Next Article in Journal
Cellular Crosstalk between Endothelial and Smooth Muscle Cells in Vascular Wall Remodeling
Previous Article in Journal
Study of the Expression and Function of Calcium-Sensing Receptor in Human Skeletal Muscle
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of Long Non-Coding RNAs and the Molecular Mechanisms Involved in Insulin Resistance

1
Laboratorio de Epidemiología Clínica y Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39087, GRO, Mexico
2
Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39087, GRO, Mexico
3
Laboratorio de Citopatología e Histoquímica, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39087, GRO, Mexico
4
CONACyT, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39087, GRO, Mexico
5
Laboratorio de Virología, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39087, GRO, Mexico
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(14), 7256; https://doi.org/10.3390/ijms22147256
Submission received: 3 June 2021 / Revised: 27 June 2021 / Accepted: 2 July 2021 / Published: 6 July 2021
(This article belongs to the Section Molecular Endocrinology and Metabolism)

Abstract

:
Long non-coding RNAs (lncRNAs) are single-stranded RNA biomolecules with a length of >200 nt, and they are currently considered to be master regulators of many pathological processes. Recent publications have shown that lncRNAs play important roles in the pathogenesis and progression of insulin resistance (IR) and glucose homeostasis by regulating inflammatory and lipogenic processes. lncRNAs regulate gene expression by binding to other non-coding RNAs, mRNAs, proteins, and DNA. In recent years, several mechanisms have been reported to explain the key roles of lncRNAs in the development of IR, including metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), imprinted maternal-ly expressed transcript (H19), maternally expressed gene 3 (MEG3), myocardial infarction-associated transcript (MIAT), and steroid receptor RNA activator (SRA), HOX transcript antisense RNA (HOTAIR), and downregulated Expression-Related Hexose/Glucose Transport Enhancer (DREH). LncRNAs participate in the regulation of lipid and carbohydrate metabolism, the inflammatory process, and oxidative stress through different pathways, such as cyclic adenosine monophosphate/protein kinase A (cAMP/PKA), phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT), polypyrimidine tract-binding protein 1/element-binding transcription factor 1c (PTBP1/SREBP-1c), AKT/nitric oxide synthase (eNOS), AKT/forkhead box O1 (FoxO1), and tumor necrosis factor-alpha (TNF-α)/c-Jun-N-terminal kinases (JNK). On the other hand, the mechanisms linked to the molecular, cellular, and biochemical actions of lncRNAs vary according to the tissue, biological species, and the severity of IR. Therefore, it is essential to elucidate the role of lncRNAs in the insulin signaling pathway and glucose and lipid metabolism. This review analyzes the function and molecular mechanisms of lncRNAs involved in the development of IR.

1. Introduction

Insulin resistance (IR) is a clinical condition defined as a reduced metabolic response to the action of insulin on target tissues, which are unable to coordinate a normal glucose-lowering response involving the suppression of endogenous glucose production, the suppression of lipolysis, cellular uptake of available plasma glucose, and glycogen synthesis. IR is a critical component of metabolic syndrome, obesity, and hyperlipidemia and is a risk factor for cardiovascular diseases (CVDs) and type 2 diabetes (T2D), especially when an insulin-resistant individual cannot secrete enough insulin from pancreatic β-cells [1,2]. Several mechanisms are involved in the etiology of IR, including mutations and post-translational modifications of the insulin receptor (IR), the insulin receptor substrate (IRS), and effector molecules located downstream. An essential factor that contributes to IR is the decreased phosphorylation of tyrosine residues (Tyr) in the β-subfraction of IR, which results in a decrease in its interaction with phosphatidylinositol 3-kinase (PI3K) and thus alters the phosphorylation and activation of protein kinase B (AKT) [3].
Several factors, such as proinflammatory cytokines, saturated fatty acids (SFAs), amino acids, endothelin 1, angiotensin II, and hyperinsulinemia, increase kinase activity [2,4]. Cellular senescence and chronic inflammation are known to be involved in the development of IR and T2D through both endocrine and metabolic effects, with the increased release of free fatty acids (FFAs) from adipocytes to the circulation. FFAs induce myocyte inflammation and metabolic dysfunction [5]. The accumulation of intramuscular lipids leads to insufficient oxidation of fatty acids by the mitochondria, which can be a causal factor in the development of IR. High concentrations of plasma FFAs are involved in the etiology of obesity-associated IR. In particular, SFAs such as palmitic acid markedly inhibit insulin-stimulated phosphorylation of IRS-1 and AKT, thereby inhibiting insulin-induced signaling. In addition, SFAs induce the expression of proinflammatory cytokines [6], an alteration that has also been associated with the development of IR.
Genome-wide association studies have identified approximately 88 loci associated with the risk of developing T2D. Most of the loci are related to insulin secretion and pancreatic β-cell function. Among the loci associated with IR are the peroxisome proliferator-activated receptor (PPAR) gamma, Kruppel-like factor 14 (KLF14), insulin receptor substrate 1 (IRS1), glucokinase regulator (GCKR), transcription factor 7-like 2 (TCF7L2), N-acetyltransferase (NAT) 1 and 2, transmembrane protein 163 (TMEM163), insulin-like growth factor (IGF1) 1, melanocortin 4 receptor (MC4R), transcription elongation regulator 1-like (TCERG1L), and sterol-C4-methyl oxidase-like (SC4MOL) [2,7,8]. In contrast, several miRNAs are involved in metabolic processes, including the regulation of insulin synthesis, secretion, and sensitivity. For instance, the overexpression of miR-29 led to IR, whereas miR-320 increased insulin sensitivity, miR-30d inhibited insulin expression, and miR-133 and miR-223 regulated glucose transporter type 4 (GLUT4) expression [9]. Other studies have reported associations between IR and let-7b, miR-144-5p, miR-34a, miR-532-5p [10], miR-27a [11], miR-34a, miR-592, and miR-194 [12].
Recently, the relationship between different long non-coding RNAs (lncRNAs) and the development and progression of metabolic diseases, including metabolic syndrome, liver diseases, obesity, IR, and diabetes, has been reported. However, the functional role of lncRNAs in the molecular mechanisms involved in IR development remains largely unknown. The aim of this review was to analyze the function and molecular mechanisms of the main lncRNAs involved in IR development.

2. Metabolic Processes of Insulin Resistance

2.1. Lipocentric and Glucocentric Approach in IR

Two of the most popular theories about IR development are the lipocentric and glucocentric approaches. In the first approach, IR is a consequence of lipotoxic effects. Lipotoxicity is considered an intracellular over-accumulation of FFAs or their derivatives, such as diacylglycerol (DAG), ceramides, and long-chain fatty acyl-coenzyme A (CoA), in adipose, liver, and muscle tissues [6,13]. This over-accumulation results in the activation of phosphokinase C (PKC). This serine/threonine kinase increases the phosphorylation of serine residues of IRS, which leads to the negative regulation on phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) signaling pathway [14,15].
Additionally, the over-accumulation of lipids in the cytoplasm generates endoplasmic reticulum stress with subsequent activation of the unfolded protein response (UPR). UPR can activate c-Jun-N-terminal kinases (JNKs), serine/threonine kinases that promote the negative regulation of the insulin receptor (INSR) and IRS by phosphorylating serine residues [16]. Therefore, JNK may promote IR via the phosphorylation of IRS molecules. In addition, JNK promotes metabolic inflammation, adiposity, and negative regulation of PPARα and fibroblast growth factor 21 (FGF21). PPARα increases the transcription of genes related to the oxidative metabolism of fatty acids and the target gene FGF21, resulting in an increase in the hormone FGF21; this hormone promotes insulin sensitivity in the liver. For this reason, the inhibition of the PPARα–FGF21 axis by JNK is a positive feedback cycle in the development of IR [17]. Moreover, a high intake of SFAs has been reported to induce oxidation, inflammation, and impairment of hepatic insulin signaling in the liver tissue [18]. The molecular mechanism of SFAs contributes to the negative regulation of insulin signaling and activates proinflammatory pathways; SFAs activate toll-like receptor 4 (TLR4), which is expressed on immune cells, in white adipose tissue, and in the liver, to stimulate downstream proinflammatory processes [13].
On the other hand, in the glucocentric approach, IR development is linked to recurrent states of hyperglycemia that generate advanced glycation end products (AGEs), which exert pathogenic effects through interaction with cell surface receptors [19]. AGEs are involved in IR through direct and indirect actions of several pathways, including the activation of protein kinase C (PKC)-α, changes in the PI3K and mitogen-activated protein kinase (MAPK) insulin signaling pathways with opposing protective effects by sirtuin 1 (SIRT1), and the induction of oxidative stress and inflammation [20,21].

