if the complementarity is not perfect).

Following the initial cleavage by Drosha, the premiRNAs are exported by a Ran-guanosine triphosphate–dependent nucleo/cytoplasmic cargo transporter named exportin 5 (Exp5) from the nucleus to the cytoplasm, where the maturation of miRNA takes place (23). Exp5 was originally known as a minor export transporter for transfer RNAs (tRNAs) (24). Recent evidence indicated that the affinity of Exp5 for premiRNA is much higher than that for tRNA (23). As individual miRNAs are abundantly present at up to 50,000 copies per cell in some organisms (25), premiRNAs are probably the main cargo for this transporter compared with other RNAs, including short hairpin RNAs and tRNAs (23, 26). Exp5 also protects nuclear premiRNA from degradation by forming a complex with premiRNA and Ran-guanosine triphosphate together (27). Once in the cytoplasm, the premiRNA is cleaved by another RNase-III enzyme, Dicer, into a small, imperfect dsRNA duplex that contains both the mature miRNA strand and its complementary strand (28–30). Dicer is an RNase III–type endonuclease of ∼200 kDa (31); it is highly conserved in almost all eukaryotic organisms such as plants and animals (14). It cleaves at a site that is about 21 to 28 nucleotides (nt), depending on the species and the type of Dicer (32), from the 3′ end of the terminus of dsRNAs (the 3′-counting rule) (31, 33, 34). In mammals, Dicer also binds to the 5′ phosphorylated end of the premiRNA and cleaves it at about 22 nt from the 5′ end (the 5′-counting rule) (35). After cleavage by Dicer, the resulted 21 to 24 nt miRNA duplex needs to be unwound to release one of the strands for entering Argonaute for function (36, 37) (Fig. 1). Usually, one strand (“passenger” strand, also known as the star strand or “-5-p”) of this short-lived duplex disappears, whereas the other strand (“guide” strand or “-3p”) becomes the mature miRNA (38). It was originally thought that the passenger strands are subject to rapid degradation (37, 39, 40). However, recently, studies found that these strands could also be loaded on Argonaute and directed repression of target mRNAs (41, 42).

In animals, most miRNAs are incorporated into the RNA-induced silencing complex (RISC) (43) to form miRNA-containing RISC, which is also known as miRNA-containing ribonucleoprotein complex or mirgonaute (14, 38). The miRNA directs the RISC to the 3′ untranslated region (UTR) of the target mRNA to negatively regulate the expression of the latter at the posttranscriptional level by promoting mRNA cleavage (44, 45) and at the translational level by serving as a translational repressor to inhibit the translation of protein from mRNA (46), or a combination of the two (47). The initial evidence for miRNA targeting 3′ UTR of RNA came from the observation that a part of lin-4 sequence was complementary with the 3′ UTR of lin-14 mRNA (11,48) and is required for the repression of lin-14 by lin-4 (49). Recent studies have reported that, in animals, the 7 nt “seed region,” mapping to positions 2 to 8 at the 5′ end, of miRNA (50, 51) plays a key role on its target recognition and is highly conserved both within and between species (47, 52). When there is a perfectly complementary sequence between the miRNA and its target, the target mRNA is cleaved (53–55). However, in mammals, this phenomenon is rarely observed; the sequences of miRNA and its target are only partially complementary (53). In this case, mRNA is deadenylated and then decapped (56–59). An in vivo study showed that the decapped mRNAs were degraded (60). However, in cultured cell extracts, deadenylated mRNAs are not further degraded but remain in the deadenylated state, followed by translational repression (56).

III. miRNAs Regulating Insulin-Signaling Pathways and the Microvasculature

Under homeostatic conditions, cellular signaling by insulin is initiated by binding to insulin receptors (61, 62). The insulin receptor is a cell surface integral membrane protein expressed on most mammalian cells such as liver, adipose, and skeletal muscle cells (63–65). Insulin exerts its effect on three main target tissues: fat, liver, and skeletal muscle (66). Binding of insulin triggers oligomerization and receptor autophosphorylation on tyrosine residues (67–69) and tyrosine phosphorylation of insulin receptor substrates (IRSs) IRS1, IRS2, IRS3, IRS4, IRS5/DOK4, and IRS6/DOK5 (70–74). This phosphorylation provides the basis for the subsequent association with downstream signal proteins that diverge into three different pathways: the phosphoinositide-3-kinase (PI3K)/Akt pathway, the CAP/Cbl/TC10 pathway, and the mitogen-activated protein-kinase (MAPK)–dependent pathway (75–77).

PI3K interacts with phosphorylated tyrosines on IRS molecules (78, 79), resulting in increased levels of phosphatidyl inositol-4,5-diphosphate to phosphatidyl inositol-3,4,5-triphosphate (80, 81) followed by activation of phosphoinositide-dependent kinase 1 (82), which further phosphorylates protein kinase B (PKB) and atypical protein kinase C (83–85). Phosphorylation of PKB inactivates glycogen synthase kinase 3 (GSK-3), which results in glycogen synthesis (86). PKB also mediates stimulation of the fatty acid synthase promotor (87), inhibition of phosphoenolpyruvate carboxykinase gene transcription (88), and translocation of the main glucose transporter GLUT4 to the plasma membrane (89). In addition, PKB regulates downstream protein synthesis and degradation, such as 4E-BP1 and eIF-4G, which is mediated by phosphorylation and activation of mammalian target of rapamycin (mTOR) (90, 91).

For glucose uptake to be fully manifested, proto-oncogene Cbl in the second pathway is recruited to the phosphorylated insulin receptor by interaction with the adapter protein CAP (92, 93), which finally results in reinforced GLUT4 translocation in parallel with but not affecting PI3K signaling in response to insulin (76, 93). The third pathway after IRS activation includes GRB2, SHP2, and several other proteins, which lead to mitogen-activated protein-kinase activation, cellular proliferation, and differentiation via gene transcription regulation (77, 94–96).

A. Visceral fat

It is well recognized that human subjects with generalized obesity suffer from a high risk of insulin resistance and its metabolic complications, such as type 2 diabetes mellitus and hypertriglyceridemia (97). Inflammation in adipose tissue, as suggested by the presence of macrophages in the form of crown-like structures, has been identified as a mediator of systemic insulin resistance (98). Obese human subjects with excess visceral adipose tissue, or abdominal obesity, are at higher risk for type 2 diabetes components than those whose fat is located predominantly in the lower body, subcutaneously (99, 100). Only a few miRNAs have been reported to directly regulate adipocytes and visceral adipose tissue during insulin resistance (Table 1).

Table 1.

List of miRNAs Involved in Insulin-Relevant Tissues

miRNA Target Function Reference
Liver
 miR-103 Promotes adipogenesis in hepatocytes (108)
 miR-122 PTP1B Dephosphorylates tyrosine residues on the IR and IRS (290)
 miR-143 Impairs insulin-stimulated Akt activation and glucose homeostasis (291)
 miR-145 Inhibits glucose uptake in HepG2 cells, diminishes the phosphorylation of Akt and IRS1, and induces insulin resistance in hepatocytes (292)
 miR-190b IGF-1 Induces insulin resistance (293)
 miR-200a Increases the expression level of leptin receptor and IRS2, reduces body weight gain, and restores liver insulin responsiveness (294)
 miR-26a Improves insulin sensitivity, decreases hepatic glucose production, and decreases fatty acid synthesis (295)
 miR-379 Increases VLDL-associated triglyceride (296)
 miR-802 Hnf1b Impairs glucose tolerance and attenuates insulin sensitivity (297)
VAT
 miR-222 ER-α Reduces GLUT4 (298)
 miR-26b PTEN Promotes insulin-stimulated glucose uptake and increases insulin-stimulated GLUT4 translocation (299)
 miR-181b PHLPP2 Reduces visceral fat inflammation and improves glucose homeostasis and insulin sensitivity by increasing adipose tissue endothelial insulin signaling and Akt/eNOS activation (101)
Skeletal muscle
 miR125b IGF2 Regulates PI3K/Akt or MAPK pathway (300, 301)
 miR-135a IRS-2 Inhibits PI3K/Akt pathway and glucose uptake (302)
 miR-182 FoxO3 Attenuates atrophy-related gene expression (303)
 miR-199a-3p IGF, PIK3r1, and mTOR Activates AKT/mTOR signal pathway (304)
 miR-21 TGFβI Promotes PI3K/Akt/mTOR signaling (305)
 miR-24 p38 Inhibits activation of p38 MAPK pathway (306)
 miR-29a IRS-1 Impairs Akt/GSK-3 pathway and glucose uptake (307)

Although alterations in adipocytes and macrophage accumulation have been clearly associated with insulin resistance (98), the role of the microvasculature (i.e., endothelial cells [ECs]) in visceral fat in regulating insulin resistance is poorly understood. A recent study by our group identified that in response to high-fat-diet–induced insulin resistance, the anti-inflammatory miRNA miR-181b (101, 103) is significantly reduced in adipose tissue ECs, but not adipocytes, from visceral fat (epididymal white adipose tissue [eWAT]) after just one week. This finding raised the possibility that “miR-181b replacement therapy” may ameliorate inflammation in visceral fat and potentially insulin resistance. Indeed, high-fat-diet–induced, insulin-resistant mice that were tail vein injected with liposomally encapsulated miR-181b exhibited markedly improved glucose homeostasis and insulin sensitivity compared with controls (101). Mechanistically, miR-181b overexpression in adipose ECs enhanced insulin-mediated Akt phosphorylation at Ser473, and reduced endothelial dysfunction, an effect that shifted macrophage polarization toward an M2 anti-inflammatory phenotype in eWAT. These effects were associated with induction of endothelial nitric oxide synthase (eNOS), nitric oxide (NO) activity, and FoxO1 phosphorylation specifically in eWAT, but not in liver or skeletal muscle. Importantly, miR-181b overexpression in peripheral blood mononuclear cells had no effect on macrophage activation, proliferation, or recruitment to visceral fat. Similarly, adipocytes overexpressing miR-181b had no direct effect on glucose uptake; however, when conditioned medium from ECs overexpressing miR-181b was added to adipocyte cultures, these adipocytes were subsequently rendered less insulin resistant with improved glucose uptake. Bioinformatics and gene profiling studies revealed that the target of miR-181b was plexkstrin homology domain leucine-rich repeat protein (PHLPP2), a phosphatase known to dephosphorylate Akt at Ser473. Indeed, small interfering RNA (siRNA)-mediated knockdown of PHLPP2 recapitulated miR-181b’s favorable effects on glucose homeostasis, insulin sensitivity, and inflammation of eWAT. Interestingly, PHLPP2 is expressed robustly in eWAT, whereas it is barely detectable in liver and skeletal muscle, which may underlie the miR-181b tissue-specific effect in eWAT. Finally, these findings have translational significance, as ECs from diabetic subjects expressed higher PHLPP2 expression compared with healthy controls. Collectively, these findings highlight that miR-181b plays a critical homeostatic role in the microvasculature of visceral fat to control EC inflammation and insulin resistance (Fig. 2).

Figure 2.

Figure 2.

miR-181b improves endothelial dysfunction and consequently improves glucose homeostasis and insulin sensitivity in white adipose tissue by targeting the phosphatase PHLPP2 and reducing its expression. Reduced PHLPP2 enhances insulin signaling and AKT and eNOS expression.

B. Liver and skeletal muscle

Insulin resistance is one of the key components of the metabolic syndrome, and it eventually leads to the development of type 2 diabetes (103, 104). During the development of insulin resistance, hepatic lipid accumulation and hepatic inflammation may be observed in liver (104). In addition, skeletal muscle is quantitatively an important site of insulin resistance in type 2 diabetic patients and a direct risk factor for the development of cardiovascular disease (105, 106). More recently, several miRNAs have been reported to regulate hepatic and skeletal muscle function in type 2 diabetes (107–110) (Table 1; Fig. 3). Although some of these miRNAs, such as miR-143, miR-26, miR-29, and miR-103, have been implicated in regulating cell types important in the vessel wall such as ECs, a definitive role for these miRNAs in the microvasculature of these relevant tissues has not been elucidated, but may provide future pathophysiological insights and targets for therapy.

Figure 3.

Figure 3.

miRNA regulation of insulin signaling and glucose homeostasis. Many miRNAs have been identified to regulate insulin signaling in adipose tissue, liver, and skeletal muscle. miR-181b targets the phosphatase PHLPP2 in ECs of adipose tissue. miR-122, miR-103, miR-802, miR-143, and miR-26a regulate insulin signaling in the liver by regulating the expression of PTP1B, Cav-1, Hnf1b, ORP8, ACSL3, and PKCδ. miR-503, miR-125b, and miR-135a act in skeletal muscle on targets, including EFNB2 and VEGFA, IGF2, and IRS2, respectively. Endothelial-enriched miRNAs are highlighted in red.

IV. Role of miRNAs Involved in Diabetes-Induced Vascular Dysfunction

It is known that type 2 diabetes is associated with an increased risk of both micro- and macrovascular complications (111). In the macrovasculature, hyperglycemia can regulate multiple proinflammatory and proatherosclerotic target genes in ECs, vascular smooth muscle cells (VSMCs), and macrophages (112, 113). The morbidity associated with diabetic microvasculature disease, including retinopathy (114), neuropathy (115), nephropathy (116), and limb ischemia, is staggering (117). miRNAs have been implicated in the epigenetic regulation of key metabolic, inflammatory, and antiangiogenic pathways in type 2 diabetes and may contribute to common disease complications (118) (Table 2).

Table 2.

List of miRNAs Involved in Diabetes-Associated Vascular Dysfunction

miRNA Target Function Reference
Macrovasculature
 let-7a/b Prevents atherosclerosis (138)
 miR-1 Prevents endothelial dysfunction (141)
 miR-125b Suv39h1 Promotes the inflammatory reaction (156)
 miR-126 Spred-1 Promotes EPC proliferation and migration (136)
 miR-143/145 Increases VSMC cell area and reduces cell proliferation (153)
 miR-144-3p ABCA1 Promotes THP-1 macrophage differentiation into foam cells (308)
 miR-146a IRAK1, IL-6 Alters insulin sensitivity (131)
 miR-155 p65 Contributes to the progression of atherosclerosis (134)
Promotes macrophage-derived foam cell formation (164)
 miR-181b Importin-α3 PHLPP2 Card10 Confers anti-inflammatory effects in the macro- and microvasculature by regulating NF-κB and insulin signaling (101, 102, 143, 144)
 miR-195 SIRT1 Increases expression of FN (309)
 miR-200 Zeb1 Induces MCP-1 and COX-2 expression and promotes monocyte binding (157)
 miR-206 Enhances cholesterol efflux (164)
 miR-21 SPI Stimulates VSMC proliferation and reduces VSMC differentiation marker gene expression (155)
 miR-24 Reduces intraplaque macrophage proliferation (165)
 miR-492 resistin Reduces insulin resistance and endothelial dysfunction (310)
 miR-9 ACAT1 Decreases THP-1 macrophage-derived foam cell formation (311)
 miR-92a MKK4 and JNK Inhibits oxidative stress–induced VSMC apoptosis (312)
Microvasculature
 miR-130a Runx3 Maintains the normal EPC function (172)
 miR-135a TRPC1 Improves renal function and ameliorates progression (313)
 miR-146 CARD10 HuR Inhibits NF-κB activation (176)
 miR-146a IRAK1 Reduces IL-1β–induced ICAM-1 expression (198)
 miR-146a FN Inhibits thrombosis (127)
 miR-146a IRAK1 TRAF6 Promotes the complications of diabetic sensory neuropathy (198)
 miR-195 Prevents SIRT1-mediated tissue damage (309)
 miR-200b VEGF Prevents the glucose-induced increased vascular permeability and angiogenesis (169)
 miR-21 DAXX Prevents endothelial apoptosis (171)
 miR-29b Abolishes TGF-β/Smad3 pathway activation (314)
 miR-29c Spry1 Inhibition of miR-29c reduces mesangial matrix accumulation and albuminuria (186)
 miR-320 Inhibits EC migration and tube formation (174)
 miR-34a Alleviates glomerular hypertrophy (315)
 miR-93 Msk2/VEGF-A MiR-93 overexpression reduces features of diabetic nephropahy (187, 188)

Hyperglycemia and insulin resistance have been identified as key players in the development of diabetic atherosclerosis, with metabolic insulin signaling being an important contributor to normal vascular function and homeostasis (119). Abnormalities in vascular EC and VSMC function and macrophage activation are important contributors to vascular complications and propensity to both macro- and microvascular dysfunction (120).

A. Role of miRNAs in endothelial dysfunction and insulin resistance/type 2 diabetes

Both insulin resistance and endothelial dysfunction are associated with increased circulating markers of inflammation (121). In physiological conditions, insulin promotes endothelium-dependent relaxation by a mechanism that involves an increase of NO production via activation of PI3K and Akt kinase pathways (110, 122). However, in pathological conditions, insulin resistance reduced bioavailability of NO production via inhibition of eNOS activation (123) and eNOS expression (124), an effect that may be mediated by TNF-α-induced NF-κB signaling pathway (123, 125). Indeed, genetic inhibition of NF-κB signaling in ECs protects mice from the development of obesity- and aging-associated insulin resistance (126). This is accompanied by decreased macrophage infiltration into adipose tissue and oxidative stress in plasma, and increased blood flow, mitochondrial content, and insulin delivery in muscle (126). These favorable metabolic phenotypes are partially explained by increased eNOS expression. Emerging studies highlight important roles for miRNA-mediated regulation of endothelial dysfunction and diabetes (Table 2).

For example, miR-146a expression was reduced in both high glucose–treated ECs and streptozotocin (STZ)-induced diabetic rats (127, 128). Further studies demonstrated that miR-146a lowered ICAM-1 expression by targeting IL-1 receptor-associated kinase (IRAK1) (129) and by inhibiting the inflammatory signaling via binding to IL-6 (130), which alters insulin sensitivity and contributes to the progression of atherosclerosis (131). miR-155 expression was increased in type 2 diabetic mice (132) and impaired endothelium-dependent vasorelaxation by directly targeting the 3′ UTR of eNOS (133). However, Wu et al. found that TNF-α–induced miR-155 serves as a negative feedback regulator in endothelial inflammation and atherosclerosis by targeting the nuclear transcription factor p65 (134). Moreover, clinical studies showed that expression of miR-126 was reduced in type 2 diabetic patients with coronary artery disease, and type 1 diabetic subjects with reduced miR-126 expression was associated with higher micro- and macro-vascular complications (118, 135–137). miR-126 promoted endothelial progenitor cell (EPC) proliferation and migration and inhibited apoptosis via targeting Spred-1, an intracellular inhibitor of the Ras/extracellular signal-regulated kinase (ERK) cascade, which may influence the PI3K/Akt/eNOS signaling pathway (136), an important insulin-signaling pathway. In addition, overexpression of let-7a and let-7b inhibited oxidized-low-density-lipoprotein–induced EC apoptosis, NO deficiency, and eNOS downregulation and attenuated oxidized-low-density-lipoprotein–induced p38MAPK phosphorylation, NF-κB nuclear translocation, IκB degradation, and PKB dephosphorylation. These findings may provide new insights into the protective properties of let-7a and let-7b in preventing atherosclerosis and potentially insulin resistance (138).

Overexpression of miR-492 reduced insulin resistance and endothelial dysfunction induced by high glucose both in vitro and in vivo by directly reducing the expression of resistin, which also plays an important role in the development of both atherosclerosis and insulin resistance (139, 140). Feng et al. (127) reported that miR-1 was reduced in STZ-induced diabetic rats and, at the same time, endothelin-1 (ET-1) expression, a key marker indicating endothelial dysfunction was increased. Furthermore, transfection of miR-1 mimics inhibited the high glucose­–mediated induction of ET-1 in human umbilical vein endothelial cells (HUVECs), suggesting that miR-1 prevents endothelial dysfunction induced by hyperglycemia (141). In addition, induction of miR-195 after high glucose treatment increased expression of fibronectin (FN) via targeting SIRT1, a regulator of aging in ECs, indicating that miR-195 has the potential to promote thrombosis via induction of FN (142).

Finally, the miRNA miR-181b not only regulates vascular inflammation in the microvasculature as highlighted above in adipose tissue ECs, but also suppresses EC inflammation in the macrovascualture (102, 143–146). Indeed, systemic delivery of miR-181b markedly reduced atherosclerotic lesion formation independent of any effects on lipid profiles and reduced arterial thrombosis in mice (143, 144) (Table 2).

B. Role of miRNAs in VSMC function and insulin resistance/type 2 diabetes

VSMCs are also targets of insulin, an effect that bears clinical importance for vasculoproliferative states, both macrovascular (e.g., coronary artery disease and intimal hyperplastic restenosis to microvascular disease (e.g., myocardial arteriogenesis, retinopathy) (147, 148). Indeed, compared with nondiabetic VSMCs, human diabetic VSMCs exhibit abnormal morphology in culture, increased proliferation, and greater adhesion and migration rates (147). Through the PI3-K pathway, insulin stimulates glucose transport, induces the well-differentiated contractile state, antagonizes the effects of platelet-derived growth factor, and increases NO production (149). When the PI3-K pathway is inhibited, insulin increases chemotaxis (150) and VSMC proliferation via the MAPK pathway (151). Moreover, by the cooperation of the two pathways, insulin activates the HIF/VEGF pathway (152). Accumulating studies highlight several VSMC-enriched miRNAs that contribute to vascular dysfunction in diabetes (Table 2).

For example, miR-143 and miR-145, two upregulated miRNAs in VSMCs isolated from type 2 diabetic patients, increased VSMC area and reduced cell proliferation. Transfection of anti–miR-143/145 conferred type 2 diabetes-VSMC characteristics into a nondiabetic phenotype, implying that miR-143/145 impaired VSMC phenotype and function, suggesting a new therapeutic intervention of vascular function in type 2 diabetic patients (153). Another miRNA, miR-21, may also be important in regulating VSMC proliferation. Insulin resistance led to a significant reduction of miR-21 expression (154), and overexpression of miR-21 stimulated VSMC proliferation and reduced VSMC differentiation marker gene expression by targeting specificity protein-1 (155) (Table 2).

Finally, miR-125b and miR-200 have been implicated in modulating VSMC inflammatory effects. In VSMCs from diabetic db/db mice, both miR-125b and miR-200 were upregulated compared with nondiabetics cells (156, 157). Via targeting histone methyltransferase Suv39h1, miR-125b increased the expression of MCP-1 and IL-6 and enhanced monocyte binding to promote inflammation in db/db mice (156). Moreover, overexpression of miR-200 mimics also induced MCP-1 and COX-2 expression and promoted monocyte adhesion via binding Zeb1, an E-box binding transcriptional repressor. Interestingly, silencing Zeb1 increased miR-200 expression, indicating a feedback regulatory loop during this process (157). Collectively, therapeutic silencing of miR-143/miR-145, miR-21, miR-125b, or miR-200 may limit VSMC-associated proliferative states or VSMC-derived inflammation important to a range of diabetic macro- or microvascular complications.

C. Role of miRNAs in macrophage function and insulin resistance/type 2 diabetes

Abundant studies indicate that monocytes are recruited to peripheral tissues (i.e., pancreas, liver, and adipose tissue) to become resident macrophages and contribute to local inflammation and development of insulin resistance (158). In the state of insulin resistance, there is a distinct switch, termed macrophage polarization (159), in the macrophage populations present from an anti-inflammatory (M2) population to an inflammatory (M1) population, which releases cytokines and chemotactic factors that exacerbate and amplify the local inflammatory environment, thereby creating a vicious cycle (160). miRNAs figure prominently in regulating macrophage activation and polarization dynamics (Table 2).

For example, through targeting the adenosine triphosphate–binding cassette transporter A1 (ABCA1), miR-144-3p mimics increased the secretion of inflammatory cytokines such as IL-6, IL-1β, and TNF-α to promote THP-1 macrophage differentiation into proinflammatory foam cells. Activation of Liver X receptors (LXRs) in macrophages promotes cholesterol efflux (161, 162) and protects against the development of insulin resistance and atherosclerosis (163). Overexpression of miR-206 in THP-1 macrophages increased LXRα expression and enhanced cholesterol efflux. Activation of LXRα also reduced miR-206 expression, indicating that there is a feedback loop between miR-206 and LXRα in regulating cholesterol efflux, vascular inflammation, insulin resistance, and atherosclerosis (164). Finally, miR-24 reduced intraplaque macrophage proliferation rates by 34% by targeting the matrix metalloproteinases-14 (MMP-14), highlighting its therapeutic potential in macrophage proinflammatory functions (165).

V. Diabetic Complications

A. miRNAs involved in aberrant angiogenic growth of ECs and diabetic retinopathy

Diabetic retinopathy is one of the leading causes of blindness (139). Nearly 60% of type 2 diabetic patients are expected to have some form of retinopathy by the first decade after diagnosis (166). Aberrant angiogenic growth of ECs is now recognized to be an important component of diabetic retinopathy (167, 168). Several miRNAs have been implicated in regulating aberrant EC growth (Fig. 4). In the retina of STZ-induced diabetic rats, miR-200b expression decreased, whereas its direct target, VEGF, increased on both the mRNA and protein level. Overexpression of miR-200b normalized VEGF expression in the diabetic rat retina and prevented the glucose-induced increased permeability and angiogenesis in HUVECs, highlighting the importance of miR-200b in the regulation of VEGF-mediated diabetic retinopathy (169). Under hypoxic conditions, miR-126 expression is reduced in diabetic retinas. Restoration of miR-126 expression halted the hypoxia-induced neovascularization by suspending cell cycle progression and inhibiting the expression of VEGF and MMP-9 (170). Inhibition of a different miRNA, miR-21, significantly enhanced the high glucose–induced endothelial cytotoxicity. Furthermore, overexpression of miR-21 inhibited the expression of death domain associated protein (DAXX), a proapoptotic mediator, whereas silencing DAXX mRNA reversed the inhibition of miR-21 on endothelial apoptosis induced by high glucose, implying that miR-21 could protect ECs from apoptosis by inhibiting DAXX expression (171). In EPCs derived from patients with diabetes, transfection of miR-130a increased the protein level of ERK/VEGF and Akt via repression of Runx3, suggesting that the miR-130a can maintain normal EPC function through ERK/VEGF and Akt signaling pathways (172). In diabetic rats, miR-320 negatively regulated glucose-induced increase of VEGF and FN (173) and inhibited EC migration and tube formation (174). Finally, in response to high glucose conditions in ECs, several miRNAs such as miR-181c, miR-20b, and miR-15a, were reduced; however, their functional effects have not been explored (175).

Figure 4.

Figure 4.

miRNA regulation of diabetic wound healing and myocardial angiogenesis. These miRNAs are involved in regulating diabetic wound healing and myocardial angiogenesis through effects on their targets that control cell proliferation, migration, apoptosis, and endothelial microvascular function. In response to diabetic conditions, several miRNAs are dysregulated (marked with an arrow).

NF-κB is a ubiquitous inducible transcription factor and is a key proinflammatory mediator of cellular pathways in the early phases of diabetic retinopathy development. In human retinal ECs, thrombin increased miR-146 expression in an NF-κB–dependent manner, and at the same time, miR-146 promoted a negative feedback to thrombin-induced NF-κB activation via targeting CARD10 (176). Human antigen R (HuR) has been reviewed as a promising therapeutic target for angiogenesis (177). Through binding to HuR, miR-146 also repressed endothelial activation by inhibiting the proinflammatory NF-κB pathway (178). miR-146a reduced the expression of the NF-κB–responsive adhesion molecule ICAM-1 by binding to the 3′ UTR of IRAK1, an upstream adapter protein activated by IL-1β in both human retinal ECs and diabetic rat retinas (179). Moreover, high glucose increased miR-195 and decreased SIRT1 expression in diabetic rat retinal ECs. Neutralization of miR-195 rescued SIRT1 expression and decreased tissue damage in diabetic retinopathy (180), highlighting an important role for miR-195-SIRT1 signaling axis in modulating retinal EC function.

B. miRNAs involved in the pathogenesis of diabetic nephropathy and diabetic microvascular kidney disease

Diabetic nephropathy is the major cause of end-stage renal disease and high mortality in diabetic patients (181, 182). Accumulating evidence has demonstrated that miRNAs regulate signaling pathways involved in the pathogenesis of diabetic nephropathy, including the microvasculature that figures prominently in this process (182) (Fig. 4). Renal hypertrophy and abnormal extracellular matrix deposition are hallmarks of diabetic nephropathy (183). FN is widely expressed in embryos and adults, especially in regions of active morphogenesis, cell migration, and inflammation (184). The main function of FN is to serve as a scaffold for cell adhesion and migration, thereby also mediating cell proliferation and differentiation (185). FN also contributes to micro- and macrovascular thrombosis (142). In kidneys of STZ-induced diabetic mice, both endothelin-1 and FN was increased with reduced expression of miR-1; in addition, miR-1 overexpression in ECs prevented the high glucose–induced ET-1 upregulation, suggesting a potential link in diabetic microvascular kidney disease (141). In HUVECs and STZ-induced diabetic rats, high glucose decreased miR-146a and increased FN expression. Interestingly, overexpression of miR-146a decreased FN expression in ECs, raising the possibility that efforts to maintain miR-146a expression in the microvasculature may be therapeutically relevant for diabetic nephropathy (127).

Finally, two other miRNAs, miR-29c and miR-93, have been implicated in regulating diabetic nephropathy through unique mechanisms directly or indirectly related to microvascular function. miR-29c inhibition reduced albuminuria and kidney mesangial matrix accumulation in a diabetic mouse model, an effect thought to be mediated by miR-29c targeting Sprouty homolog 1 (Spry1) and, in turn, regulation of Rho kinase (186). However, Spry1 has also been implicated as a MAPK antiangiogenic mediator, raising the possibility that miR-29c’s effects could also be related to angiogenic effects. miR-93, on the other hand, has been more directly associated with alterations of angiogenic signaling and diabetic nephropathy. Mice with inducible overexpression of miR-93 in podocytes showed marked improvements in diabetic nephropathy through modulating chromatin reorganization and targeting Msk2, a histone kinase, and its substrate H3S10 (187). Interestingly, miR-93 is localized within the intron of MCM7 gene, and its expression is reduced in response to high glucose. The authors showed that hyperglycemia decreased MCM7 promoter activity, which, in turn, decreased miR-93 and increased its target gene VEGF-A and VEGF signaling in kidneys, an effect that increased collagen and FN expression, paving the way to diabetic nephropathy (188). Taken together, overexpression of miR-146a and miR-93, or neutralization of miR-93, may provide potential therapeutic targets for regulating endothelial function and key steps in diabetic nephropathy.

C. miRNA regulation of diabetic neuropathy

Diabetic neuropathy is a nerve disorder that occurs in nearly 50% of patients with diabetes (189–191). It is found in patients who have had type 1 diabetes for more than five years and in the early stages of type 2 diabetes (192–194). The diabetic neuropathies are comprised of several different syndromes: distal symmetric polyneuropathy (typically presents with the gradual onset of numbness, paresthesias, or dysesthesias in the feet), median mononeuropathy (selective injury of individual nerves), visceral autonomic neuropathy (such as genitourinary dysfunction and orthostatic hypotension), small fiber and painful neuropathy, and other diabetic nerve injury (195). Several miRNAs have been reported to be involved in diabetic neuropathy (196–198) (Fig. 4).

However, only one miRNA, miR-146a, uniquely suppresses the same proinflammatory NF-κB targets in neurons and ECs relevant to the pathogenesis of diabetic neuropathy and the diabetic microvasculature. Specifically, overexpression of miR-146a in dorsal root ganglion (DRG) neurons inhibited the upregulation of the targets IRAK1 and TRAF6 (198–200). In db/db mice, which is a mouse model of diabetic peripheral neuropathy, hyperglycemia suppressed miR-146a expression and increased the protein levels of the proinflammatory adaptor proteins IRAK1 and TRAF6; similar results were also observed in cultured DRG neurons (198), which comprise thermoceptors, mechanoceptors, and itch sensors (201–203) thought to be responsible for the complication of diabetic sensory neuropathy (204–206). As such, miR-146a plays an important role in protecting DRG neurons from apoptosis under hyperglycemic conditions, suggesting that miR-146a might also prevent the development of diabetic neuropathy. Because miR-146a also confers favorable anti-inflammatory, protective effects in ECs in the microvasculature by regulating IRAK1, TRAF6, and HuR/eNOS, it may serve as a particularly important miRNA in the microvasculature and in peripheral neuropathy. Collectively, given the emerging role of EC plasticity governed by sympathetic nerve innervation (207), future studies will be necessary to explore the importance of miRNAs in peripheral nerves of patients with diabetes and the potential dual role of miRNAs regulating the microvasculature and peripheral nerves, as highlighted by miR-146a.

D. miRNAs involved in diabetic wound healing and angiogenesis

Accumulating studies suggest a number of miRNAs may be dysregulated and participate in different pathophysiological phases of diabetic wound healing (208) (Fig. 4). For example, miR-26a was shown to be induced in diabetic dermal wounds in mice. Local inhibition of miR-26a expression increased granulation tissue, induced angiogenesis, and promoted wound healing through an ID1/SMAD1 signaling pathway in ECs. In addition, local inhibition of miR-26a had no effect on leukocyte accumulation and enhanced the accumulation of myofibroblasts, which are important for wound contraction (209). Another miRNA, miR-155, known to functionally regulate diverse aspects of the immune response was found to be induced in diabetic wounds in mice (210). Deficiency of miR-155 led to a reduced inflammatory response and improved wound closure, an effect associated with increased expression of miR-155 target genes BCL6, RhoA, SHIP1, and FIZZ1 (210). In addition, expression of the members of the miR-99 family were found to be reduced in diabetic wounds, and overexpression of this miR family reduced PI3K/Akt signaling and migration and proliferation of keratinocytes, implicating their potential role in the later phase of re-epithelialization (211). The expression of a different miRNA, miR-21, was induced late (day 8) in dermal wounds of diabetic mice. Although miR-21 overexpression increased fibroblast migration in vitro, the role of altering miR-21 expression in diabetic wound healing remains unknown (208). In addition, the proinflammatory cytokine TNF-α increased expression of miR-200b in diabetic wounds, an effect that decreased GATA2 and VEGFR2 and altered the angiogenic response; conversely, miR-200b deficiency attenuated TNF-α’s effects and promoted angiogenesis (212). In diabetic limbal corneas, miR-146a was shown to be significantly increased. Overexpression of miR-146a in limbal epithelial cells decreased epithelial cell migration and wound closure in addition to phosphorylating p38 and EGFR. Ex vivo studies showed that inhibition of miR-146a in cultured human diabetic corneas promoted wound healing (213). In diabetes, several cell types exhibit impaired function including bone marrow–derived angiogenic cells (BMACs). Overexpression of miR-27b increased cell proliferation, tube formation, adhesion, and delayed apoptosis by directly targeting TSP1, TSP2, semaphorin 6A, and p66shc (Src homology 2 domain containing transforming protein 1) in BMACs. Topical transplantation of BMACs overexpressing miR-27b in wounds of diabetic mice resulted in improved wound healing and wound perfusion. Interestingly, direct delivery of miR-27b to the wounds of diabetic mice only partially improved wound healing, suggesting that additional miRNAs are likely to be involved (214). Finally, examination of human subjects with type 2 diabetes revealed distinct circulating miRNA patterns, including increased expression of miR-191, in the presence of chronic wounds (215). Interestingly, endothelial-derived miR-191 was taken up in dermal cells, leading to a reduction in the miR-191 target zonula occludens-1. Moreover, increased miR-191 expression impaired angiogenesis by suppressing migration of both diabetic dermal ECs and fibroblasts, in part, by targeting zonula occludens-1 (215). Collectively, these studies reveal important paracrine or “miracrine”-mediated effects from the microvasculature of wounds on dermal fibroblast function, an effect that may impact diabetic wound healing.