2.2. Causal Link between Oxidative Stress, Inflammation, and Insulin Resistance

Hyperglycemia contributes to increased reactive oxygen species (ROS) production by activating the PKC pathway, increasing hexosamine pathway flux, and promoting AGE production. Oxidative stress leads to altered glucose uptake in skeletal muscle and adipose tissue and decreases insulin secretion from β-cells, which can cause IR. ROS accumulation leads to non-specific oxidative damage to DNA, proteins, and lipids and consequently to cell death. Oxidative stress activates several signaling pathways, such as inhibitor of nuclear factor kappa/nuclear factor kappa B (IKKB/NF-κB) and JNK [22,23,24]. Additionally, hyperglycemia leads to the upregulation of proinflammatory factors, which induces endothelial dysfunction, leukocyte adhesion, and IR. Most cytokines and inflammatory mediators, such as tumor necrosis factor-alpha (TNF-α), monocyte chemoattractant protein-1 (MCP-1), C-reactive protein, and interleukins (IL), are upregulated in IR or high-glucose conditions [23,25]. TNF-α itself impairs insulin signaling via serine phosphorylation of IRS-1, leading to its degradation, and the translocation of GLUT4 is reduced.
Activation of IKKB/NF-κB and JNK pathways is a possible link between inflammation and IR [17,23]. The events that cause FFA-induced hepatic IR have been observed to occur via activation of PKC-δ and nicotinamide adenine dinucleotide phosphate oxidase (NADPH). These actions induce oxidative stress, leading to IKKδ- and JNK-mediated serine phosphorylation of IRS and the subsequent impairment of hepatic insulin signaling [6,26]. As previously described, the processes involved in the development of IR lead to the production of ROS, AGEs, and proinflammatory factors, which, in turn, will lead to other metabolic alterations, such as endothelial dysfunction and IR, and to the increased production of these molecules. Therefore, an unstoppable vicious cycle occurs if the necessary preventive measures are not implemented to reduce these alterations, which, of course, will lead to the generation of comorbidities associated with IR.

3. lncRNAs in Insulin Resistance

lncRNAs have been described as having an essential role in the molecular mechanisms involved in IR. They are single-stranded nucleotide sequences, ranging from approximately 200 bp to 10 kb in length, and are the most abundant non-coding RNAs in the human genome compared to microRNAs [27,28]. lncRNAs are usually polyadenylated and can have a multi-exonic structure. However, most of them are not translated into proteins, and it has been recently reported that some lncRNAs can encode small peptides. The majority of lncRNAs are transcribed by RNA polymerase II and often exhibit alternative splicing [29,30]. lncRNAs are classified by their genomic locations into (1) long intergenic ncRNAs (lincRNAs), which are located between protein-coding genes, (2) intronic lncRNAs, (3) sense lncRNAs, which are found on the sense strand of protein-coding genes and may contain exons from these genes, (4) antisense lncRNAs, which are located on the antisense strand, and (5) enhancer RNAs (eRNAs), which are produced from the bidirectional transcription of enhancer regions [29].
The subcellular localization of lncRNAs, either in the nucleus or in the cytoplasm, provides an indication of its function. Many lncRNAs are involved in gene regulatory processes at the nuclear level, including the repression or activation of transcription, activation or repression of multiple loci through chromatin remodeling, and epigenetic gene regulatory processes [31]. Additionally, lncRNAs can regulate gene expression post-transcriptionally by modulating the translation or stability of partially complementary mRNAs and interfering with RNA-binding proteins to influence splicing and translation. Competing endogenous RNAs (ceRNA) and circular RNAs are stable lncRNAs that accumulate in the cell and modulate gene expression by acting as decoys or sponges for microRNAs. Finally, some lncRNAs function post-translationally to control protein turnover by facilitating ubiquitination [32]. However, the specific characterization of lncRNA subcellular localization in IR models has focused on the endogenous competitor function and its localization in the cytoplasm, whereas the characterization of lncRNAs at the nuclear level is lacking.
At present, the GENCODE database (v.37) identifies 17,957 genes as lncRNAs [33]. Several lncRNAs have been confirmed to be related to IR, such as metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), imprinted maternally expressed transcript (H19), maternally expressed gene 3 (MEG3), myocardial infarction-associated transcript (MIAT), and steroid receptor RNA activator (SRA). However, there is still much to learn about the function, molecular processes involved, and expression of these non-coding RNAs in IR.

3.1. Metastasis-Associated Lung Adenocarcinoma Transcript 1 (MALAT1)

MALAT1 is a widely studied lncRNA; it is also known as nuclear-enriched abundant transcript 2 (NEAT2), HCN, LINC00047, NCRN00047, and PRO2853. In cancer, it has been associated with the transcriptional and post-transcriptional regulation of numerous genes involved in the regulation of the cell cycle, metastasis, cell migration, and resistance to antineoplastic drugs and with the prognosis and development of cancer [34]. Moreover, it has been reported that MALAT1 is related to the etiology and progression of metabolic diseases, such as T2D [35], metabolic syndrome [36], diabetic retinopathy [37], CVD [38], diabetic nephropathy [39], and gestational diabetes [40].
The MALAT1 gene is located on chromosome 11q13.1 and has an approximate length of 6.7 kb, and the 3’ region lacks a poly-A tail. The 3’-end of MALAT1 forms a unique triple-helix structure that protects it from 3’-5’ exonucleases. MALAT1 is highly expressed to a similar or even higher degree than some proteins, such as β-actin and D-glyceraldehyde-3-phosphate dehydrogenase. MALAT1 is predominantly located in the nucleus; however, a small proportion of this molecule can be found in the cytoplasm [34,39,41,42].
The function of MALAT1 occurs inside cells, primarily in the nucleus. In a hyperglycemic state, the global inhibition of DNA methyltransferase (DNMT) activity induces significant dysregulation in the expression of MALAT1, probably due to the hypomethylation of CpG islands in the MALAT1 promoter [43]. Additionally, MALAT1 downregulation may contribute to the dysfunction of pancreatic β-cells and the development of T2D via direct interaction and modulation of polypyrimidine tract-binding protein 1 (PTBP1), demonstrating the scaffolding activity of MALAT1. Moreover, hyperglycemia and elevated levels of FFAs also induce an increase in the transcriptional factor pancreatic and duodenal homeobox 1 (PDX1), which, in turn, increases the transcription of MALAT1 and can induce β-cell dysfunction through the PDX1/MALAT1/PTBP1 pathway [44]. These events increase the transcription of MALAT1 and its levels in the nucleus. MALAT1 indirectly promotes the overexpression of the proinflammatory genes TNF-α, IL-6, and IL-1β. This event occurs when MALAT1 is bound to enhancer of zeste polycomb homolog 2 (EZH2), which is the catalytic subunit of polycomb repressive complex 2 (PRC2), leading to histone methylation of anti-inflammatory genes, transcriptional repression, and therefore, to increased transcription of proinflammatory genes [43]. As mentioned earlier, the overexpression of proinflammatory genes results in a systemic inflammatory state with the subsequent activation of serine/threonine kinases such as JNK; the activation of JNK results in the negative feedback of the PI3K–AKT pathway activated by insulin through of mechanism above mentioned (Figure 1).
Dong et al. reported that glycated albumin induced MCP-1 expression in retinal microglia and observed that its expression was dependent on the MALAT1/miR-124 axis, which has been previously demonstrated to have an important relationship with inflammatory processes in diabetic mice and cell cultures exposed to high concentrations of glucose [45]. Another proinflammatory mechanism due to hyperglycemia mediated by MALAT1 is the upregulation of serum amyloid antigen 3 (SAA3), which stimulates the production of proinflammatory cytokines TNF-α and IL-6, promoting endothelial inflammation, which may lead to CVD [46]. Furthermore, in hepatocytes under fatty acid overload conditions, MALAT1 directly interacts with sterol regulatory element-binding transcription factor 1c (SREBP-1c), an essential regulator of lipogenesis. This binding inhibits the ubiquitination of SREBP-1c, increasing SREBP-1c activity and the expression of its target lipogenic genes, which induces intracellular lipid accumulation and IR (Figure 1) [47].
As previously described, in a hyperglycemic state, MALAT1 is involved in the increased transcription of proinflammatory and lipogenic genes, leading to the activation of threonine/serine kinases JNK and PKC. These increases are associated with negative regulation in the insulin signaling pathway (Figure 1) [15]. However, more studies are necessary to establish the relation between MALAT1, JNK, and PKC. In addition, rigorous studies are required to evaluate the effects of the hyper-insulinemic state on MALAT1 activity.

3.2. Imprinted Maternally Expressed Transcript (H19)

H19, a 2.3 kb RNA, is highly expressed in the fetal stage and evolutionarily conserved. H19 plays an important role in embryonal development, mainly in mesoderm and endoderm-derived tissues. Its expression is strongly downregulated after birth, except in cardiac and skeletal muscle. This lncRNA is transcribed exclusively from the maternal allele and resides in the H19-IGF2 locus, a highly conserved imprint located on human chromosome 11p15.5, where these two genes are reciprocally imprinted. The H19 gene comprises five exons separated by small introns and produces a spliced, capped, and polyadenylated RNA that mainly resides in the cytoplasm [48,49]. The hypermethylation of the H19 gene promoter and allele-specific methylation of the 3′ portion of H19 may be related to changes in its expression [50].
Different studies have reported that H19 overexpression contributes to IR development; however, the results have been contradictory. An increase in the expression levels of H19 associated with decreased methylation at CpG sites and reduced folate levels has been reported in the liver tissue of patients with T2D [51]. Moreover, the binding of S-adenosylhomocysteine hydrolase (SAHH) to H19 has been shown to interfere with its ability to hydrolyze S-adenosylhomocysteine (SAH), which is a potent inhibitor of S-adenosylmethionine (SAM)-dependent DNMTs. This interaction leads to decreased DNMT3B-mediated methylation of different genomic loci, which could be associated with the development of various pathological conditions [52].
Under normal physiological conditions, hepatic glucose production (HGP) is controlled by glucagon (a positive regulator) and insulin (a negative regulator) in response to fasting or postprandial states, respectively. In the fasting state, glucagon promotes gluconeogenesis and glycogenolysis through the increased expression of phosphoenolpyruvate carboxykinase (PEPCK) and G6Pase. This effect is linked to the interaction of forkhead box O1 (FoxO1) and peroxisome proliferator-activated receptor-gamma coactivator-1 alpha (PGC1α). However, after a rise in the blood glucose level with subsequent hyperinsulinemia, insulin promotes the phosphorylation of FoxO1, leading to its nuclear exclusion, which results in a decrease in PEPCK and G6Pase expression and the subsequent inhibition of HGP [53].
In this context, increased expression of H19 in the liver of diabetic mice is associated with increased HGP and glucose intolerance. In contrast, the loss of H19 function improves insulin sensitivity and suppresses HGP. H19 induces hypomethylation of the promoter of hepatocyte nuclear factor 4 alpha (HNF4A), increasing its expression and stimulating the expression of the gluconeogenic genes PEPCK1 and glucose-6-phosphatase catalytic (G6PC), which reduces the ability of insulin to suppress HGP. Additionally, the glucagon-stimulated upregulation of H19 via the cyclic adenosine monophosphate (cAMP)/PKA pathway induces nuclear translocation of HNF4A and activates the transcription of the gluconeogenic genes G6PC and PCK and, as a result, HGP (Figure 2) [54].
In contrast, decreased H19 expression in IR or associated conditions has been reported in different studies. Gui et al. found that the expression of H19 was reduced in skeletal muscle samples of obese and diabetic mice (db/db mice, used as a model of IR). In addition, they used a model of H19 overexpression in db/db mice to assess its effect on glucose and lipid metabolism by measuring multiple metabolic parameters. They found that H19-overexpressing mice had significantly improved glucose tolerance and decreased serum levels of insulin, triglycerides (TG), and cholesterol, and decreased TG levels in skeletal muscle were identified. Additionally, the overexpression of H19 in muscle increased acetyl-CoA carboxylase 1 (ACC1) and cAMP-activated protein kinase (AMPK) phosphorylation and increased the expression of peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), SIRT1, carnitine palmitoyltransferase 1b, and CD36, which suggests that H19 overexpression ameliorates glucose intolerance, dyslipidemia, and IR (Figure 2). Furthermore, using RNA pull-down and immunoprecipitation assays, the authors identified the interaction of H19 with heterogeneous nuclear ribonucleoprotein A1 (hnRNPA1), which has been reported to play an important role in glucose and lipid metabolism [55].
Gao et al. showed a double-negative feedback loop between the H19 and lethal-7 gene (Let-7) in muscle cells. Acute hyperinsulinemia enhances the activation of the PI3K/AKT pathway, promoting an increase in the biogenesis of miR-let-7, which binds to H19 for its degradation. Therefore, a decrease in H19 levels and higher bioavailability of miR-let-7 could increase glucose uptake. Moreover, miR-let-7-mediated repression of multiple components of the insulin-PI3K-mammalian target of rapamycin (mTOR) pathway, including INSR and IRS2, leads to impaired glucose tolerance [56]. Goyal et al. observed that H19 inhibition promoted an increase in the expression of gluconeogenic genes, which was associated with higher HGP and hyperglycemia and hyperinsulinemia in diabetic (db/db) mouse liver and HepG2 cells. They also identified that H19 downregulation was correlated with the enrichment of G6PC, PCK1, pyruvate carboxylase (PC), and fructose-1,6-biphosphatase (FBP) genes, which are involved in glycolysis and gluconeogenesis. In addition, H19 depletion in HepG2 cells increased the nuclear localization of FoxO1 (an essential transcriptional factor in the regulation of gluconeogenesis and in the insulin response in the liver), impaired insulin signaling, and increased gluconeogenesis, suggesting the functional relevance of decreased H19 levels in the pathophysiology of T2D [57]. Subsequently, the same working group demonstrated that the inhibition of H19 caused a significant increase in FoxO1 transcript and protein levels and impaired glucose, insulin, and pyruvate tolerance. Additionally, they identified that, in the absence of H19, p53 significantly occupied the FoxO1 promoter, suggesting that the inhibition of H19 promotes the transcription of FoxO1 through p53. Furthermore, they demonstrated that the inhibition of H19 in vivo induced hyperglycemia and hyperinsulinemia and increased the transcription of hepatic gluconeogenic genes. These results suggest that H19 inhibition deregulates both circulatory and metabolic profiles and hepatic gluconeogenesis, possibly via the p53/FoxO1 axis (Figure 2) [58].
In another study, elevated H19 expression in the skeletal muscle of mice improved insulin sensitivity. In contrast, H19 knockout (KO) decreased insulin-stimulated glucose uptake in muscle cells; this observed effect could be explained by the disruption of the H19/dual-specificity phosphatase 27 (DUSP27)/AMPK axis. The authors suggested that a reduction in H19 results in the lower expression of DUSP27 with the subsequent reduction in the phosphorylation of AMPK (at Thr172) and ACC1 (at Ser79), resulting in an insulin-resistant state. Moreover, the overexpression of DUSP27 in H19 KO mice resulted in an increase in AMPK phosphorylation and insulin-stimulated glucose uptake similar to wild-type H19 mice and improved mitochondrial biogenesis, fatty acid biosynthesis, glucose uptake, and muscle insulin sensitivity (Figure 2) [59].
Non-alcoholic fatty liver disease, a consequence of the high production and accumulation of lipids in the liver, is considered a key factor in the development of IR and T2D [60]. A mechanism has been proposed in which H19 promotes hepatic lipid synthesis by binding to PTBP1, which interacts with SREBP-1c, allowing greater stability, increasing its cleavage and nuclear translocation, and improving its transcriptional activity [61]. Activation of PI3K/pyruvate dehydrogenase kinase 1/PKB is required for insulin to induce SREBP-1c expression in the liver, which, together with glucokinase (GK), exerts a synergistic effect on glycolytic and lipogenic genes. It has been proposed that hyperinsulinemia, which prevails in IR, could induce a decrease in IRS-2 content. This event, in turn, could account for the IR-mediated repression of gluconeogenic genes, whereas insulin could still activate lipogenic genes by IRS-1. It has been suggested that mTOR complex 1 (mTORC1) is involved in SREBP-1c expression. As mTORC1 integrates not only signals arising from insulin but also nutrients, it has been suggested that nutrient signals such as amino acids could be excessive in obesity and replace the deficient insulin signal, thus worsening IR. On the other hand, endoplasmic reticulum stress, which is present in the liver of IR rodents, has been shown to activate SREBP-1c, inducing IR through the activation of stress kinases as JNK [62].
High-fat diet (HFD)-induced H19 upregulation promotes MLX-interacting protein-like (MLXIPL) expression, a glucose-dependent lipogenic transcription factor, and promotes its nuclear translocation. In addition, H19 positively regulates mTORC1 signaling and the activation of the H19/PTBP1/SREBP-1c axis; together, these events induce lipid accumulation in hepatocytes [63]. On the other hand, Liu et al. showed that elevated H19 expression promotes hepatic steatosis via the miR-130a/PPARγ axis, which induces the transcription of lipogenic SREBP-1c, ACC1, and fatty acid synthase (FAS) [64]. As a result that oxidative stress and inflammation play an important role in T2D, endothelial cells were treated with glucose in various doses and intervals, resulting in significant dose- and time-dependent upregulation of H19, and vascular endothelial growth factor (VEGF)-A expression was also significantly upregulated. In contrast, after the administration of glucose, the expression of miR-29b was considerably downregulated, and a proinflammatory environment and oxidative stress were induced, which suggests that the H19/miR-29b/VEGFA pathway plays a role in hyperglycemic-induced endothelial dysfunction. Additionally, after H19 downregulation, the expression of proinflammatory cytokines, the production of ROS, and NADPH oxidase activity all decreased, while miR-29b expression and activation of the AKT/nitric oxide synthase (eNOS) pathway increased [65]. The participation of H19 in different metabolic pathways is summarized in Figure 2.
As a major lncRNA that has been widely studied in metabolic diseases, H19 has been proposed as a biomarker for the diagnosis of IR and T2D; however, at present, this is not possible due to the contradictory findings reported by different studies to date. In a study carried out on samples of subcutaneous (SAT) and visceral (VAT) adipose tissue from women, a significant decrease in the expression levels of H19 in SAT from women with obesity and IR was identified. Additionally, the expression levels of both ACC1 and FAS, two critical molecules required for fatty acid biogenesis, were elevated in obese patients. In addition, the expression of H19 was negatively correlated with FAS, suggesting the essential participation of H19 in fatty acid biogenesis [66]. On the other hand, recent studies have investigated H19 in non-invasive samples, such as blood plasma and serum. At least two studies have reported the elevated expression of H19 in blood plasma samples from diabetic patients compared to healthy patients [65,67]. In contrast, our group identified low H19 expression in blood serum samples from patients with metabolic syndrome [68]. The contradictory results for the expression of H19, as well as the variability in the clinical characteristics of the patients, such as the timing in the progression of IR or T2D, the metabolic control, and the treatment, could contribute to the variation in the expression of lncRNAs. This suggests that, for the diagnosis or prognosis of these diseases, a set of lncRNAs should be used as biomarkers, not only one of them.