E. miRNAs involved in myocardial injury and angiogenesis

A number of miRNAs are implicated in myocardial injury and angiogenesis (Figure 4). miR-26a was shown to be important in both physiological and pathological angiogenesis by suppressing endothelial SMAD1 expression, an effect leading to reduced Id1 and increased p21WAF/CIP1 and p27 expression and EC cycle arrest (209). miR-26a expression is increased in response to acute myocardial infarction in mice and in human subjects with acute coronary syndromes. miR-26a overexpression impairs physiological angiogenic responses such as exercise-induced angiogenesis in mice and developmental angiogenic responses such as the formation of the caudal venous plexus formation in zebrafish (209). In contrast, in vivo neutralization of miR-26a reduced myocardial infarct size by rapidly inducing robust angiogenesis by two days with improved left ventricular function, suggesting a new therapeutic approach for diseases associated with impaired pathological angiogenesis, including diabetic myocardial or peripheral artery disease (209). Inhibition of miR-92a, a part of the miR-17-to-miR-92 cluster, also reduced infarct size and promoted neovascularization in response to myocardial infarction by targeting integrin-α5 and eNOS (216). The role of miR-92a in physiological angiogenesis and diabetes remains unknown. miR-126 targets sprouty-related EVH-1 domain-containing 1 and regulated developmental angiogenesis in zebrafish and neovascularization after ischemic myocardial injury in mice (217). Members of the miR-23∼miR-27∼miR-24 cluster target several known angiogenic factors, including semaphorin 6A, Sprouty2, GATA2, and p21-activated kinase PAK4, to promote myocardial and retinal neovascularization (218–221). Finally, overexpression of miR-342-5p inhibited EC proliferation and angiogenesis in vitro and in vivo through direct targeting of endoglin. In addition, miR-342-5p inhibited TGF-β signaling by repressing SMAD1/5 phosphorylation in ECs. In a pathological model of angiogenesis using lased-induced choroidal neovascularization (CNV) in mice, miR-342-5p expression was significantly induced. Overexpression of miR-342-5p in this model system inhibited neovascularization and decreased the CNV area (222). Because CNV may coexist with chronic diabetic retinopathy (223), miRNAs may figure prominently in disease pathogenesis or response to therapy.

Although there are a number of studies that focus on the role of miRs in the context of diabetic angiogenesis, only a few studies examined the role of miRNAs specifically in type 1 diabetes. For example, circulating miR-27b and miR-320a are implicated in the progression of retinopathy in type 1 diabetes. miR-27b directly targets TSP-1, an extracellular matrix protein that is implicated in EC proliferation, migration, and angiogenesis, whereas miR-320a indirectly targets TSP-1 by reducing its secretion by ECs (224). In vitro inhibition of miR-195 protects against palmitate-induced EC apoptosis, suggesting that it may play a role in EC angiogenesis and metabolic-induced ER stress. In a mouse model of STZ-induced type 1 diabetes, miR-195 expression was increased in mouse hearts, and inhibition of miR-195 increased myocardial capillary density and coronary blood flow and reduced myocardial hypertrophy, in part, by attenuating oxidative damage and caspase 3 activity. Mechanistically, silencing of miR-195 increased BCL-2 and Sirt-1 protein expression (225). Because reduced coronary flow reserve is a strong indicator of coronary microvascular dysfunction in patients with diabetes and predicts cardiac mortality (226), these findings suggest that neutralization of miR-195 may improve diabetic coronary microvascular dysfunction, an emerging therapeutic interest to reduce cardiovascular burden in this predisposed population (227).

F. miRNAs and hyperglycemic memory

Hyperglycemic memory describes a phenomenon that the detrimental effects of hyperglycemia may persist even after restoration of normal glucose levels in diabetic patients (228, 229). Large population studies revealed that the persistent risk for vascular complications is observed for both type 1 and 2 diabetic patients (230, 231). Emerging evidence demonstrate that miRNAs contribute to hyperglycemic memory. For example, it has been shown that hyperglycemia significantly affects miRNA expression in the heart of diabetic mice, and many of them remained significantly altered after three weeks of intensive glycemic control with insulin (232). In miR-125b, miR-146a-5p, and miR-29a-3p were associated with persistent impaired endothelial function and altered proinflammatory gene expression after transient high-glucose treatment (233). The role of miRNAs in the pathogenesis of diabetic complications and metabolic memory has also been recently reviewed by Prattichizzo et al. (234). The causal role of miRNAs in hyperglycemic memory awaits further investigation. Targeting these miRNAs associated with hyperglycemic memory could open new therapeutic avenues to reduce cardiovascular events or residual cardiovascular risk in patients with diabetes.

VI. Circulating miRNAs

Exosomes are spherical-to-cup-shaped, 30- to 100-nm microvesicles (235) and can be released by all cell types (236). The composition of exosomes is very complicated, and they are enriched in various lipids, proteins, miRNAs, mRNAs, and DNAs (237). After they are released, exosomes enter the circulatory system and bodily fluids, including plasma, urine, bile, and breast milk, among others (238, 239). Therefore, the content of exosomes can be transferred from host cells to recipient cells carrying their content, which can affect the function of recipient cells, thereby facilitating communication between host and recipient cells (235, 240, 241).

miRNAs can also be detected in the extracellular environment, circulating in various biological fluids such as plasma, sera, urine, cerebrospinal fluid, and tears (242). The majority of detectable miRNAs are primarily concentrated in exosomes in serum and saliva (243). Accumulating studies demonstrate that circulating miRNAs carried in exosomes may influence target cell function in an analogous manner as some circulating hormones (244, 245). In this review, we summarize the effects of exosomal miRNAs on diabetic mellitus and their potential as biomarkers.

A. Exosomal miRNAs in type 1 diabetes

β-cell dysfunction and impaired insulin production are hallmarks of diabetes. Thioredoxin-interacting protein, a cellular redox regulator, has been reported to induce the expression of miR-204 in β-cells. By targeting MAFA, a known insulin transcription factor, miR-204 can block insulin production (246). Moreover, miR-204 can also target the protein kinase R-like ER kinase (PERK) to regulate β-cell apoptosis through the unfolded protein response (247). Other studies revealed that miR-204 was abundantly expressed in exosomes of human aqueous humor (248), is regulated by the gut microbiome, and impairs endothelium-dependent vasorelaxation by targeting the Sirtuin1 lysine deacetylase (Sirt1) (249). These findings highlight the important role of circulating miR-204 in both diabetes and endothelial function.

A cross-sectional cohort study revealed that circulating miRNA levels of miR-21 and miR-210 were significantly upregulated in the plasma and urine of type 1 diabetic patients, whereas urinary miR-126 levels were significantly lower in diabetic patients. These results suggest that miRNAs may participate in the early onset of type 1 diabetes (250). Because miR-21 is also induced by TGF-β1, an essential mediator in the pathogenesis of diabetic nephropathy, considerable attention has focused on miR-21 as a potential mediator in this condition (251). For example, renal cortices from type 1 diabetic mice demonstrated significantly elevated levels of miR-21, which mediates critical pathologic features of diabetic kidney disease through inhibiting the expression of its target PTEN and increasing Akt phosphorylation (252). Collectively, these findings provide a new potential diagnostic biomarker for diabetic nephropathy in type 1 diabetic patients.

In new-onset type 1 diabetes in children, miR-25 was negatively associated with residual β-cell function and positively associated with glycemic control, suggesting a predictive biomarker for type 1 diabetes diagnosis and a new target for therapeutic intervention (251). Inflammatory cytokines contribute to the progressive deterioration of pancreatic β-cell function and development of type 1 diabetes (253, 254). In response to IL-1β, for example, miR-101a and miR-30b participate in β-cell dysfunction by decreasing insulin content and gene expression and increasing β-cell death (255).

B. Exosomal miRNAs in type 2 diabetes

Accumulating studies demonstrate that circulating miR-126 levels are decreased in the blood of patients with type 2 diabetes (256–258). miR-126 is also highly expressed in ECs (38) to maintain endothelial homeostasis and vascular integrity in part through regulating the VEGF pathway (217, 259). miR-126 was identified as a key miRNA that confers the proangiogenic properties of circulating CD34+ cells (260). The CD34+ cell subset secretes miR-126 largely in microvesicles and exosomes, which are taken-up by ECs, and promotes angiogenesis (260). However, this proangiogenic capacity of CD34+ cells from diabetic patients was significantly attenuated due to the reduction of miR-126 expression (260). Recent clinical studies also demonstrated that, in peripheral whole blood, circulating miR-126 levels in type 2 diabetic patients are lower than in healthy controls and increased significantly after improved glycemic control. These findings suggest that circulating levels of miR-126 may be considered as a potential blood-based biomarker, perhaps useful for monitoring therapeutic responses in type 2 diabetic patients, including diabetic angiogenic responses important to diabetic coronary or peripheral artery disease (258, 261).

Serum miR-130b levels were significantly decreased in type 2 diabetic patients with diabetic nephropathy (262), and increased expression of miR-130b improves renal tubulointerstitial fibrosis by binding to the 3′ UTR of Snail mRNA both in vitro and in vivo (263). Furthermore, miR-130b can also be shuttled by microvesicles and transferred into target cells to regulate recipient cell function (264). Therefore, microvesicle-shuttled miR-130b has the potential to be a circulating therapeutic tool in diabetic nephropathy.

Several other miRNAs may serve as circulating biomarkers or mediators in type 2 diabetes. For instance, miR-1 has the potential to prevent endothelial dysfunction induced by hyperglycemia (141). Several studies have also found that plasma miR-1 levels were significantly higher in patients with myocardial infarction (265). In addition, microarray profiling studies identified the miRNAs miR-144, miR-146a, miR-150, and miR-182 as four diabetes-related miRNAs in rat peripheral blood (128). Among these miRNAs, circulating miR-144 levels were correlated with a reduction in the expression of its target, IRS1, at both the mRNA and protein levels (128), implying a new potential diagnostic and therapeutic target of type 2 diabetes. High-glucose treatment also reduced the miR-126 content of endothelial apoptotic bodies, which was similar to circulating microvesicles in plasma of patients with type 2 diabetes (256). miR-375 was recently characterized to be a pancreatic islet–specific miRNA, and knockdown of endogenous pri-mir-375 expression almost completely protected NIT-1 cells from palmitate-induced lipoapoptosis by repressing its target V1 expression, suggesting a potential therapeutic target for β-cell dysfunction and β-mass loss in type 2 diabetes (266). Interestingly, miR-375 is significantly increased in type 2 diabetic patients and has the potential to serve as a new biomarker for type 2 diabetes (267, 268). Taken together, emerging studies indicate that these exosomal miRNAs in the circulation of type 2 diabetic subjects may not only serve as potential biomarkers, but they may also regulate key processes in the pathology of a range of type 2 diabetes–associated conditions.

VII. Conclusions and Future Therapeutic Opportunities

A. Rationale for targeting miRNAs in the diabetic microvasculature

Therapeutic targeting of noncoding RNAs such as miRNAs represent an emerging frontier in the treatment of a variety of chronic disease states, including diabetes and a range of diabetes-associated complications that manifest with microvasculature dysfunction. Although several classes of RNA therapeutics are currently under development, including miRNA mimetics and inhibitors, siRNAs, and antisense oligonucleotides, only one antisense oligonucleotide drug, mipomersen, which targets apoB, is currently approved by the U.S. Food and Drug Administration for patients with homozygous familial hypercholesterolemia (269). In contrast to single-gene targeting using siRNA, therapeutic targeting of miRNA offers the ability to regulate gene networks important to the pathogenesis of complex diseases such as diabetes. Effective delivery of miRNA mimics or inhibitors to the microvasculature may facilitate improved cellular signaling and maladaptive biological processes in the local environmental of injured diabetic tissues.

The ability of miRNAs to circulate in exosomes and microvesicles provides a unique opportunity to use these “hormone-like” noncoding RNAs as biomarkers not only for the diagnosis and prognosis in a range of diabetes-associated conditions with altered microvascular function, but also in response to current or emerging therapies. In addition, because miRNAs may be expressed in specific cell types or in response to certain stimuli, careful interpretation is required for determining which miRNAs may best serve as potential biomarkers and therapeutic targets. Furthermore, miRNA target genes may be differentially expressed in a tissue- or cell-specific manner, which may be important for distinct diabetic microvascular environments.

B. Obstacles and challenges for effective miRNA delivery and targeting

Modulation of miRNAs may be facilitated locally or systemically using a range of chemically modified antisense microRNA inhibitors (antimiRs) (to inhibit miRNA) or miRNA mimics (to overexpress miRNA). Naked antimiRs are readily degraded by endogenous circulating or tissue RNases. However, several strategies have been used to chemically modify antimiR oligonucleotides to enhance target stability, affinity, or tissue uptake (270–272). For example, locked nucleic acids exhibit high binding efficiencies and improved stability with the addition of a methylene link between the 2′-oxygen and the 4′-carbon resulting in a locked position and reducing the flexibility of the ribose ring (273). Other modifications include ribose 2’-OH group modifications, such as 2′4′–constrained 2′O′-ethyl and 2’methoxyethyl, and phosphorothioate modification. Because a recent study highlighted that phosphorothioate modifications may facilitate nucleotide-based drugs to bind and activate platelets eliciting thrombus formation in response to carotid injury, pulmonary thromboembolism, and mesenteric artery injury in mice (274), careful scrutiny of chemical modifications will be required to minimize potential side effects and unanticipated toxicities. In contrast to their double-stranded counterparts (miRNA mimics), single-stranded antimiR oligonucleotides can be formulated in saline or phosphate-buffered saline for subcutaneous or intravenous delivery and do not require lipid- or nanoparticle-based delivery systems. Upon systemic delivery, these antimiRs rapidly leave the plasma and are taken up by multiple tissues, most notably the liver, the spleen, the kidney, adipose tissue, and bone marrow (275). Upon cellular uptake, the antimiR generates a high-affinity, stable bond, with the miRNA reducing the availability of the endogenous miRNA for binding to the 3′ UTR of the mRNA target(s). For a general view of miRNA targets, delivery of antimiRs, and therapeutic developments, please refer to the seminal review by van Rooij et al. (276).

C. Future prospects and unique platforms for miRNAs

Preclinical studies in nonhuman primates using naked antimiR oligonucleotides have shown promise for targeting miRNAs expressed highly in the liver, such as miR-122 or miR-33 (277). Cholesterol analogs have been added to antimiRs to facilitate cellular uptake, particularly in the liver, and this enhances their incorporation into high-density lipoprotein and low-density lipoprotein (278). Alternative approaches for miRNA inhibition include the use of competitive miRNA inhibitors, such as miRNA sponges or decoy transcripts that contain miRNA binding sites complementary to the seed sequence of the miRNA of interest (279).

Several approaches have been established to reconstitute miRNAs in disease states in which miRNA deficiency occurs. For instance, reduced miR-181b expression in the microvasculature of visceral fat is associated with the onset of insulin resistance, and delivery of miR-181b mimics was used to restore its expression and ameliorate insulin resistance in mice (101). Because of their double-stranded nature, the delivery of therapeutic miRNA molecules in vivo has many of the same challenges as siRNAs. As such, drug delivery vehicles such as liposomes, polymeric micelles, and nanoparticle-based drug carriers are being developed to deliver these oligonucleotides to cells and tissues. One of the major challenges associated with miRNA mimics replacement technologies include the ability to target miRNA to a specific cell type and the potential requirement of multiple doses to achieve sustained target repression. Viral vectors can be used to direct sustained expression, including short hairpin RNAs that can be processed in the target cell to the mature miRNA, an approach that has been used in multiple preclinical studies (280, 281). However, viral delivery systems will require careful scrutiny for clinical use. Cell-type–specific ligands, peptides, and nanoparticles are likely to provide unique delivery platforms that enable sustained and prolonged miRNA expression or knockdown.

In addition to chemical modification of miRs or antimiRs, nanotechnology is also an effective tool for systemic delivery of miRNAs and may provide enhanced specificity for targeting the diabetic vasculature. Typically, nanotechnology includes nanosized particles composed of different entities such as lipids, polymers, and inorganic materials (282, 283). These nanoparticles can also be modified to selectively deliver miRNAs. For example, to selectively deliver antimiR-712 to aortic ECs, cationic lipopartcles incorporating the peptide (VHPK) to target VCAM-1 were used, an effect resulting in inhibition of atherosclerosis by targeting TIMP3 (284). In a similar approach, loading miR-146 and miR-181b into E-selectin–targeting multistage nanoparticles effectively transported these two miRNAs to ECs, an effect that ameliorated endothelial inflammation and atherosclerosis (285). These modified nanocarriers may improve therapeutic effects and also reduce “off-target” effects, thereby providing a potential framework for targeting, for example, the diabetic microvasculature in pathologies such as diabetic wound healing, retinopathy, or impaired myocardial angiogenesis, among others. Finally, other potential vehicles for modulating miRNA expression in the vasculature include exosomes, a class of microvesicles released by cells that contain cell-specific proteins (286) that can recognize target cells (287). Therefore, engineering cell-specific exosomes enriched with a cell surface signature relevant to the diabetic microvasculature may hold promise for serving as an ideal miRNA carrier for a range of diabetic pathologies.

D. Examples of miRNA mimics and inhibitors in clinical trials

Several miRNA-based therapeutics are currently in clinical trials. The first is a locked nucleic acid directed against miR-122 (Miravirsen), which targets hepatitis C virus RNA (288). In a phase 2 study, Miraversen demonstrated dose-dependent antiviral activity maintained over four weeks (289). Another antimiR in clinical development is an inhibitor to miR-155 for cutaneous T cell lymphoma (ClinicalTrials.gov no. NCT02580552). miRNA mimics in earlier-stage clinical trials include intradermal delivery of miR-29b mimics for patients with cutaneous scleroderma (ClinicalTrials.gov no. NCT02603224) and MRX34, a miR-34 mimic encapsulated in a liposomal nanoparticle formulation, which was to be examined in patients with a range of advanced solid tumors and hematologic malignancies. Due to multiple immune-related severe adverse events, the phase 1 clinical trial investigating MRX34 was halted, raising concerns about both dosing and targets for this miR-34 mimic (ClinicalTrials.gov no. NCT01829971). Nevertheless, the results of clinical trials involving miRNA mimics or inhibitors provide new therapeutic opportunities for “fine-tuning” disease management involving the diabetic microvasculture.