3.3. Maternally Expressed 3 (MEG3)

The MEG3 gene is located on chromosome 14q32.2, contains ten exons with an approximate length of 1700 nucleotides, is a maternally expressed imprinted gene with different isoforms generated by alternative splicing, and is expressed in many normal tissues. Several miRNAs have been shown to regulate the expression of MEG3 at the post-transcriptional level. In turn, MEG3 acts as competing endogenous RNA for several miRNAs [69], including miR-466i-5p, miR-574-5p, and miR-770-5p. Interestingly, the genes targeted by miR-466i-5p, miR-574-5p, and miR-770-5p were identified as hub genes modulated by MEG3 in IR conditions; the target genes include prolyl 3-hydroxylase 2 (LEPREL1), integrin alpha M (ITGAM), and collagen type VI alpha chain (COL6A2), which are expressed in adipose tissue and involved in regulating inflammation, fibrosis, and insulin signaling, respectively [70].
Mondal et al. mapped MEG3 binding sites across the genome in BT-549 cells; they reported more than 6800 sites of union. A significant proportion of these sites are located distal to the promoter regions. The authors demonstrated that transforming growth factor-beta (TGF-β) pathway genes are direct targets of MEG3. These results are consistent with another investigation reporting that MEG3 expression was downregulated upon TGF-β1 treatment in human hepatic stellate cells and that its overexpression inhibited TGF-β1-stimulated cell proliferation and induced apoptosis [71]. In this regard, the participation of the TGF-β1 signaling pathway in the development of IR has been identified, either through the effect of miR335-5p on the inhibition of vasohibin-1 expression and stimulation of the TGF-β signaling pathway [72] or through the overexpression of tribbles (protein kinase) induced by TGF-β/glass bottom boat axis [73].
In a mouse model of HFD-induced IR, Chen et al. identified the overexpression of MEG3 and early growth response protein-2 (EGR2), while miR-185 was downregulated. In addition, MEG3 and miR-185-5 expression were negatively correlated, while MEG3 and EGR2 expression were positively correlated. Additionally, in HepG2 cells, the authors observed that elevated MEG3 expression aggravated palmitate-induced IR by regulating the miR-185-5p/EGR2 axis, suggesting that MEG3 may be a useful target in new therapeutic strategies for T2D (Figure 3) [74].
Zhu et al. reported that the expression of MEG3 was upregulated in HFD and obese (ob/ob) mice. Moreover, high fat enhanced MEG3 expression in hepatocytes through histone acetylation. MEG3 overexpression significantly increased FoxO1, G6PC, and PCK1 expression and hepatic gluconeogenesis and suppressed insulin-stimulated glycogen synthesis in primary hepatocytes, which led to increased hepatic IR [75]. Subsequently, the same group identified increased MEG3 expression and activating transcription factor 4 (ATF4) (a factor that regulates gluconeogenesis by affecting the transcriptional activity of FoxO1) and decreased miR-214 expression in liver tissue from HFD and ob/ob mice. Additionally, they showed that MEG3 knockdown upregulated miR-214 expression and downregulated ATF4 expression. These findings suggest that MEG3 functions as a ceRNA of miR-214 to upregulate ATF4 expression, leading to the activation of FoxO1 and FAS, which in turn promotes lipogenesis and IR [76]. In the same year, these researchers proposed that cAMP response element-binding protein (CREB) induces MEG3 upregulation and increases the hepatic gluconeogenic genes PGC-1α, PEPCK, and G6PC through the CREB–CREB-regulated transcriptional coactivator (CRTC2) complex, which contributes to the pathogenesis of hepatic gluconeogenesis, suggesting that MEG3 acts as a ceRNA to upregulate CRTC2 via miR-302a-3p (Figure 3) [77].
Furthermore, in a study carried out in samples of SAT and VAT adipose tissue from women, MEG3 expression in SAT had a significant relationship with obesity indices. In addition, MEG3 expression in SAT was positively correlated with PPARγ and FAS expression (Figure 3), suggesting an essential role for MEG3 in obesity-related conditions [66]. Cheng et al. identified an increase in the expression of MEG3 in endothelial cells isolated from white adipose tissue compared to those from the skeletal muscle or liver of obese mice. Additionally, they observed that MEG3 knockdown induced endothelial senescence in vitro and cellular senescence of hepatic endothelium, promoting obesity-induced IR in obese mice. Researchers have suggested that the manipulation of MEG3 expression may represent a novel approach to managing obesity-associated hepatic endothelial senescence and IR [78].
Additionally, significant MEG3 overexpression has been reported in patients with T2D, and this overexpression was positively correlated with poor glycemic control, IR, transcriptional markers of senescence, and inflammation [35]. Liu et al. reported that MEG expression was markedly downregulated, while miR-140-5p was upregulated in circulating endothelial progenitor cells (EPCs) of patients with metabolic syndrome. miR-140-5p is associated with cellular senescence. Moreover, treatment with pioglitazone significantly increased MEG3 expression and decreased miR-140-5p levels in circulating EPCs, suggesting that MEG3 acts as a ceRNA of miR-140-5p (Figure 3) [79].

3.4. Myocardial Infarction-Associated Transcript (MIAT)

MIAT is also known as Gomafu or retinal non-coding RNA 2; its gene is located on the short arm of human chromosome 22q12.1. MIAT contains multiple spliced exons, with a final transcript size of approximately 10,000 nucleotides, which is polyadenylated; it is not exported to the cytoplasm and is concentrated in the cell nucleus [80]. MIAT can bind to and sequester miRNAs via base pairing, functioning as a ceRNA to regulate mRNA expression. For example, MIAT inhibited high-mobility group box 1 (HMGB1) expression by competitively binding to miR-204-5p to regulate the injury of cerebral artery occlusion after cerebral ischemia in rats [81]. Some studies have identified relationships between MIAT and several diseases, such as schizophrenia, prostate cancer, lung cancer, diabetes, and complications of diabetes [82]. Significantly increased levels of MIAT have been reported in patients with T2D, possibly related to poor glycemic control, IR, cellular senescence, and inflammation [35]. Moreover, the plasma levels of MIAT were significantly increased in patients with acute myocardial infarction (AMI) [83]. Genetic variants in MIAT were initially recognized as a plausible biomarker linked to the development of AMI in the Japanese population [84].
As in most lncRNAs, the ceRNA function is a molecular mechanism through which MIAT exerts its pathogenic effects. In this regard, MIAT decreases the levels of miR-29c, resulting in an increase in lysyl oxidase-like 2 (LOXL2), an enzyme that catalyzes the oxidative deamination of lysine and hydroxylysine residues in tropocollagen and tropoelastin [85]. LOXL2 is a highly expressed protein in hepatic IR in response to lipotoxicity. Circulating LOXL2 levels were increased in T2D patients with clinical fibrosis [86]. In contrast, a significant increase in MIAT has been reported in diabetic rats and endothelial cells exposed to high glucose concentrations. Additionally, MIAT overexpression in endothelial cells has been reported to upregulate VEGF levels by inhibiting miR-150-5p, which suggests that there is an interplay among MIAT, miR-150-5p, and VEGF to promote angiogenesis, which is related to metabolic diseases (Figure 3) [87].
Ying et al. reported that a reduction in miR-150 increases the plasma levels of proinflammatory cytokines such as TNF-α, IL-1β, and IL-6, with the subsequent activation of JNK; these events could participate in the development of IR [88]. Hepatic IR could be generated by the inhibition of MIAT, which leads to the overexpression of miR-145 and a significant reduction in the phosphorylation of AKT, affecting the PI3K/AKT pathway, which leads to IR and decreased cell viability [89,90]. miR-214 is another molecular target of MIAT [91]. Reduction in miR-214 is associated with the IR state in cellular or animal models and in obese patients [92]. As mentioned above, MIAT decreases the levels of miRNAs with anti-inflammatory functions. It has been suggested that a reduction in miRNAs linked to anti-inflammatory functions induces an inflammatory response and the subsequent activation of serine/threonine kinases, such as JNK, a negative regulator of insulin activity via the PI3K/AKT pathway [93]. Moreover, MIAT promotes the upregulation of FoxO1, and this effect is associated with the ceRNA activity of MIAT on miR-139-5p in hepatocytes [94] (Figure 3). Considering the above-mentioned studies, MIAT, as with other lncRNAs, could impact the development of IR by regulating important genes involved in immune response signaling pathways, a mechanism that has become increasingly recognized as important in the development of metabolic diseases, including IR.

3.5. Steroid Receptor RNA Activator (SRA)

SRA is a type of lncRNA that coordinates the functions of various transcription factors, enhances steroid receptor-dependent gene expression, and serves as a distinct scaffold for corepressor complexes. SRA functions as an RNA coactivator for several nuclear hormone receptors, including androgen, estrogen, progesterone, glucocorticoid, thyroid hormone, retinoic acid, and vitamin D receptors [95,96,97], and is predominantly cytoplasmic [95]. SRA is encoded by the SRA1 gene, which is located on the short arm of human chromosome 5q31.3. The SRA1 gene is unique in that it encodes RNA transcripts of different lengths and produces an mRNA that encodes a 236/237-amino-acid protein through alternative splicing [98].
Interestingly, SRA is highly expressed in white adipose tissue (WAT) and functions as a transcriptional coactivator of PPARγ in in vitro models, suggesting that adipogenesis and adipocyte function can be affected by the SRA/PPARγ axis. Moreover, SRA increases insulin-stimulated glucose uptake and the phosphorylation of the downstream targets AKT and FoxO1 in adipose tissue [97]. Furthermore, SRA1 knockout mice were observed to have improved insulin sensitivity, decreased liver fat, and increased AKT phosphorylation in WAT, liver, and gastrocnemius muscle [99]. In primary mouse hepatocytes, Chen et al. identified that SRA expression stimulated the insulin-induced phosphorylation of AKT, extracellular signal-regulated kinase 1/2 (ERK1/2), and FoxO1, which decreased when SRA expression was inhibited. The researchers hypothesized that SRA functions as a scaffold to recruit a repressive complex in the liver to suppress FoxO1 and PPARγ-mediated adipose triglyceride lipase (ATGL) transcription [100]. SRA regulates multiple signaling pathways, including SRA/PPARγ/ATGL, AKT/FoxO1, and TNF-α/JNK pathways [98]. These data indicate that SRA plays an essential role in insulin signaling and the regulation of β-oxidation.

3.6. HOX Transcript Antisense RNA (HOTAIR)

HOX transcript antisense RNA (HOTAIR) is a 2.2 kb lncRNA that is generated from the transcription of the antisense strand of the HOXC gene located on human chromosome 12q13.13. HOTAIR induces gene silencing by interacting with gene silencing machinery. The 5’-end of HOTAIR interacts with PRC2, and its 3’-end interacts with lysine-specific demethylase 1 (LSD1). The recruits of PRC2 and LSD1 by HOX transcript antisense RNA (HOTAIR) result in the methylation of histone H3 on lysine 27 and demethylation of H3 on lysine 4, which leads to gene silencing [101,102,103]. Moreover, HOTAIR is overexpressed in several types of cancer; in these pathological states, HOTAIR can be associated with the induction of metastasis, anticancer drug resistance, and epithelial–mesenchymal transition [104].
Li et al. investigated the role of HOTAIR in hepatic IR; their results showed that HOTAIR was overexpressed in patients with T2D compared with normal subjects, and animals on a high-fat diet or with diabetes showed the same upregulation of HOTAIR. Moreover, HOTAIR overexpression in HepG2 cells was associated with the elevated expression of PCK and G6PC genes and the subsequent reduction in glycogen levels and an increase in glucose delivery. In addition, the overexpression of HOTAIR in HepG2 cells decreased the phosphorylation levels of AKT and glycogen synthase kinase 3 beta (GSK3β) after insulin stimulation. Finally, the author suggested that overexpression of HOTAIR decreases the levels of SIRT1, and the overexpression of SIRT1 decreases the insulin-resistant state originating in HepG2 cells by the overexpression of HOTAIR [105].
HOTAIR overexpression in the serum of patients with diabetic retinopathy, along with a decrease in miR-20b expression, was reported. In addition, miR-120b was observed to be negatively correlated with poor glycemic control. HOTAIR was found to have complementary sequences to miR-20b [106]. Majumder et al. demonstrated that HOTAIR expression was consistently increased in the kidneys in both humans and mice with diabetes. Additionally, in glomerular podocytes exposed to high glucose levels, the authors identified high levels of HOTAIR expression, and this upregulation was dependent on the p65 subunit of the proinflammatory transcription factor NF-κB, which could be involved in the pathogenesis of diabetic kidney disease [107]. Similarly, HOTAIR overexpression was found in human retinal endothelial cells (HRECs) exposed to high doses of glucose. In addition, HOTAIR was localized in both the nucleus and perinuclear/cytosolic regions [108].
In contrast, Gao et al. found that HOTAIR was downregulated, whereas, as a target of HOTAIR, miR-34a was upregulated in the heart of diabetic mice and in high-glucose-treated H9c2 cells. Additionally, HOTAIR overexpression has been shown to protect against inflammation and oxidative stress induced by diabetes via the HOTAIR/miR-34a/SIRT1 axis [109]. In another study, the downregulated expression of HOTAIR was identified in both myocardial tissue and serum from patients with diabetic cardiomyopathy compared to patients with diabetes without cardiomyopathy and healthy controls. Interestingly, no significant differences were found in HOTAIR expression between patients with diabetes without cardiomyopathy and healthy subjects. In addition, both the expression level of HOTAIR and the phosphorylation of AKT were decreased in human cardiomyocytes exposed to higher doses of glucose. Thus, HOTAIR overexpression may upregulate the phosphorylation of AKT [110]. The above-described findings suggest an important role for HOTAIR in the pathogenesis of IR, diabetes, and diabetes-associated complications. However, much remains to be elucidated, and future research is needed to identify the effect of HOTAIR on the molecular mechanisms involved in metabolic diseases such as IR and diabetes.

3.7. Growth Arrest-Specific 5 (GAS5)

Growth arrest-specific 5 (GAS5), whose gene is located on chromosome 1q25.1, was first characterized as non-coding lncRNA by Coccia et al., followed by Raho et al. [111,112]. Interestingly, the locus that encodes GAS5 harbors several C/D box small nucleolar RNAs (snoRNAs), RNAs related to RNA methylation, and splicing and editing of mRNAs [113]. GAS5 was shown to participate in the cell cycle and modulation of insulin secretion in mouse pancreatic β-cells. Therefore, its participation in the maintenance of the identity and function of β-cells has also been implicated in the pathogenesis of T2D in mice [114]. However, in a study conducted in humans, GAS5 was one of the lncRNAs that showed a greater significant association in terms of its altered expression with T2D. However, this association was lost when adjusted for HOMA-IR and senescence markers, which means that this association could be closely linked to IR and downstream inflammatory signaling [35]. In contrast, in disorders associated with IR, such as polycystic ovary syndrome (PCOS), GAS5 expression was found to be downregulated: GAS5 expression was decreased in the serum of PCOS patients with IR, and the researchers found a negative correlation between HOMA-IR and GAS5 expression. GAS5 is a suggested biomarker for the diagnostic and predictive value of IR [115].

3.8. Gm15622

Gm15622 was significantly enriched in the liver and overexpressed due to an HFD. This overexpression favored the accumulation of lipids and liver steatosis in obese mice as a consequence of feeding an HFD. This lncRNA plays a vital role in the expression of lipid synthesis genes such as FAS. Gm15622 regulates SREBP-1c expression by competing for the binding of miR-742-3p in alpha mouse liver 12 (AML-12) cells. In other words, Gm15622 can act as a molecular sponge or ceRNA for miR-742-3p. Interestingly, metformin inhibits FAS and SREBP-1c expression by targeting Gm15622. Thus, metformin exerts its effect by regulating a network of genes involving Gm15622, miR-742-3p, and SREBP-1c [116]. However, further experimental studies are needed to demonstrate the existence of a gene orthologous to Gm28220202 in humans and to identify its role in IR and/or diabetes.