In conclusion, accumulating studies highlight critical roles for miRNAs in diverse aspects of diabetic microvascular disease, including insulin resistance, diabetic wound healing, myocardial angiogenesis, diabetic retinopathy, and diabetic nephropathy, and a range of important cell signaling pathways in disease pathogenesis. miRNAs may offer new targets for early detection and therapeutic intervention, paving the way for unique means to identify and target the devastating sequelae of diabetic microvasculature disease.

Acknowledgments

This work was supported by the National Institutes of Health (HL115141, HL117994, HL134849, and GM115605; to M.W.F.), the Arthur K. Watson Charitable Trust (to M.W.F.), the Dr. Ralph and Marian Falk Medical Research Trust (to M.W.F.), the American Heart Association (SDG#15SDG25400012; to X.S.), a Boston Nutrition Obesity Research Center Pilot and Feasibility award under National Institutes of Health/National Institute of Diabetes and Digestive and Kidney Diseases P30KD046200 (to X.S.), a Watkins Discovery Award (to B.I.), and an American Diabetes Association grant (#1-16-JDF-046; to B.I.).

Disclosure Summary: The authors have nothing to disclose.

Footnotes

Abbreviations:
Akt/PKB
protein kinase B
antimiR
antisense microRNA inhibitor
BMAC
bone marrow–derived angiogenic cell
CNV
choroidal neovascularization
DAXX
death domain associated protein
DRG
dorsal root ganglion
dsRNA
double-stranded RNA
EC
endothelial cell
eNOS
endothelial nitric oxide synthase
EPC
endothelial progenitor cell
eWAT
epididymal white adipose tissue
FN
fibronectin
HuR
human antigen R
HUVEC
human umbilical vein endothelial cell
IRS
insulin receptor substrate
MAPK
mitogen-activated protein kinase
miRNA
microRNA
mRNA
messenger RNA
mTOR
mammalian target of rapamycin
NO
nitric oxide
nt
nucleotide
premiRNA
precursor-miRNA
pri-miRNA
primary-miRNA
RISC
RNA-induced silencing complex
RNase
ribonuclease
tRNA
transfer RNA
UTR
untranslated region
VSMC
vascular smooth muscle cell.