3.9. Regulator of Insulin Sensitivity and Autophagy (Risa)

Regulator of insulin sensitivity and autophagy (Risa) or AK044604 is a potential lncRNA identified in different tissues, including the heart, kidney, liver, lung, muscle, spleen, thyroid, and white adipose tissue. Risa is located at the 10qB4 region close to the locus of SIRT1, an essential regulator of insulin sensitivity, autophagy, and metabolism. Risa is a polyadenylated lncRNA, and its overexpression significantly reduced the insulin-induced phosphorylation level of the insulin receptor, AKT, and GSK3β, which suggests that the overexpression of Risa induces IR [117]. Although this example illustrates a potential lncRNA that participates in the induction of IR and related diseases, studies in patients with IR and T2D are necessary.

3.10. Downregulated Expression-Related Hexose/Glucose Transport Enhancer (DREH)

Downregulated expression-related hexose/glucose transport enhancer (DREH) was initially named for its downregulated expression by hepatitis B virus X protein (HBx). DREH is an lncRNA in HBx transgenic mice [118]. Although the role of this lncRNA has not been directly demonstrated in IR, some studies suggest that DREH could be involved. For example, metformin treatment has been shown to reduce IR [119]. DREH expression does not facilitate glucose uptake in adipocytes unless its expression is lowered [120]. Interestingly, treatment with metformin leads to the dysregulation of several lncRNAs, including the deregulation of DREH expression [121].
As previously described, several mechanisms have been reported to explain the key role of lncRNAs in the development of IR. However, studies have identified several molecular targets for only a few lncRNAs described in this manuscript. On the other hand, the mechanisms linked to molecular, cellular, and biochemical actions of lncRNAs vary according to the tissue, biological species, and the severity of IR (Figure 4). Table 1 summarizes studies that researched the association between the aberrant expression of other lncRNAs and their main molecular targets during IR or associated pathologies.

4. Conclusions and Future Perspectives

Insulin resistance is a multifactorial disease associated with metabolic syndrome and is considered a risk factor for the development of T2D, dyslipidemias, obesity, and cardiovascular diseases. Environmental risk factors, such as a sedentary lifestyle, stress, and malnutrition, and genetic factors, such as single-nucleotide polymorphisms (SNPs) and copy number variation, can contribute to the development of IR. However, in recent years, non-coding RNAs, including miRNAs and lncRNAs, have acquired increased relevance, the latter being the most clinically important because they are attributed various functions, such as acting as master regulators of different genetic, epigenetic, and metabolic processes, under normal physiological conditions and in metabolic disorders related to diseases. Various authors have associated lncRNAs, such as MIAT, MALAT1, H19, MEG3, and HOTAIR, with inflammatory and lipogenic processes that can lead to IR development through interactions with mRNAs, miRNAs, transcription factors, and proteins. However, a direct relationship between lncRNAs and the insulin signaling pathway or glucose metabolism has not yet been identified. Most of the presented results are from in vivo and in vitro models, and studies in patients need to be conducted to demonstrate the utility of these molecules by generating and implementing panels of the lncRNAs described here. In conclusion, it is essential to continue conducting research that clarifies the direct involvement of lncRNAs in the molecular mechanisms that contribute to IR and their molecular interaction networks that regulate insulin signaling pathways. In addition, it is necessary to demonstrate the usefulness of these biomolecules as potential biomarkers for the diagnosis and prognosis of the development of IR, as well as their potential application as therapeutic targets.

Author Contributions

V.A.T.-F., E.F.-A. and Ó.d.M.-H.: conceptualization, writing, reviewing, and editing; F.O.B.-A., M.A.R.-V. and B.E.E.-C.: writing, reviewing, and editing; C.A.L.-V. and V.A.T.-F.: preparation of figures; L.d.C.A.-R., N.N.-T. and M.R.: reviewing and editing. All authors have read and agreed to the published version of the manuscript.

Funding

No funding was received for this article.

Institutional Review Board Statement

Not applicable for studies not involving humans or animals.

Informed Consent Statement

Not applicable for studies not involving humans.

Data Availability Statement

Not applicable.

Acknowledgments

The figures included in this manuscript were self-made in the BioRender online application.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ACC1Acetyl-CoA carboxylase 1
AGEsAdvanced glycation end products
AKTProtein kinase B
AMIAcute myocardial infarction
AML-12Alpha mouse liver 12 cells
AMPKAMP-activated protein kinase
ATF4Activating transcription factor 4
ATGLAdipose triglyceride lipase
cAMPCyclic adenosine monophosphate
ceRNACompeting endogenous RNA
CoACoenzyme A
CPT1bCarnitine palmitoyltransferase 1b
CREBcAMP responsive element-binding protein
CRTC2Regulated transcription coactivator-2
CVDCardiovascular diseases
DAGDiacylglycerol
DNMTDNA methyltransferase
DREHHexose/glucose transport enhancer
DUSP27Dual-specificity phosphatase 27
EGR2Early growth response protein 2
eNOSNitric oxide synthase
EPCsEndothelial progenitor cells
ERK1/2Extracellular signal-regulated kinase 1/2
EZH2Enhancer of zeste polycomb repressive complex 2
FASFatty acid synthase
FBPFructose-1,6-biphosphatase
FFAFree fatty acids
FGFFibroblast growth factor
FoxO1Forkhead box O1
G6PCGlucose-6-phosphatase catalytic
GAS5Growth arrest specific 5
GKGlucokinase
GLUT4Glucose transporter type 4
GSK3βGlycogen synthase kinase 3 beta
HBxHepatitis B virus X
HFDHigh-fat diet
HGPHepatic glucose production
HMGB1High-mobility group box 1
HNF4AHepatocyte nuclear factor 4 alpha
hnRNPA1Heterogeneous nuclear ribonucleoprotein A1
HOMA-IRInsulin resistance- homeostatic model assessment
HOTAIRHOX transcript antisense RNA
HRECsHuman retinal endothelia cells
IGFInsulin-like growth factor
IKKInhibitor of nuclear factor kappa
ILInterleukin
INSRInsulin receptor
Let-7Lethal-7
LOXL2Lysyl oxidase like 2
IRInsulin resistance
IRSInsulin receptor substrate
JNKc-Jun-N-terminal kinase
KSRPK-homology splicing regulatory protein
LSD1Lysine-specific demethylase 1
MALAT1Metastasis associated lung adenocarcinoma transcript 1
MAPKMitogen-activated protein kinase
MCP-1Monocyte chemoattractant protein-1
MEG3Maternally expressed gene 3
MIATMyocardial infarction associated transcript
MLXIPLMLX-interacting protein-like
mTORMammalian target of rapamycin
mTORC1mTOR complex 1
NADPHNicotinamide adenine dinucleotide phosphate oxidase
NF-κBNuclear factor kappa B
PCPyruvate carboxylase
PCK/PEPCKPhosphoenolpyruvate carboxykinase
PCOSPolycystic ovary syndrome
PDK1Pyruvate dehydrogenase kinase 1
PDX1Pancreatic and duodenal homeobox 1
PGC-1αPeroxisome proliferator-activated receptor gamma coactivator 1-alpha
PI3KPhosphatidylinositol 3 kinase
PKPhosphokinase
PPARPeroxisome proliferator-activated receptor
PRC2Polycomb repressive complex 2
PTBP1Polypyrimidine tract-binding protein 1
RisaRegulator of insulin sensitivity and autophagy
ROSReactive oxygen species
SAA3Serum amyloid antigen 3
SAHS-adenosylhomocysteine
SAHHS-adenosylhomocysteine hydrolase
SATSubcutaneous adipose tissue
SCO1Cytochrome C oxidase
SFASaturated fatty acids
SIRT1Sirtuin 1
SRASteroid receptor RNA activator
SREBP-1cSterol regulatory element-binding transcription factor 1c
T2DType 2 diabetes
TGTriglycerides
TGF-βTransforming growth factor-beta
TNF-αTumor necrosis factor-α
UPRUnfolded protein response
VASH1Vasohibin-1
VATVisceral adipose tissue
VEGFAVascular endothelial growth factor-A
WATWhite adipose tissue