References

  • 1.Mlinar B, Marc J, Janež A, Pfeifer M. Molecular mechanisms of insulin resistance and associated diseases. Clin Chim Acta. 2007;375(1-2):20–35. [DOI] [PubMed] [Google Scholar]
  • 2.Tewari N, Awad S, Macdonald IA, Lobo DN. Obesity-related insulin resistance: implications for the surgical patient. Int J Obes. 2015;39(11):1575–1588. [DOI] [PubMed] [Google Scholar]
  • 3.DeFronzo RA. Pathogenesis of type 2 diabetes mellitus. Med Clin North Am. 2004;88(4):787–835, ix. [DOI] [PubMed] [Google Scholar]
  • 4.Zenni GC, McLane MP, Law WR, Raymond RM. Hepatic insulin resistance during chronic hyperdynamic sepsis. Circ Shock. 1992;37(3):198–208. [PubMed] [Google Scholar]
  • 5.Reynisdottir S, Ellerfeldt K, Wahrenberg H, Lithell H, Arner P. Multiple lipolysis defects in the insulin resistance (metabolic) syndrome. J Clin Invest. 1994;93(6):2590–2599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Iwatsuka H, Shino A. Studies on diabetogenic action of obesity in mice: congenital insulin resistance of KK mice. Endocrinol Jpn. 1970;17(6):535–540. [DOI] [PubMed] [Google Scholar]
  • 7.Duong M, Petit JM, Piroth L, Grappin M, Buisson M, Chavanet P, Hillon P, Portier H. Association between insulin resistance and hepatitis C virus chronic infection in HIV-hepatitis C virus-coinfected patients undergoing antiretroviral therapy. J Acquir Immune Defic Syndr. 2001;27(3):245–250. [DOI] [PubMed] [Google Scholar]
  • 8.Li XH, Gong QM, Ling Y, Huang C, Yu DM, Gu LL, Liao XW, Zhang DH, Hu XQ, Han Y, Kong XF, Zhang XX. Inherent lipid metabolic dysfunction in glycogen storage disease IIIa. Biochem Biophys Res Commun. 2014;455(1-2):90–97. [DOI] [PubMed] [Google Scholar]
  • 9.Ambros V. MicroRNAs: tiny regulators with great potential. Cell. 2001;107(7):823–826. [DOI] [PubMed] [Google Scholar]
  • 10.Chalfie M, Horvitz HR, Sulston JE. Mutations that lead to reiterations in the cell lineages of C. elegans. Cell. 1981;24(1):59–69. [DOI] [PubMed] [Google Scholar]
  • 11.Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell. 1993;75(5):843–854. [DOI] [PubMed] [Google Scholar]
  • 12.Reinhart BJ, Slack FJ, Basson M, Pasquinelli AE, Bettinger JC, Rougvie AE, Horvitz HR, Ruvkun G. The 21-nucleotide let-7 RNA regulates developmental timing in Caenorhabditis elegans. Nature. 2000;403(6772):901–906. [DOI] [PubMed] [Google Scholar]
  • 13.Lee Y, Jeon K, Lee JT, Kim S, Kim VN. MicroRNA maturation: stepwise processing and subcellular localization. EMBO J. 2002;21(17):4663–4670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Kim VN. MicroRNA biogenesis: coordinated cropping and dicing. Nat Rev Mol Cell Biol. 2005;6(5):376–385. [DOI] [PubMed] [Google Scholar]
  • 15.Lee Y, Kim M, Han J, Yeom KH, Lee S, Baek SH, and Kim VN. MicroRNA genes are transcribed by RNA polymerase II. EMBO J 2004;23(20):4051–4060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Lee Y, Ahn C, Han J, Choi H, Kim J, Yim J, Lee J, Provost P, Rådmark O, Kim S, Kim VN. The nuclear RNase III Drosha initiates microRNA processing. Nature. 2003;425(6956):415–419. [DOI] [PubMed] [Google Scholar]
  • 17.Gregory RI, Yan KP, Amuthan G, Chendrimada T, Doratotaj B, Cooch N, Shiekhattar R. The Microprocessor complex mediates the genesis of microRNAs. Nature. 2004;432(7014):235–240. [DOI] [PubMed] [Google Scholar]
  • 18.Denli AM, Tops BBJ, Plasterk RHA, Ketting RF, Hannon GJ. Processing of primary microRNAs by the Microprocessor complex. Nature. 2004;432(7014):231–235. [DOI] [PubMed] [Google Scholar]
  • 19.Han J, Lee Y, Yeom KH, Kim YK, Jin H, Kim VN. The Drosha-DGCR8 complex in primary microRNA processing. Genes Dev. 2004;18(24):3016–3027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Han J, Pedersen JS, Kwon SC, Belair CD, Kim YK, Yeom KH, Yang WY, Haussler D, Blelloch R, Kim VN. Posttranscriptional crossregulation between Drosha and DGCR8. Cell. 2009;136(1):75–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Yeom KH, Lee Y, Han J, Suh MR, Kim VN. Characterization of DGCR8/Pasha, the essential cofactor for Drosha in primary miRNA processing. Nucleic Acids Res. 2006;34(16):4622–4629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Kadener S, Rodriguez J, Abruzzi KC, Khodor YL, Sugino K, Marr MT II, Nelson S, Rosbash M. Genome-wide identification of targets of the drosha-pasha/DGCR8 complex. RNA. 2009;15(4):537–545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Lund E, Güttinger S, Calado A, Dahlberg JE, Kutay U. Nuclear export of microRNA precursors. Science. 2004;303(5654):95–98. [DOI] [PubMed] [Google Scholar]
  • 24.Calado A, Treichel N, Müller EC, Otto A, Kutay U. Exportin‐5‐mediated nuclear export of eukaryotic elongation factor 1A and tRNA. EMBO J 2002;21(22):6216–6224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Lim LP, Lau NC, Weinstein EG, Abdelhakim A, Yekta S, Rhoades MW, Burge CB, Bartel DP. The microRNAs of Caenorhabditis elegans. Genes Dev. 2003;17(8):991–1008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Yi R, Qin Y, Macara IG, Cullen BR. Exportin-5 mediates the nuclear export of pre-microRNAs and short hairpin RNAs. Genes Dev. 2003;17(24):3011–3016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Zeng Y, Cullen BR. Structural requirements for pre-microRNA binding and nuclear export by Exportin 5. Nucleic Acids Res. 2004;32(16):4776–4785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Hutvágner G, McLachlan J, Pasquinelli AE, Bálint E, Tuschl T, Zamore PD. A cellular function for the RNA-interference enzyme Dicer in the maturation of the let-7 small temporal RNA. Science. 2001;293(5531):834–838. [DOI] [PubMed] [Google Scholar]
  • 29.Grishok A, Pasquinelli AE, Conte D, Li N, Parrish S, Ha I, Baillie DL, Fire A, Ruvkun G, Mello CC. Genes and mechanisms related to RNA interference regulate expression of the small temporal RNAs that control C. elegans developmental timing. Cell. 2001;106(1):23–34. [DOI] [PubMed] [Google Scholar]
  • 30.Ketting RF, Fischer SE, Bernstein E, Sijen T, Hannon GJ, Plasterk RH. Dicer functions in RNA interference and in synthesis of small RNA involved in developmental timing in C. elegans. Genes Dev. 2001;15(20):2654–2659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Zhang H, Kolb FA, Jaskiewicz L, Westhof E, Filipowicz W. Single processing center models for human Dicer and bacterial RNase III. Cell. 2004;118(1):57–68. [DOI] [PubMed] [Google Scholar]
  • 32.Jaskiewicz L, Filipowicz W, Paddison P, and Vogt P, eds. RNA Interference. Springer Berlin Heidelberg; 2008:77–97. [Google Scholar]
  • 33.Macrae IJ, Zhou K, Li F, Repic A, Brooks AN, Cande WZ, Adams PD, Doudna JA. Structural basis for double-stranded RNA processing by Dicer. Science. 2006;311(5758):195–198. [DOI] [PubMed] [Google Scholar]
  • 34.MacRae IJ, Zhou K, Doudna JA. Structural determinants of RNA recognition and cleavage by Dicer. Nat Struct Mol Biol. 2007;14(10):934–940. [DOI] [PubMed] [Google Scholar]
  • 35.Park JE, Heo I, Tian Y, Simanshu DK, Chang H, Jee D, Patel DJ, Kim VN. Dicer recognizes the 5′ end of RNA for efficient and accurate processing. Nature. 2011;475(7355):201–205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Carthew RW, Sontheimer EJ. Origins and mechanisms of miRNAs and siRNAs. Cell. 2009;136(4):642–655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Finnegan EF, Pasquinelli AE. MicroRNA biogenesis: regulating the regulators. Crit Rev Biochem Mol Biol. 2013;48(1):51–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Rawal S, Manning P, Katare R. Cardiovascular microRNAs: as modulators and diagnostic biomarkers of diabetic heart disease. Cardiovasc Diabetol. 2014;13(1):44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Khvorova A, Reynolds A, Jayasena SD. Functional siRNAs and miRNAs exhibit strand bias. Cell. 2003;115(2):209–216. [DOI] [PubMed] [Google Scholar]
  • 40.Schwarz DS, Hutvágner G, Du T, Xu Z, Aronin N, Zamore PD. Asymmetry in the assembly of the RNAi enzyme complex. Cell. 2003;115(2):199–208. [DOI] [PubMed] [Google Scholar]
  • 41.Okamura K, Phillips MD, Tyler DM, Duan H, Chou YT, Lai EC. The regulatory activity of microRNA* species has substantial influence on microRNA and 3′ UTR evolution. Nat Struct Mol Biol. 2008;15(4):354–363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Winter J, Diederichs S. Argonaute-3 activates the let-7a passenger strand microRNA. RNA Biol. 2013;10(10):1631–1643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Hammond SM, Boettcher S, Caudy AA, Kobayashi R, Hannon GJ. Argonaute2, a link between genetic and biochemical analyses of RNAi. Science. 2001;293(5532):1146–1150. [DOI] [PubMed] [Google Scholar]
  • 44.Zeng Y, Wagner EJ, Cullen BR. Both natural and designed micro RNAs can inhibit the expression of cognate mRNAs when expressed in human cells. Mol Cell. 2002;9(6):1327–1333. [DOI] [PubMed] [Google Scholar]
  • 45.Zeng Y, Yi R, Cullen BR. MicroRNAs and small interfering RNAs can inhibit mRNA expression by similar mechanisms. Proc Natl Acad Sci USA. 2003;100(17):9779–9784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Lim LP, Lau NC, Garrett-Engele P, Grimson A, Schelter JM, Castle J, Bartel DP, Linsley PS, Johnson JM. Microarray analysis shows that some microRNAs downregulate large numbers of target mRNAs. Nature. 2005;433(7027):769–773. [DOI] [PubMed] [Google Scholar]
  • 47.Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell. 2009;136(2):215–233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Wightman B, Ha I, Ruvkun G. Posttranscriptional regulation of the heterochronic gene lin-14 by lin-4 mediates temporal pattern formation in C. elegans. Cell. 1993;75(5):855–862. [DOI] [PubMed] [Google Scholar]
  • 49.Wightman B, Bürglin TR, Gatto J, Arasu P, Ruvkun G. Negative regulatory sequences in the lin-14 3′-untranslated region are necessary to generate a temporal switch during Caenorhabditis elegans development. Genes Dev. 1991;5(10):1813–1824. [DOI] [PubMed] [Google Scholar]
  • 50.Brennecke J, Stark A, Russell RB, Cohen SM. Principles of microRNA-target recognition. PLoS Biol. 2005;3(3):e85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Lewis BP, Burge CB, Bartel DP. Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell. 2005;120(1):15–20. [DOI] [PubMed] [Google Scholar]
  • 52.Wheeler BM, Heimberg AM, Moy VN, Sperling EA, Holstein TW, Heber S, Peterson KJ. The deep evolution of metazoan microRNAs. Evol Dev. 2009;11(1):50–68. [DOI] [PubMed] [Google Scholar]
  • 53.Meister G, Tuschl T. Mechanisms of gene silencing by double-stranded RNA. Nature. 2004;431(7006):343–349. [DOI] [PubMed] [Google Scholar]
  • 54.Martinez J, Patkaniowska A, Urlaub H, Lührmann R, Tuschl T. Single-stranded antisense siRNAs guide target RNA cleavage in RNAi. Cell. 2002;110(5):563–574. [DOI] [PubMed] [Google Scholar]
  • 55.Yekta S, Shih IH, Bartel DP. MicroRNA-directed cleavage of HOXB8 mRNA. Science. 2004;304(5670):594–596. [DOI] [PubMed] [Google Scholar]
  • 56.Wakiyama M, Takimoto K, Ohara O, Yokoyama S. Let-7 microRNA-mediated mRNA deadenylation and translational repression in a mammalian cell-free system. Genes Dev. 2007;21(15):1857–1862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Rehwinkel J, Behm-Ansmant I, Gatfield D, Izaurralde E. A crucial role for GW182 and the DCP1:DCP2 decapping complex in miRNA-mediated gene silencing. RNA. 2005;11(11):1640–1647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Behm-Ansmant I, Rehwinkel J, Doerks T, Stark A, Bork P, Izaurralde E. mRNA degradation by miRNAs and GW182 requires both CCR4:NOT deadenylase and DCP1:DCP2 decapping complexes. Genes Dev. 2006;20(14):1885–1898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Wu L, Fan J, Belasco JG. MicroRNAs direct rapid deadenylation of mRNA. Proc Natl Acad Sci USA. 2006;103(11):4034–4039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Bagga S, Bracht J, Hunter S, Massirer K, Holtz J, Eachus R, Pasquinelli AE. Regulation by let-7 and lin-4 miRNAs results in target mRNA degradation. Cell. 2005;122(4):553–563. [DOI] [PubMed] [Google Scholar]
  • 61.Rieser P. Tryptophan residues may be at the insulin receptor site in muscle. Life Sci. 1967;6(12):1269–1275. [DOI] [PubMed] [Google Scholar]
  • 62.Freychet P, Roth J, Neville DM Jr. Insulin receptors in the liver: specific binding of (125 I)insulin to the plasma membrane and its relation to insulin bioactivity. Proc Natl Acad Sci USA. 1971;68(8):1833–1837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Zick Y. The insulin receptor: structure and function. Crit Rev Biochem Mol Biol. 1989;24(3):217–269. [DOI] [PubMed] [Google Scholar]
  • 64.Arakaki RF, Hedo JA, Collier E, Gorden P. Effects of castanospermine and 1-deoxynojirimycin on insulin receptor biogenesis. Evidence for a role of glucose removal from core oligosaccharides. J Biol Chem. 1987;262(24):11886–11892. [PubMed] [Google Scholar]
  • 65.Cuatrecasas P. Affinity chromatography and purification of the insulin receptor of liver cell membranes. Proc Natl Acad Sci USA. 1972;69(5):1277–1281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Zierath JR, Krook A, Wallberg-Henriksson H. Insulin action and insulin resistance in human skeletal muscle. Diabetologia. 2000;43(7):821–835. [DOI] [PubMed] [Google Scholar]
  • 67.Rosen OM. Insulin receptor as a tyrosine protein kinase. Ann N Y Acad Sci. 1986;463(1):13–19. [DOI] [PubMed] [Google Scholar]
  • 68.Kubar J, Van Obberghen E. Oligomeric states of the insulin receptor: binding and autophosphorylation properties. Biochemistry. 1989;28(3):1086–1093. [DOI] [PubMed] [Google Scholar]
  • 69.Mynarcik DC, Whittaker J. Insulin receptor transmembrane signaling: evidence for an intermolecular oligomerization mechanism of activation. J Recept Signal Transduct Res. 1995;15(7-8):887–904. [DOI] [PubMed] [Google Scholar]
  • 70.Sun XJ, Rothenberg P, Kahn CR, Backer JM, Araki E, Wilden PA, Cahill DA, Goldstein BJ, White MF. Structure of the insulin receptor substrate IRS-1 defines a unique signal transduction protein. Nature. 1991;352(6330):73–77. [DOI] [PubMed] [Google Scholar]
  • 71.Sun XJ, Wang L-M, Zhang Y, Yenush L, Myers MG Jr, Glasheen E, Lane WS, Pierce JH, White MF. Role of IRS-2 in insulin and cytokine signalling. Nature. 1995;377(6545):173–177. [DOI] [PubMed] [Google Scholar]
  • 72.Lavan BE, Lane WS, Lienhard GE. The 60-kDa phosphotyrosine protein in insulin-treated adipocytes is a new member of the insulin receptor substrate family. J Biol Chem. 1997;272(17):11439–11443. [DOI] [PubMed] [Google Scholar]
  • 73.Lavan BE, Fantin VR, Chang ET, Lane WS, Keller SR, Lienhard GE. A novel 160-kDa phosphotyrosine protein in insulin-treated embryonic kidney cells is a new member of the insulin receptor substrate family. J Biol Chem. 1997;272(34):21403–21407. [DOI] [PubMed] [Google Scholar]
  • 74.Cai D, Dhe-Paganon S, Melendez PA, Lee J, Shoelson SE. Two new substrates in insulin signaling, IRS5/DOK4 and IRS6/DOK5. J Biol Chem. 2003;278(28):25323–25330. [DOI] [PubMed] [Google Scholar]
  • 75.Shepherd PR, Withers DJ, Siddle K. Phosphoinositide 3-kinase: the key switch mechanism in insulin signalling. Biochem J. 1998;333(3):471–490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Chiang SH, Baumann CA, Kanzaki M, Thurmond DC, Watson RT, Neudauer CL, Macara IG, Pessin JE, Saltiel AR. Insulin-stimulated GLUT4 translocation requires the CAP-dependent activation of TC10. Nature. 2001;410(6831):944–948. [DOI] [PubMed] [Google Scholar]
  • 77.Yang Y, Farese RV. Insulin activates myelin basic protein (p42 MAP) kinase by a protein kinase C-independent pathway in rat adipocytes. Dissociation from glucose transport. FEBS Lett. 1993;333(3):287–290. [DOI] [PubMed] [Google Scholar]
  • 78.Shaw LM. Identification of insulin receptor substrate 1 (IRS-1) and IRS-2 as signaling intermediates in the α6β4 integrin-dependent activation of phosphoinositide 3-OH kinase and promotion of invasion. Mol Cell Biol. 2001;21(15):5082–5093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Qu BH, Karas M, Koval A, LeRoith D. Insulin receptor substrate-4 enhances insulin-like growth factor-I-induced cell proliferation. J Biol Chem. 1999;274(44):31179–31184. [DOI] [PubMed] [Google Scholar]
  • 80.Stephens L, Eguinoa A, Corey S, Jackson T, Hawkins PT. Receptor stimulated accumulation of phosphatidylinositol (3,4,5)-trisphosphate by G-protein mediated pathways in human myeloid derived cells. EMBO J. 1993;12(6):2265–2273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Stephens LR, Hughes KT, Irvine RF. Pathway of phosphatidylinositol(3,4,5)-trisphosphate synthesis in activated neutrophils. Nature. 1991;351(6321):33–39. [DOI] [PubMed] [Google Scholar]
  • 82.Currie RA, Walker KS, Gray A, Deak M, Casamayor A, Downes CP, Cohen P, Alessi DR, Lucocq J. Role of phosphatidylinositol 3,4,5-trisphosphate in regulating the activity and localization of 3-phosphoinositide-dependent protein kinase-1. Biochem J. 1999;337(3):575–583. [PMC free article] [PubMed] [Google Scholar]
  • 83.Alessi DR, Cohen P. Mechanism of activation and function of protein kinase B. Curr Opin Genet Dev. 1998;8(1):55–62. [DOI] [PubMed] [Google Scholar]
  • 84.Banfić H, Tang X, Batty IH, Downes CP, Chen C, Rittenhouse SE. A novel integrin-activated pathway forms PKB/Akt-stimulatory phosphatidylinositol 3,4-bisphosphate via phosphatidylinositol 3-phosphate in platelets. J Biol Chem. 1998;273(1):13–16. [DOI] [PubMed] [Google Scholar]
  • 85.Egawa K, Maegawa H, Shi K, Nakamura T, Obata T, Yoshizaki T, Morino K, Shimizu S, Nishio Y, Suzuki E, Kashiwagi A. Membrane localization of 3-phosphoinositide-dependent protein kinase-1 stimulates activities of Akt and atypical protein kinase C but does not stimulate glucose transport and glycogen synthesis in 3T3-L1 adipocytes. J Biol Chem. 2002;277(41):38863–38869. [DOI] [PubMed] [Google Scholar]
  • 86.Cross DAE, Alessi DR, Cohen P, Andjelkovich M, Hemmings BA. Inhibition of glycogen synthase kinase-3 by insulin mediated by protein kinase B. Nature. 1995;378(6559):785–789. [DOI] [PubMed] [Google Scholar]
  • 87.Wang D, Sul HS. Insulin stimulation of the fatty acid synthase promoter is mediated by the phosphatidylinositol 3-kinase pathway. Involvement of protein kinase B/Akt. J Biol Chem. 1998;273(39):25420–25426. [DOI] [PubMed] [Google Scholar]
  • 88.Agati JM, Yeagley D, Quinn PG. Assessment of the roles of mitogen-activated protein kinase, phosphatidylinositol 3-kinase, protein kinase B, and protein kinase C in insulin inhibition of cAMP-induced phosphoenolpyruvate carboxykinase gene transcription. J Biol Chem. 1998;273(30):18751–18759. [DOI] [PubMed] [Google Scholar]
  • 89.Hill MM, Clark SF, Tucker DF, Birnbaum MJ, James DE, Macaulay SL. A role for protein kinase Bbeta/Akt2 in insulin-stimulated GLUT4 translocation in adipocytes. Mol Cell Biol. 1999;19(11):7771–7781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Scott PH, Brunn GJ, Kohn AD, Roth RA, Lawrence JC Jr. Evidence of insulin-stimulated phosphorylation and activation of the mammalian target of rapamycin mediated by a protein kinase B signaling pathway. Proc Natl Acad Sci USA. 1998;95(13):7772–7777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Dennis PB, Fumagalli S, Thomas G. Target of rapamycin (TOR): balancing the opposing forces of protein synthesis and degradation. Curr Opin Genet Dev. 1999;9(1):49–54. [DOI] [PubMed] [Google Scholar]