References

  1. Petersen, M.C.; Shulman, G.I. Mechanisms of insulin action and insulin resistance. Physiol. Rev. 2018, 98, 2133–2223. [Google Scholar] [CrossRef] [Green Version]
  2. Brown, A.E.; Walker, M. Genetics of insulin resistance and the metabolic syndrome. Curr. Cardiol. Rep. 2016, 18, 75. [Google Scholar] [CrossRef] [Green Version]
  3. White, M.F. Insulin signaling in health and disease. Science 2003, 302, 1710–1711. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Gutiérrez-Rodelo, C.; Roura-Guiberna, A.; Olivares-Reyes, J.A. Molecular mechanisms of insulin resistance: An update. Gac Med. Mex. 2017, 153, 214–228. [Google Scholar] [PubMed]
  5. Wu, H.; Ballantyne, C.M. Skeletal muscle inflammation and insulin resistance in obesity. J. Clin. Investig. 2017, 127, 43–54. [Google Scholar] [CrossRef] [PubMed]
  6. Engin, A.B. What Is Lipotoxicity? Adv. Exp. Med. Biol. 2017, 960, 197–220. [Google Scholar]
  7. Dupuis, J.; Langenberg, C.; Prokopenko, I.; Saxena, R.; Soranzo, N.; Jackson, A.U.; Wheeler, E.; Glazer, N.L.; Bouatia-Naji, N.; Gloyn, A.L.; et al. New genetic loci implicated in fasting glucose homeostasis and their impact on type 2 diabetes risk. Nat. Genet. 2010, 42, 105–116. [Google Scholar] [CrossRef] [PubMed]
  8. Watanabe, R.M. The genetics of insulin resistance: Where’s Waldo? Curr. Diab. Rep. 2010, 10, 476–484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Vivacqua, A.; De Marco, P.; Belfiore, A.; Maggiolini, M. Recent advances on the role of microRNAs in both insulin resistance and cancer. Curr. Pharm. Des. 2017, 23, 3658–3666. [Google Scholar] [CrossRef]
  10. Jones, A.; Danielson, K.M.; Benton, M.C.; Ziegler, O.; Shah, R.; Stubbs, R.S.; Das, S.; Macartney-Coxson, D. miRNA signatures of insulin resistance in obesity. Obesity 2017, 25, 1734–1744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Chen, T.; Zhang, Y.; Liu, Y.; Zhu, D.; Yu, J.; Li, G.; Sun, Z.; Wang, W.; Jiang, H.; Hong, Z. MiR-27a promotes insulin resistance and mediates glucose metabolism by targeting PPAR-γ-mediated PI3K/AKT signaling. Aging 2019, 11, 7510–7524. [Google Scholar] [CrossRef] [PubMed]
  12. Włodarski, A.; Strycharz, J.; Wróblewski, A.; Kasznicki, J.; Drzewoski, J.; Śliwińska, A. The role of microRNAs in metabolic syndrome-related oxidative stress. Int. J. Mol. Sci. 2020, 21, 6902. [Google Scholar] [CrossRef] [PubMed]
  13. Yang, Q.; Vijayakumar, A.; Kahn, B.B. Metabolites as regulators of insulin sensitivity and metabolism. Nat. Rev. Mol. Cell. Biol. 2018, 19, 654–672. [Google Scholar] [CrossRef] [PubMed]
  14. Yazıcı, D.; Sezer, H. Insulin resistance, obesity and lipotoxicity. Adv. Exp. Med. Biol. 2017, 960, 277–304. [Google Scholar] [PubMed]
  15. Mittra, S.; Bansal, V.S.; Bhatnagar, P.K. From a glucocentric to a lipocentric approach towards metabolic syndrome. Drug Discov. Today. 2008, 13, 211–218. [Google Scholar] [CrossRef] [PubMed]
  16. Villalobos-Labra, R.; Subiabre, M.; Toledo, F.; Pardo, F.; Sobrevia, L. Endoplasmic reticulum stress and development of insulin resistance in adipose, skeletal, liver, and foetoplacental tissue in diabesity. Mol. Aspects Med. 2019, 66, 49–61. [Google Scholar] [CrossRef] [Green Version]
  17. Solinas, G.; Becattini, B. JNK at the crossroad of obesity, insulin resistance, and cell stress response. Mol. Metab. 2017, 6, 174–184. [Google Scholar] [CrossRef] [PubMed]
  18. Tamer, F.; Ulug, E.; Akyol, A.; Nergiz-Unal, R. The potential efficacy of dietary fatty acids and fructose induced inflammation and oxidative stress on the insulin signaling and fat accumulation in mice. Food Chem. Toxicol. 2020, 135, 110914. [Google Scholar] [CrossRef]
  19. Song, F.; Schmidt, A.M. Glycation and insulin resistance: Novel mechanisms and unique targets? Arterioscler. Thromb. Vasc. Biol. 2012, 32, 1760–1765. [Google Scholar] [CrossRef] [Green Version]
  20. Ruiz, H.H.; Ramasamy, R.; Schmidt, A.M. Advanced glycation end products: Building on the concept of the "common soil" in metabolic disease. Endocrinology 2020, 161, bqz006. [Google Scholar] [CrossRef]
  21. Ottum, M.S.; Mistry, A.M. Advanced glycation end-products: Modifiable environmental factors profoundly mediate insulin resistance. J. Clin. Biochem. Nutr. 2015, 57, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Luc, K.; Schramm-Luc, A.; Guzik, T.J.; Mikolajczyk, T.P. Oxidative stress and inflammatory markers in prediabetes and diabetes. J. Physiol. Pharmacol. 2019, 70, 809–824. [Google Scholar]
  23. Yaribeygi, H.; Farrokhi, F.R.; Butler, A.E.; Sahebkar, A. Insulin resistance: Review of the underlying molecular mechanisms. J. Cell Physiol. 2019, 234, 8152–8161. [Google Scholar] [CrossRef]
  24. Keane, K.N.; Cruzat, V.F.; Carlessi, R.; de Bittencourt, P.I., Jr.; Newsholme, P. Molecular events linking oxidative stress and inflammation to insulin resistance and β-cell dysfunction. Oxid. Med. Cell. Longev. 2015, 2015, 181643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Zhao, M.X.; Zhou, B.; Ling, L.; Xiong, X.Q.; Zhang, F.; Chen, Q.; Li, Y.H.; Kang, Y.M.; Zhu, G.Q. Salusin-β contributes to oxidative stress and inflammation in diabetic cardiomyopathy. Cell Death Dis. 2017, 8, e2690. [Google Scholar] [CrossRef] [Green Version]
  26. Pereira, S.; Park, E.; Mori, Y.; Haber, C.A.; Han, P.; Uchida, T.; Stavar, L.; Oprescu, A.I.; Koulajian, K.; Ivovic, A.; et al. FFA-induced hepatic insulin resistance in vivo is mediated by PKCδ, NADPH oxidase, and oxidative stress. Am. J. Physiol. Endocrinol. Metab. 2014, 307, E34–E46. [Google Scholar] [CrossRef] [Green Version]
  27. Xiao, Y.; Hu, J.; Yin, W. Systematic identification of non-coding RNAs. Adv. Exp. Med. Biol. 2018, 1094, 9–18. [Google Scholar]
  28. Marchese, F.P.; Raimondi, I.; Huarte, M. The multidimensional mechanisms of long noncoding RNA function. Genome Biol. 2017, 18, 206. [Google Scholar] [CrossRef] [Green Version]
  29. Mathy, N.W.; Chen, X.M. Long non-coding RNAs (lncRNAs) and their transcriptional control of inflammatory responses. J. Biol. Chem. 2017, 292, 12375–12382. [Google Scholar] [CrossRef] [Green Version]
  30. Derrien, T.; Johnson, R.; Bussotti, G.; Tanzer, A.; Djebali, S.; Tilgner, H.; Guernec, G.; Martin, D.; Merkel, A.; Knowles, D.G.; et al. The GENCODE v7 catalog of human long noncoding RNAs: Analysis of their gene structure, evolution, and expression. Genome Res. 2012, 22, 1775–1789. [Google Scholar] [CrossRef] [Green Version]
  31. Hombach, S.; Kretz, M. Non-coding RNAs: Classification, biology and functioning. Adv. Exp. Med. Biol. 2016, 937, 3–17. [Google Scholar]
  32. Yoon, J.H.; Kim, J.; Gorospe, M. Long noncoding RNA turnover. Biochimie 2015, 117, 15–21. [Google Scholar] [CrossRef] [Green Version]
  33. Frankish, A.; Diekhans, M.; Ferreira, A.M.; Johnson, R.; Jungreis, I.; Loveland, J.; Mudge, J.M.; Sisu, C.; Wright, J.; Armstrong, J.; et al. GENCODE reference annotation for the human and mouse genomes. Nucleic Acids Res. 2019, 47, D766–D773. [Google Scholar] [CrossRef] [Green Version]