  • 92.Ribon V, Printen JA, Hoffman NG, Kay BK, Saltiel ARA. A novel, multifuntional c-Cbl binding protein in insulin receptor signaling in 3T3-L1 adipocytes. Mol Cell Biol. 1998;18(2):872–879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Baumann CA, Ribon V, Kanzaki M, Thurmond DC, Mora S, Shigematsu S, Bickel PE, Pessin JE, Saltiel AR. CAP defines a second signalling pathway required for insulin-stimulated glucose transport. Nature. 2000;407(6801):202–207. [DOI] [PubMed] [Google Scholar]
  • 94.Skolnik EY, Batzer A, Li N, Lee CH, Lowenstein E, Mohammadi M, Margolis B, Schlessinger J. The function of GRB2 in linking the insulin receptor to Ras signaling pathways. Science. 1993;260(5116):1953–1955. [DOI] [PubMed] [Google Scholar]
  • 95.Xu H, Goldfarb M. Multiple effector domains within SNT1 coordinate ERK activation and neuronal differentiation of PC12 cells. J Biol Chem. 2001;276(16):13049–13056. [DOI] [PubMed] [Google Scholar]
  • 96.Avruch J, Khokhlatchev A, Kyriakis J, Luo Z, Tzivion G, Vavvas D, and Zhang X. Ras activation of the Raf kinase: tyrosine kinase recruitment of the MAP kinase cascade. Recent Prog Horm Res. 2001;56:127–155. [DOI] [PubMed] [Google Scholar]
  • 97.Garg A. Regional adiposity and insulin resistance. J Clin Endocrinol Metab. 2004;89(9):4206–4210. [DOI] [PubMed] [Google Scholar]
  • 98.Patel P, Abate N. Body fat distribution and insulin resistance. Nutrients. 2013;5(6):2019–2027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Hall AM, Kou K, Chen Z, Pietka TA, Kumar M, Korenblat KM, Lee K, Ahn K, Fabbrini E, Klein S, Goodwin B, Finck BN. Evidence for regulated monoacylglycerol acyltransferase expression and activity in human liver. J Lipid Res. 2012;53(5):990–999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Hamdy O, Porramatikul S, Al-Ozairi E. Metabolic obesity: the paradox between visceral and subcutaneous fat. Curr Diabetes Rev. 2006;2(4):367–373. [DOI] [PubMed] [Google Scholar]
  • 101.Sun X, Lin J, Zhang Y, Kang S, Belkin N, Wara AK, Icli B, Hamburg NM, Li D, Feinberg MW. MicroRNA-181b improves glucose homeostasis and insulin sensitivity by regulating endothelial function in white adipose tissue. Circ Res. 2016;118(5):810–821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Sun X, Icli B, Wara AK, Belkin N, He S, Kobzik L, Hunninghake GM, Vera MP, Blackwell TS, Baron RM, Feinberg MW; MICU Registry . MicroRNA-181b regulates NF-κB-mediated vascular inflammation. J Clin Invest. 2012;122(6):1973–1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Kim B, Feldman EL. Insulin resistance as a key link for the increased risk of cognitive impairment in the metabolic syndrome. Exp Mol Med. 2015;47(3):e149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Gruben N, Shiri-Sverdlov R, Koonen DPY, Hofker MH. Nonalcoholic fatty liver disease: A main driver of insulin resistance or a dangerous liaison? Biochim Biophys Acta. 2014;1842(11):2329–2343. [DOI] [PubMed] [Google Scholar]
  • 105.DeFronzo RA, Gunnarsson R, Björkman O, Olsson M, Wahren J. Effects of insulin on peripheral and splanchnic glucose metabolism in noninsulin-dependent (type II) diabetes mellitus. J Clin Invest. 1985;76(1):149–155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Gallagher IJ, Scheele C, Keller P, Nielsen AR, Remenyi J, Fischer CP, Roder K, Babraj J, Wahlestedt C, Hutvagner G, Pedersen BK, Timmons JA. Integration of microRNA changes in vivo identifies novel molecular features of muscle insulin resistance in type 2 diabetes. Genome Med. 2010;2(2):9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Ferreira DMS, Simão AL, Rodrigues CMP, Castro RE. Revisiting the metabolic syndrome and paving the way for microRNAs in non-alcoholic fatty liver disease. FEBS J. 2014;281(11):2503–2524. [DOI] [PubMed] [Google Scholar]
  • 108.Xu Q, Li Y, Shang YF, Wang HL, Yao MX. miRNA-103: molecular link between insulin resistance and nonalcoholic fatty liver disease. World J Gastroenterol. 2015;21(2):511–516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Zhao E, Keller MP, Rabaglia ME, Oler AT, Stapleton DS, Schueler KL, Neto EC, Moon JY, Wang P, Wang IM, Lum P, Ivanovska I, Greenawalt D, Tsang J, Choi Y, Kleinhanz R, Shang J, Zhou YP, Howard AD, Zhang BB, Kenziorski C, Thornberry NA, Yandell BS, Schadt EE, Attie AD. Obesity and genetics regulate microRNAs in islets, liver and adipose of diabetic mice. Mamm Genome. 2009;20(8):476–485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Liang T, Liu C, Ye Z. Deep sequencing of small RNA repertoires in mice reveals metabolic disorders-associated hepatic miRNAs. PLoS One. 2013;8(11):e80774. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Wei M, Gaskill SP, Haffner SM, Stern MP. Effects of diabetes and level of glycemia on all-cause and cardiovascular mortality. The San Antonio Heart Study. Diabetes Care. 1998;21(7):1167–1172. [DOI] [PubMed] [Google Scholar]
  • 112.Piga R, Naito Y, Kokura S, Handa O, Yoshikawa T. Short-term high glucose exposure induces monocyte-endothelial cells adhesion and transmigration by increasing VCAM-1 and MCP-1 expression in human aortic endothelial cells. Atherosclerosis. 2007;193(2):328–334. [DOI] [PubMed] [Google Scholar]
  • 113.Quagliaro L, Piconi L, Assaloni R, Da Ros R, Maier A, Zuodar G, Ceriello A. Intermittent high glucose enhances ICAM-1, VCAM-1 and E-selectin expression in human umbilical vein endothelial cells in culture: the distinct role of protein kinase C and mitochondrial superoxide production. Atherosclerosis. 2005;183(2):259–267. [DOI] [PubMed] [Google Scholar]
  • 114.Ejaz S, Chekarova I, Ejaz A, Sohail A, Lim CW. Importance of pericytes and mechanisms of pericyte loss during diabetes retinopathy. Diabetes Obes Metab. 2008;10(1):53–63. [DOI] [PubMed] [Google Scholar]
  • 115.Schratzberger P, Walter DH, Rittig K, Bahlmann FH, Pola R, Curry C, Silver M, Krainin JG, Weinberg DH, Ropper AH, Isner JM. Reversal of experimental diabetic neuropathy by VEGF gene transfer. J Clin Invest. 2001;107(9):1083–1092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Wolf G, Chen S, Ziyadeh FN. From the periphery of the glomerular capillary wall toward the center of disease: podocyte injury comes of age in diabetic nephropathy. Diabetes. 2005;54(6):1626–1634. [DOI] [PubMed] [Google Scholar]
  • 117.Orasanu G, Plutzky J. The pathologic continuum of diabetic vascular disease. J Am Coll Cardiol. 2009;53(5, Suppl)S35–S42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Zampetaki A, Kiechl S, Drozdov I, Willeit P, Mayr U, Prokopi M, Mayr A, Weger S, Oberhollenzer F, Bonora E, Shah A, Willeit J, Mayr M. Plasma microRNA profiling reveals loss of endothelial miR-126 and other microRNAs in type 2 diabetes. Circ Res. 2010;107(6):810–817. [DOI] [PubMed] [Google Scholar]
  • 119.Syed Ikmal SIQ, Zaman Huri H, Vethakkan SR, Wan Ahmad WA. Potential biomarkers of insulin resistance and atherosclerosis in type 2 diabetes mellitus patients with coronary artery disease. Int J Endocrinol. 2013;2013:698567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Kim JA, Montagnani M, Koh KK, Quon MJ. Reciprocal relationships between insulin resistance and endothelial dysfunction: molecular and pathophysiological mechanisms. Circulation. 2006;113(15):1888–1904. [DOI] [PubMed] [Google Scholar]
  • 121.Xu H, Barnes GT, Yang Q, Tan G, Yang D, Chou CJ, Sole J, Nichols A, Ross JS, Tartaglia LA, Chen H. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J Clin Invest. 2003;112(12):1821–1830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Dantas APV, Fortes ZB, and de Carvalho MHC. Vascular disease in diabetic women: why do they miss the female protection? Exp Diabetes Res. 2012;2012:570598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Kim F, Gallis B, Corson MA. TNF-α inhibits flow and insulin signaling leading to NO production in aortic endothelial cells. Am J Physiol Cell Physiol. 2001;280(5):C1057–1065 [DOI] [PubMed] [Google Scholar]
  • 124.Anderson HDI, Rahmutula D, Gardner DG. Tumor necrosis factor-α inhibits endothelial nitric-oxide synthase gene promoter activity in bovine aortic endothelial cells. J Biol Chem. 2004;279(2):963–969. [DOI] [PubMed] [Google Scholar]
  • 125.Li X, Commane M, Jiang Z, Stark GR. IL-1-induced NFkappa B and c-Jun N-terminal kinase (JNK) activation diverge at IL-1 receptor-associated kinase (IRAK). Proc Natl Acad Sci USA. 2001;98(8):4461–4465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Hasegawa Y, Saito T, Ogihara T, Ishigaki Y, Yamada T, Imai J, Uno K, Gao J, Kaneko K, Shimosawa T, Asano T, Fujita T, Oka Y, Katagiri H. Blockade of the nuclear factor-κB pathway in the endothelium prevents insulin resistance and prolongs life spans. Circulation. 2012;125(9):1122–1133. [DOI] [PubMed] [Google Scholar]
  • 127.Feng B, Chen S, McArthur K, Wu Y, Sen S, Ding Q, Feldman RD, Chakrabarti S. miR-146a-Mediated extracellular matrix protein production in chronic diabetes complications. Diabetes. 2011;60(11):2975–2984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Karolina DS, Armugam A, Tavintharan S, Wong MTK, Lim SC, Sum CF, Jeyaseelan K. MicroRNA 144 impairs insulin signaling by inhibiting the expression of insulin receptor substrate 1 in type 2 diabetes mellitus. PLoS One. 2011;6(8):e22839. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Olivieri F, Lazzarini R, Recchioni R, Marcheselli F, Rippo MR, Di Nuzzo S, Albertini MC, Graciotti L, Babini L, Mariotti S, Spada G, Abbatecola AM, Antonicelli R, Franceschi C, Procopio AD. miR-146a as marker of senescence-associated pro-inflammatory status in cells involved in vascular remodelling. Age (Dordr). 2013;35(4):1157–1172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Wang HJ, Lo WY, Lin LJ. Angiotensin-(1-7) decreases glycated albumin-induced endothelial interleukin-6 expression via modulation of miR-146a. Biochem Biophys Res Commun. 2013;430(3):1157–1163. [DOI] [PubMed] [Google Scholar]
  • 131.Yudkin JS, Kumari M, Humphries SE, Mohamed-Ali V. Inflammation, obesity, stress and coronary heart disease: is interleukin-6 the link? Atherosclerosis. 2000;148(2):209–214. [DOI] [PubMed] [Google Scholar]
  • 132.Kwon DN, Chang B-, Kim JH. MicroRNA dysregulation in liver and pancreas of CMP-Neu5Ac hydroxylase null mice disrupts insulin/PI3K-AKT signaling. Biomed Res Int. 2014;2014:236385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Sun HX, Zeng DY, Li RT, Pang RP, Yang H, Hu YL, Zhang Q, Jiang Y, Huang LY, Tang YB, Yan GJ, Zhou JG. Essential role of microRNA-155 in regulating endothelium-dependent vasorelaxation by targeting endothelial nitric oxide synthase. Hypertension. 2012;60(6):1407–1414. [DOI] [PubMed] [Google Scholar]
  • 134.Wu XY, Fan WD, Fang R, Wu GF. Regulation of microRNA-155 in endothelial inflammation by targeting nuclear factor (NF)-κB P65. J Cell Biochem. 2014;115(11):1928–1936. [DOI] [PubMed] [Google Scholar]
  • 135.Jansen F, Yang X, Hoelscher M, Cattelan A, Schmitz T, Proebsting S, Wenzel D, Vosen S, Franklin BS, Fleischmann BK, Nickenig G, Werner N. Endothelial microparticle-mediated transfer of MicroRNA-126 promotes vascular endothelial cell repair via SPRED1 and is abrogated in glucose-damaged endothelial microparticles. Circulation. 2013;128(18):2026–2038. [DOI] [PubMed] [Google Scholar]
  • 136.Meng S, Cao JT, Zhang B, Zhou Q, Shen CX, Wang CQ. Downregulation of microRNA-126 in endothelial progenitor cells from diabetes patients, impairs their functional properties, via target gene Spred-1. J Mol Cell Cardiol. 2012;53(1):64–72. [DOI] [PubMed] [Google Scholar]
  • 137.Barutta F, Bruno G, Matullo G, Chaturvedi N, Grimaldi S, Schalkwijk C, Stehouwer CD, Fuller JH, Gruden G. MicroRNA-126 and micro-/macrovascular complications of type 1 diabetes in the EURODIAB Prospective Complications Study. Acta Diabetol. 2016;54(2):133–139. [DOI] [PubMed] [Google Scholar]
  • 138.Bao MH, Zhang YW, Lou XY, Cheng Y, Zhou HH. Protective effects of let-7a and let-7b on oxidized low-density lipoprotein induced endothelial cell injuries. PLoS One. 2014;9(9):e106540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Moura J, Børsheim E, Carvalho E. The role of microRNAs in diabetic complications-special emphasis on wound healing. Genes (Basel). 2014;5(4):926–956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Steppan CM, Bailey ST, Bhat S, Brown EJ, Banerjee RR, Wright CM, Patel HR, Ahima RS, Lazar MA. The hormone resistin links obesity to diabetes. Nature. 2001;409(6818):307–312. [DOI] [PubMed] [Google Scholar]
  • 141.Feng B, Cao Y, Chen S, Ruiz M, Chakrabarti S. miRNA-1 regulates endothelin-1 in diabetes. Life Sci. 2014;98(1):18–23. [DOI] [PubMed] [Google Scholar]
  • 142.Maurer LM, Tomasini-Johansson BR, Mosher DF. Emerging roles of fibronectin in thrombosis. Thromb Res. 2010;125(4):287–291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Sun X, He S, Wara AK, Icli B, Shvartz E, Tesmenitsky Y, Belkin N, Li D, Blackwell TS, Sukhova GK, Croce K, Feinberg MW. Systemic delivery of microRNA-181b inhibits nuclear factor-κB activation, vascular inflammation, and atherosclerosis in apolipoprotein E-deficient mice. Circ Res. 2014;114(1):32–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Lin J, He S, Sun X, Franck G, Deng Y, Yang D, Haemmig S, Wara AK, Icli B, Li D, Feinberg MW. MicroRNA-181b inhibits thrombin-mediated endothelial activation and arterial thrombosis by targeting caspase recruitment domain family member 10. FASEB J. 2016;30(9):3216–3226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Sun X, Sit A, Feinberg MW. Role of miR-181 family in regulating vascular inflammation and immunity. Trends Cardiovasc Med. 2014;24(3):105–112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Sun X, Belkin N, Feinberg MW. Endothelial microRNAs and atherosclerosis. Curr Atheroscler Rep. 2013;15(12):372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Faries PL, Rohan DI, Takahara H, Wyers MC, Contreras MA, Quist WC, King GL, Logerfo FW. Human vascular smooth muscle cells of diabetic origin exhibit increased proliferation, adhesion, and migration. J Vasc Surg. 2001;33(3):601–607. [DOI] [PubMed] [Google Scholar]
  • 148.Johansson GS, Arnqvist HJ. Insulin and IGF-I action on insulin receptors, IGF-I receptors, and hybrid insulin/IGF-I receptors in vascular smooth muscle cells. Am J Physiol Endocrinol Metab. 2006; 291(5):E1124–E1130. [DOI] [PubMed] [Google Scholar]
  • 149.Muniyappa R, Montagnani M, Koh KK, Quon MJ. Cardiovascular actions of insulin. Endocr Rev. 2007;28(5):463–491. [DOI] [PubMed] [Google Scholar]
  • 150.Wang CCL, Gurevich I, Draznin B. Insulin affects vascular smooth muscle cell phenotype and migration via distinct signaling pathways. Diabetes. 2003;52(10):2562–2569. [DOI] [PubMed] [Google Scholar]
  • 151.Peuler JD, Phare SM, Iannucci AR, Hodorek MJ. Differential inhibitory effects of antidiabetic drugs on arterial smooth muscle cell proliferation. Am J Hypertens. 1996;9(2):188–192. [DOI] [PubMed] [Google Scholar]
  • 152.Doronzo G, Russo I, Mattiello L, Anfossi G, Bosia A, Trovati M. Insulin activates vascular endothelial growth factor in vascular smooth muscle cells: influence of nitric oxide and of insulin resistance. Eur J Clin Invest. 2004;34(10):664–673. [DOI] [PubMed] [Google Scholar]
  • 153.Riches K, Alshanwani AR, Warburton P, O’Regan DJ, Ball SG, Wood IC, Turner NA, Porter KE. Elevated expression levels of miR-143/5 in saphenous vein smooth muscle cells from patients with Type 2 diabetes drive persistent changes in phenotype and function. J Mol Cell Cardiol. 2014;74:240–250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Ling HY, Hu B, Hu XB, Zhong J, Feng SD, Qin L, Liu G, Wen GB, Liao DF. miRNA-21 reverses high glucose and high insulin induced insulin resistance in 3T3-L1 adipocytes through targeting phosphatase and tensin homologue. Exp Clin Endocrinol Diabetes. 2012;120(9):553–559. [DOI] [PubMed] [Google Scholar]
  • 155.Yang G, Pei Y, Cao Q, Wang R. MicroRNA-21 represses human cystathionine gamma-lyase expression by targeting at specificity protein-1 in smooth muscle cells. J Cell Physiol. 2012;227(9):3192–3200. [DOI] [PubMed] [Google Scholar]
  • 156.Villeneuve LM, Kato M, Reddy MA, Wang M, Lanting L, Natarajan R. Enhanced levels of microRNA-125b in vascular smooth muscle cells of diabetic db/db mice lead to increased inflammatory gene expression by targeting the histone methyltransferase Suv39h1. Diabetes. 2010;59(11):2904–2915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Reddy MA, Jin W, Villeneuve L, Wang M, Lanting L, Todorov I, Kato M, Natarajan R. Pro-inflammatory role of microRNA-200 in vascular smooth muscle cells from diabetic mice. Arterioscler Thromb Vasc Biol. 2012;32(3):721–729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Kraakman MJ, Murphy AJ, Jandeleit-Dahm K, Kammoun HL. Macrophage polarization in obesity and type 2 diabetes: weighing down our understanding of macrophage function? Front Immunol. 2014;5:470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Chang RC, Ying W, Bazer FW, Zhou B. MicroRNAs control macrophage formation and activation: the inflammatory link between obesity and cardiovascular diseases. Cells. 2014;3(3):702–712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Stöhr R, Federici M. Insulin resistance and atherosclerosis: convergence between metabolic pathways and inflammatory nodes. Biochem J. 2013;454(1):1–11. [DOI] [PubMed] [Google Scholar]
  • 161.Chawla A, Boisvert WA, Lee CH, Laffitte BA, Barak Y, Joseph SB, Liao D, Nagy L, Edwards PA, Curtiss LK, Evans RM, Tontonoz P. A PPAR γ-LXR-ABCA1 pathway in macrophages is involved in cholesterol efflux and atherogenesis. Mol Cell. 2001;7(1):161–171. [DOI] [PubMed] [Google Scholar]
  • 162.Laffitte BA, Repa JJ, Joseph SB, Wilpitz DC, Kast HR, Mangelsdorf DJ, Tontonoz P. LXRs control lipid-inducible expression of the apolipoprotein E gene in macrophages and adipocytes. Proc Natl Acad Sci USA. 2001;98(2):507–512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Song C, Hiipakka RA, Liao S. Auto-oxidized cholesterol sulfates are antagonistic ligands of liver X receptors: implications for the development and treatment of atherosclerosis. Steroids. 2001;66(6):473–479. [DOI] [PubMed] [Google Scholar]
  • 164.Tian FJ, An LN, Wang GK, Zhu JQ, Li Q, Zhang YY, Zeng A, Zou J, Zhu RF, Han XS, Shen N, Yang HT, Zhao XX, Huang S, Qin YW, Jing Q. Elevated microRNA-155 promotes foam cell formation by targeting HBP1 in atherogenesis. Cardiovasc Res. 2014;103(1):100–110. [DOI] [PubMed] [Google Scholar]
  • 165.Di Gregoli K, Jenkins N, Salter R, White S, Newby AC, Johnson JL. MicroRNA-24 regulates macrophage behavior and retards atherosclerosis. Arterioscler Thromb Vasc Biol. 2014;34(9):1990–2000. [DOI] [PubMed] [Google Scholar]
  • 166.Tarr JM, Kaul K, Chopra M, Kohner EM, Chibber R. Pathophysiology of diabetic retinopathy. ISRN Ophthalmol. 2013;2013:343560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.D’Amore PA, Thompson RW. Mechanisms of angiogenesis. Annu Rev Physiol. 1987;49(1):453–464. [DOI] [PubMed] [Google Scholar]
  • 168.Klagsbrun M, D’Amore PA. Regulators of angiogenesis. Annu Rev Physiol. 1991;53(1):217–239. [DOI] [PubMed] [Google Scholar]
  • 169.McArthur K, Feng B, Wu Y, Chen S, Chakrabarti S. MicroRNA-200b regulates vascular endothelial growth factor-mediated alterations in diabetic retinopathy. Diabetes. 2011;60(4):1314–1323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Ye P, Liu J, He F, Xu W, Yao K. Hypoxia-induced deregulation of miR-126 and its regulative effect on VEGF and MMP-9 expression. Int J Med Sci. 2013;11(1):17–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Zeng J, Xiong Y, Li G, Liu M, He T, Tang Y, Chen Y, Cai L, Jiang R, Tao J. miR-21 is overexpressed in response to high glucose and protects endothelial cells from apoptosis. Exp Clin Endocrinol Diabetes. 2013;121(7):425–430. [DOI] [PubMed] [Google Scholar]
  • 172.Meng S, Cao I, Zhang X, Fan Y, Fang L, Wang C, Lv Z, Fu D, Li Y. Downregulation of MicroRNA-130a contributes to endothelial progenitor cell dysfunction in diabetic patients via its target Runx3. PLoS One. 2013;8(7). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Feng B, Chakrabarti S. miR-320 regulates glucose-induced gene expression in diabetes. ISRN Endocrinol. 2012;2012:549875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Wang XH, Qian RZ, Zhang W, Chen SF, Jin HM, Hu RM. MicroRNA-320 expression in myocardial microvascular endothelial cells and its relationship with insulin-like growth factor-1 in type 2 diabetic rats. Clin Exp Pharmacol Physiol. 2009;36(2):181–188. [DOI] [PubMed] [Google Scholar]
  • 175.Zitman-Gal T, Green J, Pasmanik-Chor M, Golan E, Bernheim J, Benchetrit S. Vitamin D manipulates miR-181c, miR-20b and miR-15a in human umbilical vein endothelial cells exposed to a diabetic-like environment. Cardiovasc Diabetol. 2014;13:8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Cowan C, Muraleedharan CK, O’Donnell JJ III, Singh PK, Lum H, Kumar A, Xu S. MicroRNA-146 inhibits thrombin-induced NF-κB activation and subsequent inflammatory responses in human retinal endothelial cells. Invest Ophthalmol Vis Sci. 2014;55(8):4944–4951. [DOI] [PubMed] [Google Scholar]
  • 177.Dong R, Yang GD, Luo NA, Qu YQ. HuR: a promising therapeutic target for angiogenesis. Gland Surg. 2014;3(3):203–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Cheng HS, Sivachandran N, Lau A, Boudreau E, Zhao JL, Baltimore D, Delgado-Olguin P, Cybulsky MI, Fish JE. MicroRNA-146 represses endothelial activation by inhibiting pro-inflammatory pathways. EMBO Mol Med. 2013;5(7):1017–1034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Wang Q, Bozack SN, Yan Y, Boulton ME, Grant MB, Busik JV. Regulation of retinal inflammation by rhythmic expression of MiR-146a in diabetic retina. Invest Ophthalmol Vis Sci. 2014;55(6):3986–3994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Mortuza R, Feng B, Chakrabarti S. miR-195 regulates SIRT1-mediated changes in diabetic retinopathy. Diabetologia. 2014;57(5):1037–1046. [DOI] [PubMed] [Google Scholar]
  • 181.Dronavalli S, Duka I, Bakris GL. The pathogenesis of diabetic nephropathy. Nat Clin Pract Endocrinol Metab. 2008;4(8):444–452. [DOI] [PubMed] [Google Scholar]
  • 182.Wu H, Kong L, Zhou S, Cui W, Xu F, Luo M, Li X, Tan Y, Miao L. The role of microRNAs in diabetic nephropathy. J Diabetes Res. 2014;2014:920134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Thrailkill KM, Clay Bunn R, Fowlkes JL. Matrix metalloproteinases: their potential role in the pathogenesis of diabetic nephropathy. Endocrine. 2009;35(1):1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Halper J, Kjaer M, Halper J, ed. Progress in Heritable Soft Connective Tissue Diseases. Springer Netherlands; 2014:31–47. [Google Scholar]
  • 185.Pankov R, Yamada KM. Fibronectin at a glance. J Cell Sci. 2002;115(20):3861–3863. [DOI] [PubMed] [Google Scholar]
  • 186.Long J, Wang Y, Wang W, Chang BH, Danesh FR. MicroRNA-29c is a signature microRNA under high glucose conditions that targets Sprouty homolog 1, and its in vivo knockdown prevents progression of diabetic nephropathy. J Biol Chem. 2011;286(13):11837–11848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Badal SS, Wang Y, Long J, Corcoran DL, Chang BH, Truong LD, Kanwar YS, Overbeek PA, Danesh FR. miR-93 regulates Msk2-mediated chromatin remodelling in diabetic nephropathy. Nat Commun. 2016;7:12076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Long J, Wang Y, Wang W, Chang BH, Danesh FR. Identification of microRNA-93 as a novel regulator of vascular endothelial growth factor in hyperglycemic conditions. J Biol Chem. 2010;285(30):23457–23465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Cachia MJ, Peakman M, Zanone M, Watkins PJ, Vergani D. Reproducibility and persistence of neural and adrenal autoantibodies in diabetic autonomic neuropathy. Diabet Med. 1997;14(6):461–465. [DOI] [PubMed] [Google Scholar]
  • 190.Callaghan BC, Cheng HT, Stables CL, Smith AL, Feldman EL. Diabetic neuropathy: clinical manifestations and current treatments. Lancet Neurol. 2012;11(6):521–534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Maser RE, Steenkiste AR, Dorman JS, Nielsen VK, Bass EB, Manjoo Q, Drash AL, Becker DJ, Kuller LH, Greene DA. Epidemiological correlates of diabetic neuropathy. Report from Pittsburgh Epidemiology of Diabetes Complications Study. Diabetes. 1989;38(11):1456–1461. [DOI] [PubMed] [Google Scholar]
  • 192.Partanen J, Niskanen L, Lehtinen J, Mervaala E, Siitonen O, Uusitupa M. Natural history of peripheral neuropathy in patients with non-insulin-dependent diabetes mellitus. N Engl J Med. 1995;333(2):89–94. [DOI] [PubMed] [Google Scholar]
  • 193.Novella SP, Inzucchi SE, Goldstein JM. The frequency of undiagnosed diabetes and impaired glucose tolerance in patients with idiopathic sensory neuropathy. Muscle Nerve. 2001;24(9):1229–1231. [DOI] [PubMed] [Google Scholar]
  • 194.Singleton JR, Smith AG, Bromberg MB. Increased prevalence of impaired glucose tolerance in patients with painful sensory neuropathy. Diabetes Care. 2001;24(8):1448–1453. [DOI] [PubMed] [Google Scholar]
  • 195.Gooch C, Podwall D. The diabetic neuropathies. Neurologist. 2004;10(6):311–322. [DOI] [PubMed] [Google Scholar]
  • 196.Gong Q, Lu Z, Huang Q, Ruan L, Chen J, Liang Y, Wang H, Yue Y, Feng S. Altered microRNAs expression profiling in mice with diabetic neuropathic pain. Biochem Biophys Res Commun. 2015;456(2):615–620. [DOI] [PubMed] [Google Scholar]
  • 197.Ciccacci C, Morganti R, Di Fusco D, D’Amato C, Cacciotti L, Greco C, Rufini S, Novelli G, Sangiuolo F, Marfia GA, Borgiani P, Spallone V. Common polymorphisms in MIR146a, MIR128a and MIR27a genes contribute to neuropathy susceptibility in type 2 diabetes. Acta Diabetol. 2014;51(4):663–671. [DOI] [PubMed] [Google Scholar]
  • 198.Wang L, Chopp M, Szalad A, Zhang Y, Wang X, Zhang R, Liu X, Jia L, Zhang ZG. The role of miR-146a in dorsal root ganglia neurons of experimental diabetic peripheral neuropathy. Neuroscience. 2014;259:155–163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Taganov KD, Boldin MP, Chang KJ, Baltimore D. NF-kappaB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. Proc Natl Acad Sci USA. 2006;103(33):12481–12486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Hou J, Wang P, Lin L, Liu X, Ma F, An H, Wang Z, Cao X. MicroRNA-146a feedback inhibits RIG-I-dependent Type I IFN production in macrophages by targeting TRAF6, IRAK1, and IRAK2. J Immunol. 2009;183(3):2150–2158. [DOI] [PubMed] [Google Scholar]
  • 201.Patapoutian A. Making the pain connection. Neuron. 2001;31(1):4–6. [DOI] [PubMed] [Google Scholar]
  • 202.Patapoutian A, Peier AM, Story GM, Viswanath V. ThermoTRP channels and beyond: mechanisms of temperature sensation. Nat Rev Neurosci. 2003;4(7):529–539. [DOI] [PubMed] [Google Scholar]
  • 203.Liu Y, Ma Q. Generation of somatic sensory neuron diversity and implications on sensory coding. Curr Opin Neurobiol. 2011;21(1):52–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Pickup JC. Inflammation and activated innate immunity in the pathogenesis of type 2 diabetes. Diabetes Care. 2004;27(3):813–823. [DOI] [PubMed] [Google Scholar]
  • 205.Kishi M, Tanabe J, Schmelzer JD, Low PA. Morphometry of dorsal root ganglion in chronic experimental diabetic neuropathy. Diabetes. 2002;51(3):819–824. [DOI] [PubMed] [Google Scholar]
  • 206.Schmeichel AM, Schmelzer JD, Low PA. Oxidative injury and apoptosis of dorsal root ganglion neurons in chronic experimental diabetic neuropathy. Diabetes. 2003;52(1):165–171. [DOI] [PubMed] [Google Scholar]
  • 207.Icli B, Feinberg MW. Plasticity of Arterial and Venous Endothelial Cell Identity: Some Nerve! Circ Res. 2016;119(5):574–576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Shilo S, Roy S, Khanna S, Sen CK. MicroRNA in cutaneous wound healing: a new paradigm. DNA Cell Biol. 2007;26(4):227–237. [DOI] [PubMed] [Google Scholar]
  • 209.Icli B, Wara AK, Moslehi J, Sun X, Plovie E, Cahill M, Marchini JF, Schissler A, Padera RF, Shi J, Cheng HW, Raghuram S, Arany Z, Liao R, Croce K, MacRae C, Feinberg MW. MicroRNA-26a regulates pathological and physiological angiogenesis by targeting BMP/SMAD1 signaling. Circ Res. 2013;113(11):1231–1241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.van Solingen C, Araldi E, Chamorro-Jorganes A, Fernández-Hernando C, Suárez Y. Improved repair of dermal wounds in mice lacking microRNA-155. J Cell Mol Med. 2014;18(6):1104–1112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Jin Y, Tymen SD, Chen D, Fang ZJ, Zhao Y, Dragas D, Dai Y, Marucha PT, Zhou X. MicroRNA-99 family targets AKT/mTOR signaling pathway in dermal wound healing. PLoS One. 2013;8(5):e64434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Chan YC, Roy S, Khanna S, Sen CK. Downregulation of endothelial microRNA-200b supports cutaneous wound angiogenesis by desilencing GATA binding protein 2 and vascular endothelial growth factor receptor 2. Arterioscler Thromb Vasc Biol. 2012;32(6):1372–1382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Winkler MA, Dib C, Ljubimov AV, Saghizadeh M. Targeting miR-146a to treat delayed wound healing in human diabetic organ-cultured corneas. PLoS One. 2014;9(12):e114692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Wang JM, Tao J, Chen DD, Cai JJ, Irani K, Wang Q, Yuan H, Chen AF. MicroRNA miR-27b rescues bone marrow-derived angiogenic cell function and accelerates wound healing in type 2 diabetes mellitus. Arterioscler Thromb Vasc Biol. 2014;34(1):99–109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Dangwal S, Stratmann B, Bang C, Lorenzen JM, Kumarswamy R, Fiedler J, Falk CS, Scholz CJ, Thum T, Tschoepe D. Impairment of wound healing in patients with type 2 diabetes mellitus influences circulating microRNA patterns via inflammatory cytokines. Arterioscler Thromb Vasc Biol. 2015;35(6):1480–1488. [DOI] [PubMed] [Google Scholar]
  • 216.Bonauer A, Carmona G, Iwasaki M, Mione M, Koyanagi M, Fischer A, Burchfield J, Fox H, Doebele C, Ohtani K, Chavakis E, Potente M, Tjwa M, Urbich C, Zeiher AM, Dimmeler S. MicroRNA-92a controls angiogenesis and functional recovery of ischemic tissues in mice. Science. 2009;324(5935):1710–1713. [DOI] [PubMed] [Google Scholar]
  • 217.Fish JE, Santoro MM, Morton SU, Yu S, Yeh RF, Wythe JD, Ivey KN, Bruneau BG, Stainier DY, Srivastava D. miR-126 regulates angiogenic signaling and vascular integrity. Dev Cell. 2008;15(2):272–284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Zhou Q, Gallagher R, Ufret-Vincenty R, Li X, Olson EN, Wang S. Regulation of angiogenesis and choroidal neovascularization by members of microRNA-23~27~24 clusters. Proc Natl Acad Sci USA. 2011;108(20):8287–8292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Urbich C, Kaluza D, Frömel T, Knau A, Bennewitz K, Boon RA, Bonauer A, Doebele C, Boeckel JN, Hergenreider E, Zeiher AM, Kroll J, Fleming I, Dimmeler S. MicroRNA-27a/b controls endothelial cell repulsion and angiogenesis by targeting semaphorin 6A. Blood. 2012;119(6):1607–1616. [DOI] [PubMed] [Google Scholar]
  • 220.Bang C, Fiedler J, Thum T. Cardiovascular importance of the microRNA-23/27/24 family. Microcirculation. 2012;19(3):208–214. [DOI] [PubMed] [Google Scholar]
  • 221.Fiedler J, Jazbutyte V, Kirchmaier BC, Gupta SK, Lorenzen J, Hartmann D, Galuppo P, Kneitz S, Pena JT, Sohn-Lee C, Loyer X, Soutschek J, Brand T, Tuschl T, Heineke J, Martin U, Schulte-Merker S, Ertl G, Engelhardt S, Bauersachs J, Thum T. MicroRNA-24 regulates vascularity after myocardial infarction. Circulation. 2011;124(6):720–730. [DOI] [PubMed] [Google Scholar]
  • 222.Yan XC, Cao J, Liang L, Wang L, Gao F, Yang ZY, Duan JL, Chang TF, Deng SM, Liu Y, Dou GR, Zhang J, Zheng QJ, Zhang P, Han H. miR-342-5p Is a Notch Downstream Molecule and Regulates Multiple Angiogenic Pathways Including Notch, Vascular Endothelial Growth Factor and Transforming Growth Factor β Signaling. J Am Heart Assoc. 2016;5(2):e003042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Campochiaro PA, Aiello LP, Rosenfeld PJ. Anti-vascular endothelial growth factor agents in the treatment of retinal disease: from bench to bedside. Ophthalmology. 2016;123(10S):S78–S88. [DOI] [PubMed] [Google Scholar]
  • 224.Zampetaki A, Willeit P, Burr S, Yin X, Langley SR, Kiechl S, Klein R, Rossing P, Chaturvedi N, Mayr M. Angiogenic microRNAs linked to incidence and progression of diabetic retinopathy in type 1 diabetes. Diabetes. 2016;65(1):216–227. [DOI] [PubMed] [Google Scholar]
  • 225.Zheng D, Ma J, Yu Y, Li M, Ni R, Wang G, Chen R, Li J, Fan GC, Lacefield JC, Peng T. Silencing of miR-195 reduces diabetic cardiomyopathy in C57BL/6 mice. Diabetologia. 2015;58(8):1949–1958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Murthy VL, Naya M, Foster CR, Gaber M, Hainer J, Klein J, Dorbala S, Blankstein R, Di Carli MF. Association between coronary vascular dysfunction and cardiac mortality in patients with and without diabetes mellitus. Circulation. 2012;126(15):1858–1868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Garg R, Rao AD, Baimas-George M, Hurwitz S, Foster C, Shah RV, Jerosch-Herold M, Kwong RY, Di Carli MF, Adler GK. Mineralocorticoid receptor blockade improves coronary microvascular function in individuals with type 2 diabetes. Diabetes. 2015;64(1):236–242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Ceriello A. Hypothesis: the “metabolic memory”, the new challenge of diabetes. Diabetes Res Clin Pract. 2009;86(Suppl 1):S2–S6. [DOI] [PubMed] [Google Scholar]
  • 229.Ceriello A, Ihnat MA, Thorpe JE. Clinical review 2: The “metabolic memory”: is more than just tight glucose control necessary to prevent diabetic complications? J Clin Endocrinol Metab. 2009;94(2):410–415. [DOI] [PubMed] [Google Scholar]
  • 230.Holman RR, Paul SK, Bethel MA, Matthews DR, Neil HA. 10-year follow-up of intensive glucose control in type 2 diabetes. N Engl J Med. 2008;359(15):1577–1589. [DOI] [PubMed] [Google Scholar]
  • 231.Nathan DM, Cleary PA, Backlund JY, Genuth SM, Lachin JM, Orchard TJ, Raskin P, Zinman B; Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and Complications (DCCT/EDIC) Study Research Group . Intensive diabetes treatment and cardiovascular disease in patients with type 1 diabetes. N Engl J Med. 2005;353(25):2643–2653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Costantino S, Paneni F, Lüscher TF, Cosentino F. MicroRNA profiling unveils hyperglycaemic memory in the diabetic heart. Eur Heart J. 2016;37(6):572–576. [DOI] [PubMed] [Google Scholar]
  • 233.Zhong X, Liao Y, Chen L, Liu G, Feng Y, Zeng T, Zhang J. The microRNAs in the pathogenesis of metabolic memory. Endocrinology. 2015;156(9):3157–3168. [DOI] [PubMed] [Google Scholar]
  • 234.Prattichizzo F, Giuliani A, De Nigris V, Pujadas G, Ceka A, La Sala L, Genovese S, Testa R, Procopio AD, Olivieri F, Ceriello A. Extracellular microRNAs and endothelial hyperglycaemic memory: a therapeutic opportunity? Diabetes Obes Metab. 2016;18(9):855–867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Cai S, Cheng X, Pan X, Li J. Emerging role of exosomes in liver physiology and pathology. Hepatol Res. 2016;47(2):194–203. [DOI] [PubMed] [Google Scholar]
  • 236.Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol. 2013;200(4):373–383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Kahlert C, Kalluri R. Exosomes in tumor microenvironment influence cancer progression and metastasis. J Mol Med (Berl). 2013;91(4):431–437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Hu G, Drescher KM, Chen XM. Exosomal miRNAs: biological properties and therapeutic potential. Front Genet. 2012;3:56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Masyuk AI, Huang BQ, Ward CJ, Gradilone SA, Banales JM, Masyuk TV, Radtke B, Splinter PL, LaRusso NF. Biliary exosomes influence cholangiocyte regulatory mechanisms and proliferation through interaction with primary cilia. Am J Physiol Gastrointest Liver Physiol. 2010;299(4):G990–G999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Aliotta JM, Pereira M, Johnson KW, de Paz N, Dooner MS, Puente N, Ayala C, Brilliant K, Berz D, Lee D, Ramratnam B, McMillan PN, Hixson DC, Josic D, Quesenberry PJ. Microvesicle entry into marrow cells mediates tissue-specific changes in mRNA by direct delivery of mRNA and induction of transcription. Exp Hematol. 2010;38(3):233–245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Luo S-S, Ishibashi O, Ishikawa G, Ishikawa T, Katayama A, Mishima T, Takizawa T, Shigihara T, Goto T, Izumi A, Ohkuchi A, Matsubara S, Takeshita T, Takizawa T. Human villous trophoblasts express and secrete placenta-specific microRNAs into maternal circulation via exosomes. Biol Reprod. 2009;81(4):717–729. [DOI] [PubMed] [Google Scholar]
  • 242.Ghai V, Wang K. Recent progress toward the use of circulating microRNAs as clinical biomarkers. Arch Toxicol. 2016;90(12):2959–2978. [DOI] [PubMed] [Google Scholar]
  • 243.Gallo A, Tandon M, Alevizos I, Illei GG. The majority of microRNAs detectable in serum and saliva is concentrated in exosomes. PLoS One. 2012;7(3):e30679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.Kosaka N, Iguchi H, Yoshioka Y, Takeshita F, Matsuki Y, Ochiya T. Secretory mechanisms and intercellular transfer of microRNAs in living cells. J Biol Chem. 2010;285(23):17442–17452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.Mittelbrunn M, Gutiérrez-Vázquez C, Villarroya-Beltri C, González S, Sánchez-Cabo F, González MÁ, Bernad A, Sánchez-Madrid F. Unidirectional transfer of microRNA-loaded exosomes from T cells to antigen-presenting cells. Nat Commun. 2011;2:282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246.Xu G, Chen J, Jing G, Shalev A. Thioredoxin-interacting protein regulates insulin transcription through microRNA-204. Nat Med. 2013;19(9):1141–1146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Xu G, Chen J, Jing G, Grayson TB, Shalev A. miR-204 targets PERK and regulates UPR signaling and β-cell apoptosis. Mol Endocrinol. 2016;30(8):917–924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Dismuke WM, Challa P, Navarro I, Stamer WD, Liu Y. Human aqueous humor exosomes. Exp Eye Res. 2015;132:73–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Vikram A, Kim YR, Kumar S, Li Q, Kassan M, Jacobs JS, Irani K. Vascular microRNA-204 is remotely governed by the microbiome and impairs endothelium-dependent vasorelaxation by downregulating Sirtuin1. Nat Commun. 2016;7:12565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250.Osipova J, Fischer DC, Dangwal S, Volkmann I, Widera C, Schwarz K, Lorenzen JM, Schreiver C, Jacoby U, Heimhalt M, Thum T, Haffner D. Diabetes-associated microRNAs in pediatric patients with type 1 diabetes mellitus: a cross-sectional cohort study. J Clin Endocrinol Metab. 2014;99(9):E1661–E1665. [DOI] [PubMed] [Google Scholar]
  • 251.Nielsen LB, Wang C, Sørensen K, Bang-Berthelsen CH, Hansen L, Andersen MLM, Hougaard P, Juul A, Zhang CY, Pociot F, Mortensen HG. Circulating levels of microRNA from children with newly diagnosed type 1 diabetes and healthy controls: evidence that miR-25 associates to residual beta-cell function and glycaemic control during disease progression. Exp Diabetes Res. 2012;2012:896362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252.Dey N, Das F, Mariappan MM, Mandal CC, Ghosh-Choudhury N, Kasinath BS, Choudhury GG. MicroRNA-21 orchestrates high glucose-induced signals to TOR complex 1, resulting in renal cell pathology in diabetes. J Biol Chem. 2011;286(29):25586–25603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253.Eizirik DL, Colli ML, Ortis F. The role of inflammation in insulitis and beta-cell loss in type 1 diabetes. Nat Rev Endocrinol. 2009;5(4):219–226. [DOI] [PubMed] [Google Scholar]
  • 254.McDaniel ML, Kwon G, Hill JR, Marshall CA, Corbett JA. Cytokines and nitric oxide in islet inflammation and diabetes. Proc Soc Exp Biol Med. 1996;211(1):24–32. [DOI] [PubMed] [Google Scholar]
  • 255.Zheng Y, Wang Z, Tu Y, Shen H, Dai Z, Lin J, Zhou Z. miR-101a and miR-30b contribute to inflammatory cytokine-mediated β-cell dysfunction. LaboratoryInvestigation. 2015;95(12)1387–1397. [DOI] [PubMed]
  • 256.Zampetaki A, Kiechl S, Drozdov I, Willeit P, Mayr U, Prokopi M, Mayr A, Weger S, Oberhollenzer F, Bonora E, Shah A, Willeit J, Mayr M. Plasma microRNA profiling reveals loss of endothelial miR-126 and other microRNAs in type 2 diabetes. Circ Res. 2010;107(6):810–817. [DOI] [PubMed] [Google Scholar]
  • 257.Ortega FJ, Mercader JM, Moreno-Navarrete JM, Rovira O, Guerra E, Esteve E, Xifra G, Martínez C, Ricart W, Rieusset J, Rome S, Karczewska-Kupczewska M, Straczkowski M, Fernández-Real JM. Profiling of circulating microRNAs reveals common microRNAs linked to type 2 diabetes that change with insulin sensitization. Diabetes Care. 2014;37(5):1375–1383. [DOI] [PubMed] [Google Scholar]
  • 258.Liu Y, Gao G, Yang C, Zhou K, Shen B, Liang H, Jiang X. The role of circulating microRNA-126 (miR-126): a novel biomarker for screening prediabetes and newly diagnosed type 2 diabetes mellitus. Int J Mol Sci. 2014;15(6):10567–10577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259.Wang S, Aurora AB, Johnson BA, Qi X, McAnally J, Hill JA, Richardson JA, Bassel-Duby R, Olson EN. The endothelial-specific microRNA miR-126 governs vascular integrity and angiogenesis. Dev Cell. 2008;15(2):261–271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 260.Mocharla P, Briand S, Giannotti G, Dörries C, Jakob P, Paneni F, Lüscher T, Landmesser U. AngiomiR-126 expression and secretion from circulating CD34(+) and CD14(+) PBMCs: role for proangiogenic effects and alterations in type 2 diabetics. Blood. 2013;121(1):226–236. [DOI] [PubMed] [Google Scholar]
  • 261.Al-Kafaji G, Al-Mahroos G, Al-Muhtaresh HA, Skrypnyk C, Sabry MA, Ramadan AR. Decreased expression of circulating microRNA-126 in patients with type 2 diabetic nephropathy: A potential blood-based biomarker. Exp Ther Med. 2016;12(2):815–822. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262.Lv C, Zhou YH, Wu C, Shao Y, Lu CL, Wang QY. The changes in miR-130b levels in human serum and the correlation with the severity of diabetic nephropathy. Diabetes Metab Res Rev. 2015;31(7):717–724. [DOI] [PubMed] [Google Scholar]
  • 263.Bai X, Geng J, Zhou Z, Tian J, Li X. MicroRNA-130b improves renal tubulointerstitial fibrosis via repression of Snail-induced epithelial-mesenchymal transition in diabetic nephropathy. Sci Rep. 2016;6:20475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 264.Pan S, Yang X, Jia Y, Li R, Zhao R. Microvesicle-shuttled miR-130b reduces fat deposition in recipient primary cultured porcine adipocytes by inhibiting PPAR-g expression. J Cell Physiol. 2014;229(5):631–639. [DOI] [PubMed] [Google Scholar]
  • 265.Ai J, Zhang R, Li Y, Pu J, Lu Y, Jiao J, Li K, Yu B, Li Z, Wang R, Wang L, Li Q, Wang N, Shan H, Li Z, Yang B. Circulating microRNA-1 as a potential novel biomarker for acute myocardial infarction. Biochem Biophys Res Commun. 2010;391(1):73–77. [DOI] [PubMed] [Google Scholar]
  • 266.Li Y, Xu X, Liang Y, Liu S, Xiao H, Li F, Cheng H, Fu Z. miR-375 enhances palmitate-induced lipoapoptosis in insulin-secreting NIT-1 cells by repressing myotrophin (V1) protein expression. Int J Clin Exp Pathol. 2010;3(3):254–264. [PMC free article] [PubMed] [Google Scholar]
  • 267.Higuchi C, Nakatsuka A, Eguchi J, Teshigawara S, Kanzaki M, Katayama A, Yamaguchi S, Takahashi N, Murakami K, Ogawa D, Sasaki S, Makino H, Wada J. Identification of circulating miR-101, miR-375 and miR-802 as biomarkers for type 2 diabetes. Metabolism. 2015;64(4):489–497. [DOI] [PubMed] [Google Scholar]
  • 268.Erener S, Mojibian M, Fox JK, Denroche HC, Kieffer TJ. Circulating miR-375 as a biomarker of β-cell death and diabetes in mice. Endocrinology. 2013;154(2):603–608. [DOI] [PubMed] [Google Scholar]
  • 269.Raal FJ, Santos RD, Blom DJ, Marais AD, Charng MJ, Cromwell WC, Lachmann RH, Gaudet D, Tan JL, Chasan-Taber S, Tribble DL, Flaim JD, Crooke ST. Mipomersen, an apolipoprotein B synthesis inhibitor, for lowering of LDL cholesterol concentrations in patients with homozygous familial hypercholesterolaemia: a randomised, double-blind, placebo-controlled trial. Lancet. 2010;375(9719):998–1006. [DOI] [PubMed] [Google Scholar]
  • 270.Broderick JA, Zamore PD. MicroRNA therapeutics. Gene Ther. 2011;18(12):1104–1110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271.Feinberg MW, Moore KJ. MicroRNA regulation of atherosclerosis. Circ Res. 2016;118(4):703–720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272.Andreou I, Sun X, Stone PH, Edelman ER, Feinberg MW. miRNAs in atherosclerotic plaque initiation, progression, and rupture. Trends Mol Med. 2015;21(5):307–318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273.Grünweller A, Hartmann RK. Locked nucleic acid oligonucleotides: the next generation of antisense agents? BioDrugs. 2007;21(4):235–243. [DOI] [PubMed] [Google Scholar]
  • 274.Flierl U, Nero TL, Lim B, Arthur JF, Yao Y, Jung SM, Gitz E, Pollitt AY, Zaldivia MT, Jandrot-Perrus M, Schäfer A, Nieswandt B, Andrews RK, Parker MW, Gardiner EE, Peter K. Phosphorothioate backbone modifications of nucleotide-based drugs are potent platelet activators. J Exp Med. 2015;212(2):129–137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 275.Stenvang J, Petri A, Lindow M, Obad S, Kauppinen S. Inhibition of microRNA function by antimiR oligonucleotides. Silence. 2012;3(1):1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 276.van Rooij E, Purcell AL, Levin AA. Developing microRNA therapeutics. Circ Res. 2012;110(3):496–507. [DOI] [PubMed] [Google Scholar]
  • 277.Rayner KJ, Esau CC, Hussain FN, McDaniel AL, Marshall SM, van Gils JM, Ray TD, Sheedy FJ, Goedeke L, Liu X, Khatsenko OG, Kaimal V, Lees CJ, Fernandez-Hernando C, Fisher EA, Temel RE, Moore KJ. Inhibition of miR-33a/b in non-human primates raises plasma HDL and lowers VLDL triglycerides. Nature. 2011;478(7369):404–407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278.Semple SC, Akinc A, Chen J, Sandhu AP, Mui BL, Cho CK, Sah DW, Stebbing D, Crosley EJ, Yaworski E, Hafez IM, Dorkin JR, Qin J, Lam K, Rajeev KG, Wong KF, Jeffs LB, Nechev L, Eisenhardt ML, Jayaraman M, Kazem M, Maier MA, Srinivasulu M, Weinstein MJ, Chen Q, Alvarez R, Barros SA, De S, Klimuk SK, Borland T, Kosovrasti V, Cantley WL, Tam YK, Manoharan M, Ciufolini MA, Tracy MA, de Fougerolles A, MacLachlan I, Cullis PR, Madden TD, Hope MJ. Rational design of cationic lipids for siRNA delivery. Nat Biotechnol. 2010;28(2):172–176. [DOI] [PubMed] [Google Scholar]
  • 279.Ebert MS, Neilson JR, Sharp PA. MicroRNA sponges: competitive inhibitors of small RNAs in mammalian cells. Nat Methods. 2007;4(9):721–726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 280.Rayner KJ, Suárez Y, Dávalos A, Parathath S, Fitzgerald ML, Tamehiro N, Fisher EA, Moore KJ, Fernández-Hernando C. miR-33 contributes to the regulation of cholesterol homeostasis. Science. 2010;328(5985):1570–1573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 281.Gentner B, Schira G, Giustacchini A, Amendola M, Brown BD, Ponzoni M, Naldini L. Stable knockdown of microRNA in vivo by lentiviral vectors. Nat Methods. 2009;6(1):63–66. [DOI] [PubMed] [Google Scholar]
  • 282.Yu B, Zhang Y, Zheng W, Fan C, Chen T. Positive surface charge enhances selective cellular uptake and anticancer efficacy of selenium nanoparticles. Inorg Chem. 2012;51(16):8956–8963. [DOI] [PubMed] [Google Scholar]
  • 283.Osaka T, Nakanishi T, Shanmugam S, Takahama S, Zhang H. Effect of surface charge of magnetite nanoparticles on their internalization into breast cancer and umbilical vein endothelial cells. Colloids Surf B Biointerfaces. 2009;71(2):325–330. [DOI] [PubMed] [Google Scholar]
  • 284.Kheirolomoom A, Kim CW, Seo JW, Kumar S, Son DJ, Gagnon MKJ, Ingham ES, Ferrara KW, Jo H. Multifunctional nanoparticles facilitate molecular targeting and miRNA delivery to inhibit atherosclerosis in ApoE(-/-) mice. ACS Nano. 2015;9(9):8885–8897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 285.Ma S, Tian XY, Zhang Y, Mu C, Shen H, Bismuth J, Pownall HJ, Huang Y, Wong WT. E-selectin-targeting delivery of microRNAs by microparticles ameliorates endothelial inflammation and atherosclerosis. Sci Rep. 2016;6:22910. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 286.Yu S, Cao H, Shen B, Feng J. Tumor-derived exosomes in cancer progression and treatment failure. Oncotarget. 2015;6(35):37151–37168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 287.Hoshino A, Costa-Silva B, Shen T-L, Rodrigues G, Hashimoto A, Tesic Mark M, Molina H, Kohsaka S, Di Giannatale A, Ceder S, Singh S, Williams C, Soplop N, Uryu K, Pharmer L, King T, Bojmar L, Davies AE, Ararso Y, Zhang T, Zhang H, Hernandez J, Weiss JM, Dumont-Cole VD, Kramer K, Wexler LH, Narendran A, Schwartz GK, Healey JH, Sandstrom P, Labori KJ, Kure EH, Grandgenett PM, Hollingsworth MA, de Sousa M, Kaur S, Jain M, Mallya K, Batra SK, Jarnagin WR, Brady MS, Fodstad O, Muller V, Pantel K, Minn AJ, Bissell MJ, Garcia BA, Kang Y, Rajasekhar VK, Ghajar CM, Matei I, Peinado H, Bromberg J, Lyden D. Tumour exosome integrins determine organotropic metastasis. Nature. 2015;527(7578):329–335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 288.Jopling CL, Yi M, Lancaster AM, Lemon SM, Sarnow P. Modulation of hepatitis C virus RNA abundance by a liver-specific MicroRNA. Science. 2005;309(5740):1577–1581. [DOI] [PubMed] [Google Scholar]
  • 289.Janssen HL, Reesink HW, Lawitz EJ, Zeuzem S, Rodriguez-Torres M, Patel K, van der Meer AJ, Patick AK, Chen A, Zhou Y, Persson R, King BD, Kauppinen S, Levin AA, Hodges MR. Treatment of HCV infection by targeting microRNA. N Engl J Med. 2013;368(18):1685–1694. [DOI] [PubMed] [Google Scholar]
  • 290.Yang YM, Seo SY, Kim TH, Kim SG. Decrease of microRNA-122 causes hepatic insulin resistance by inducing protein tyrosine phosphatase 1B, which is reversed by licorice flavonoid. Hepatology. 2012;56(6):2209–2220. [DOI] [PubMed] [Google Scholar]
  • 291.Jordan SD, Krüger M, Willmes DM, Redemann N, Wunderlich FT, Brönneke HS, Merkwirth C, Kashkar H, Olkkonen VM, Böttger T, Braun T, Seibler J, Brüning JC. Obesity-induced overexpression of miRNA-143 inhibits insulin-stimulated AKT activation and impairs glucose metabolism. Nat Cell Biol. 2011;13(4):434–446. [DOI] [PubMed] [Google Scholar]
  • 292.Wen F, Yang Y, Jin D, Sun J, Yu X, Yang Z. miRNA-145 is involved in the development of resistin-induced insulin resistance in HepG2 cells. Biochem Biophys Res Commun. 2014;445(2):517–523. [DOI] [PubMed] [Google Scholar]
  • 293.Hung TM, Ho CM, Liu YC, Lee JL, Liao YR, Wu YM, Ho MC, Chen CH, Lai HS, Lee PH. Up-regulation of microRNA-190b plays a role for decreased IGF-1 that induces insulin resistance in human hepatocellular carcinoma. PLoS One. 2014;9(2):e89446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 294.Crépin D, Benomar Y, Riffault L, Amine H, Gertler A, Taouis M. The over-expression of miR-200a in the hypothalamus of ob/ob mice is linked to leptin and insulin signaling impairment. Mol Cell Endocrinol. 2014;384(1-2):1–11. [DOI] [PubMed] [Google Scholar]
  • 295.Fu X, Dong B, Tian Y, Lefebvre P, Meng Z, Wang X, Pattou F, Han W, Wang X, Lou F, Jove R, Staels B, Moore DD, Huang W. MicroRNA-26a regulates insulin sensitivity and metabolism of glucose and lipids. J Clin Invest. 2015;125(6):2497–2509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 296.de Guia RM, Rose AJ, Sommerfeld A, Seibert O, Strzoda D, Zota A, Feuchter Y, Krones-Herzig A, Sijmonsma T, Kirilov M, Sticht C, Gretz N, Dallinga-Thie G, Diederichs S, Klöting N, Blüher M, Berriel Diaz M, Herzig S. microRNA-379 couples glucocorticoid hormones to dysfunctional lipid homeostasis. EMBO J. 2015;34(3):344–360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 297.Kornfeld JW, Baitzel C, Könner AC, Nicholls HT, Vogt MC, Herrmanns K, Scheja L, Haumaitre C, Wolf AM, Knippschild U, Seibler J, Cereghini S, Heeren J, Stoffel M, Brüning JC. Obesity-induced overexpression of miR-802 impairs glucose metabolism through silencing of Hnf1b. Nature. 2013;494(7435):111–115. [DOI] [PubMed] [Google Scholar]
  • 298.Shi Z, Zhao C, Guo X, Ding H, Cui Y, Shen R, Liu J. Differential expression of microRNAs in omental adipose tissue from gestational diabetes mellitus subjects reveals miR-222 as a regulator of ERα expression in estrogen-induced insulin resistance. Endocrinology. 2014;155(5):1982–1990. [DOI] [PubMed] [Google Scholar]
  • 299.Xu G, Ji C, Song G, Zhao C, Shi C, Song L, Chen L, Yang L, Huang F, Pang L, Zhang N, Zhao Y, Guo X. miR-26b modulates insulin sensitivity in adipocytes by interrupting the PTEN/PI3K/AKT pathway. Int J Obes. 2015;39(10):1523–1530. [DOI] [PubMed] [Google Scholar]
  • 300.Chisalita SI, Johansson GS, Liefvendahl E, Bäck K, Arnqvist HJ. Human aortic smooth muscle cells are insulin resistant at the receptor level but sensitive to IGF1 and IGF2. J Mol Endocrinol. 2009;43(6):231–239. [DOI] [PubMed] [Google Scholar]
  • 301.Taniguchi CM, Emanuelli B, Kahn CR. Critical nodes in signalling pathways: insights into insulin action. Nat Rev Mol Cell Biol. 2006;7(2):85–96. [DOI] [PubMed] [Google Scholar]
  • 302.Agarwal P, Srivastava R, Srivastava AK, Ali S, Datta M. miR-135a targets IRS2 and regulates insulin signaling and glucose uptake in the diabetic gastrocnemius skeletal muscle. Biochim Biophys Acta. 2013;1832(8):1294–1303. [DOI] [PubMed] [Google Scholar]
  • 303.Hudson MB, Rahnert JA, Zheng B, Woodworth-Hobbs ME, Franch HA, Russ Price S. miR-182 attenuates atrophy-related gene expression by targeting FoxO3 in skeletal muscle. Am J Physiol Cell Physiol. 2014;307(4):C314–C319. [DOI] [PMC free article] [PubMed]
  • 304.Jia L, Li YF, Wu GF, Song ZY, Lu HZ, Song CC, Zhang QL, Zhu JY, Yang GS, Shi XE. miRNA-199a-3p regulates C2C12 myoblast differentiation through IGF-1/AKT/mTOR signal pathway. Int J Mol Sci. 2013;15(1):296–308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 305.Bai L, Liang R, Yang Y, Hou X, Wang Z, Zhu S, Wang C, Tang Z, Li K. MicroRNA-21 regulates PI3K/Akt/mTOR signaling by targeting TGFβI during Skeletal muscle development in pigs. PLoS One. 2015;10(5):e0119396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 306.Huang B, Qin W, Zhao B, Shi Y, Yao C, Li J, Xiao H, Jin Y. MicroRNA expression profiling in diabetic GK rat model. Acta Biochim Biophys Sin (Shanghai). 2009;41(6):472–477. [DOI] [PubMed] [Google Scholar]
  • 307.Yang WM, Jeong HJ, Park SY, Lee W. Induction of miR-29a by saturated fatty acids impairs insulin signaling and glucose uptake through translational repression of IRS-1 in myocytes. FEBS Lett. 2014;588(13):2170–2176. [DOI] [PubMed] [Google Scholar]
  • 308.Hu YW, Hu YR, Zhao JY, Li SF, Ma X, Wu SG, Lu JB, Qiu YR, Sha YH, Wang YC, Gao JJ, Zheng L, Wang Q. An agomir of miR-144-3p accelerates plaque formation through impairing reverse cholesterol transport and promoting pro-inflammatory cytokine production. PLoS One. 2014;9(4):e94997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 309.Mortuza R, Chen S, Feng B, Sen S, Chakrabarti S. High glucose induced alteration of SIRTs in endothelial cells causes rapid aging in a p300 and FOXO regulated pathway. PLoS One. 2013;8(1):e54514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 310.Ying C, Sui-Xin L, Kang-Ling X, Wen-Liang Z, Lei D, Yuan L, Fan Z, Chen Z. MicroRNA-492 reverses high glucose-induced insulin resistance in HUVEC cells through targeting resistin. Mol Cell Biochem. 2014;391(1-2):117–125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 311.Xu J, Hu G, Lu M, Xiong Y, Li Q, Chang CCY, Song B, Chang T, Li B. miR-9 reduces human acyl-coenzyme A:cholesterol acyltransferase-1 to decrease THP-1 macrophage-derived foam cell formation. Acta Biochim Biophys Sin (Shanghai). 2013;45(11):953–962. [DOI] [PubMed] [Google Scholar]
  • 312.Zhang L, Zhou M, Wang Y, Huang W, Qin G, Weintraub NL, Tang Y. miR-92a inhibits vascular smooth muscle cell apoptosis: role of the MKK4-JNK pathway. Apoptosis. 2014;19(6):975–983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 313.He F, Peng F, Xia X, Zhao C, Luo Q, Guan W, Li Z, Yu X, Huang F. miR-135a promotes renal fibrosis in diabetic nephropathy by regulating TRPC1. Diabetologia. 2014;57(8):1726–1736. [DOI] [PubMed] [Google Scholar]
  • 314.Zhang X, Gong X, Han S, Zhang Y. miR-29b protects dorsal root ganglia neurons from diabetic rat. Cell Biochem Biophys. 2014;70(2):1105–1111. [DOI] [PubMed] [Google Scholar]
  • 315.Zhang L, He S, Guo S, Xie W, Xin R, Yu H, Yang F, Qiu J, Zhang D, Zhou S, Zhang K. Down-regulation of miR-34a alleviates mesangial proliferation in vitro and glomerular hypertrophy in early diabetic nephropathy mice by targeting GAS1. J Diabetes Complications. 2014;28(3):259–264. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Endocrine Reviews are provided here courtesy of The Endocrine Society

RESOURCES