  34. Zhang, X.; Hamblin, M.H.; Yin, K.J. The long noncoding RNA Malat1: Its physiological and pathophysiological functions. RNA Biol. 2017, 14, 1705–1714. [Google Scholar] [CrossRef]
  35. Sathishkumar, C.; Prabu, P.; Mohan, V.; Balasubramanyam, M. Linking a role of lncRNAs (long non-coding RNAs) with insulin resistance, accelerated senescence, and inflammation in patients with type 2 diabetes. Hum. Genom. 2018, 12, 41. [Google Scholar] [CrossRef]
  36. Pielok, A.; Marycz, K. Non-coding RNAs as potential novel biomarkers for early diagnosis of hepatic insulin resistance. Int. J. Mol. Sci. 2020, 21, 4182. [Google Scholar] [CrossRef] [PubMed]
  37. Losko, M.; Kotlinowski, J.; Jura, J. Long noncoding rnas in metabolic syndrome related disorders. Mediat. Inflamm. 2016, 2016, 5365209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Yan, Y.; Song, D.; Song, X.; Song, C. The role of lncRNA MALAT1 in cardiovascular disease. IUBMB Life 2020, 72, 334–342. [Google Scholar] [CrossRef] [PubMed]
  39. Li, X.; Zeng, L.; Cao, C.; Lu, C.; Lian, W.; Han, J.; Zhang, X.; Zhang, J.; Tang, T.; Li, M. Long noncoding RNA MALAT1 regulates renal tubular epithelial pyroptosis by modulated miR-23c targeting of ELAVL1 in diabetic nephropathy. Exp. Cell Res. 2017, 350, 327–335. [Google Scholar] [CrossRef] [PubMed]
  40. Zhang, Y.; Wu, H.; Wang, F.; Ye, M.; Zhu, H.; Bu, S. Long non-coding RNA MALAT1 expression in patients with gestational diabetes mellitus. Int. J. Gynaecol. Obstet. 2018, 140, 164–169. [Google Scholar] [CrossRef] [PubMed]
  41. Liu, J.Y.; Yao, J.; Li, X.M.; Song, Y.C.; Wang, X.Q.; Li, Y.J.; Yan, B.; Jiang, Q. Pathogenic role of lncRNA-MALAT1 in endothelial cell dysfunction in diabetes mellitus. Cell Death Dis. 2014, 5, e1506. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Zhou, X.; Liu, S.; Cai, G.; Kong, L.; Zhang, T.; Ren, Y.; Wu, Y.; Mei, M.; Zhang, L.; Wang, X. Long non coding RNA MALAT1 promotes tumor growth and metastasis by inducing epithelial-mesenchymal transition in oral squamous cell carcinoma. Sci. Rep. 2015, 5, 15972. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Biswas, S.; Thomas, A.A.; Chen, S.; Aref-Eshghi, E.; Feng, B.; Gonder, J.; Sadikovic, B.; Chakrabarti, S. MALAT1: An epigenetic regulator of inflammation in diabetic retinopathy. Sci. Rep. 2018, 8, 6526. [Google Scholar] [CrossRef]
  44. Xiong, L.; Gong, Y.; Wu, L.; Li, J.; He, W.; Zhu, X.; Xiao, H. LncRNA-Malat1 is involved in lipotoxicity-induced ß-cell dysfunction and the therapeutic effect of exendin-4 via Ptbp1. Endocrinology 2020, 161, bqaa065. [Google Scholar] [CrossRef]
  45. Dong, N.; Xu, B.; Shi, H. Long noncoding RNA MALAT1 acts as a competing endogenous RNA to regulate Amadori-glycated albumin-induced MCP-1 expression in retinal microglia by a microRNA-124-dependent mechanism. Inflamm. Res. 2018, 67, 913–925. [Google Scholar] [CrossRef]
  46. Puthanveetil, P.; Chen, S.; Feng, B.; Gautam, A.; Chakrabarti, S. Long non-coding RNA MALAT1 regulates hyperglycaemia induced inflammatory process in the endothelial cells. J. Cell. Mol. Med. 2015, 19, 1418–1425. [Google Scholar] [CrossRef]
  47. Yan, C.; Chen, J.; Chen, N. Long noncoding RNA MALAT1 promotes hepatic steatosis and insulin resistance by increasing nuclear SREBP-1c protein stability. Sci. Rep. 2016, 6, 22640. [Google Scholar] [CrossRef]
  48. Brannan, C.I.; Dees, E.C.; Ingram, R.S.; Tilghman, S.M. The product of the H19 gene may function as an RNA. Mol. Cell. Biol. 1990, 10, 28–36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Gabory, A.; Jammes, H.; Dandolo, L. The H19 locus: Role of an imprinted non-coding RNA in growth and development. Bioessays 2010, 32, 473–480. [Google Scholar] [CrossRef] [PubMed]
  50. Bao, M.H.; Szeto, V.; Yang, B.B.; Zhu, S.Z.; Sun, H.S.; Feng, Z.P. Long non-coding RNAs in ischemic stroke. Cell Death Dis. 2018, 9, 281. [Google Scholar] [CrossRef] [Green Version]
  51. Nilsson, E.; Matte, A.; Perfilyev, A.; de Mello, V.D.; Käkelä, P.; Pihlajamäki, J.; Ling, C. Epigenetic alterations in human liver from subjects with type 2 diabetes in parallel with reduced folate levels. J. Clin. Endocrinol. Metab. 2015, 100, E1491–E1501. [Google Scholar] [CrossRef] [PubMed]
  52. Zhou, J.; Yang, L.; Zhong, T.; Mueller, M.; Men, Y.; Zhang, N.; Xie, J.; Giang, K.; Chung, H.; Sun, X.; et al. H19 lncRNA alters DNA methylation genome wide by regulating S-adenosylhomocysteine hydrolase. Nat. Commun. 2015, 6, 10221. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Ramnanan, C.; Edgerton, D.S.; Cherrington, A.D. The role of insulin in the regulation of PEPCK and gluconeogenesis in vivo. US Endocrinol. 2009, 5, 34–39. [Google Scholar] [CrossRef]
  54. Zhang, N.; Geng, T.; Wang, Z.; Zhang, R.; Cao, T.; Camporez, J.P.; Cai, S.Y.; Liu, Y.; Dandolo, L.; Shulman, G.I.; et al. Elevated hepatic expression of H19 long noncoding RNA contributes to diabetic hyperglycemia. JCI Insight 2018, 3, e120304. [Google Scholar] [CrossRef] [Green Version]
  55. Gui, W.; Zhu, W.F.; Zhu, Y.; Tang, S.; Zheng, F.; Yin, X.; Lin, X.; Li, H. LncRNAH19 improves insulin resistance in skeletal muscle by regulating heterogeneous nuclear ribonucleoprotein A1. Cell Commun. Signal. 2020, 18, 173. [Google Scholar] [CrossRef] [PubMed]
  56. Gao, Y.; Wu, F.; Zhou, J.; Yan, L.; Jurczak, M.J.; Lee, H.Y.; Yang, L.; Mueller, M.; Zhou, X.B.; Dandolo, L.; et al. The H19/let-7 double-negative feedback loop contributes to glucose metabolism in muscle cells. Nucleic Acids Res. 2014, 42, 13799–13811. [Google Scholar] [CrossRef] [PubMed]
  57. Goyal, N.; Sivadas, A.; Shamsudheen, K.V.; Jayarajan, R.; Verma, A.; Sivasubbu, S.; Scaria, V.; Datta, M. RNA sequencing of db/db mice liver identifies lncRNA H19 as a key regulator of gluconeogenesis and hepatic glucose output. Sci. Rep. 2017, 7, 8312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Goyal, N.; Tiwary, S.; Kesharwani, D.; Datta, M. Long non-coding RNA H19 inhibition promotes hyperglycemia in mice by upregulating hepatic FoxO1 levels and promoting gluconeogenesis. J. Mol. Med. 2019, 97, 115–126. [Google Scholar] [CrossRef]
  59. Geng, T.; Liu, Y.; Xu, Y.; Jiang, Y.; Zhang, N.; Wang, Z.; Carmichael, G.G.; Taylor, H.S.; Li, D.; Huang, Y. H19 lncRNA promotes skeletal muscle insulin sensitivity in part by targeting AMPK. Diabetes 2018, 67, 2183–2198. [Google Scholar] [CrossRef] [Green Version]
  60. Santoleri, D.; Titchenell, P.M. Resolving the paradox of hepatic insulin resistance. Cell Mol. Gastroenterol. Hepatol. 2019, 7, 447–456. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Liu, C.; Yang, Z.; Wu, J.; Zhang, L.; Lee, S.; Shin, D.J.; Tran, M.; Wang, L. Long noncoding RNA H19 interacts with polypyrimidine tract-binding protein 1 to reprogram hepatic lipid homeostasis. Hepatology 2018, 67, 1768–1783. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Ferré, P.; Foufelle, F. Hepatic steatosis: A role for de novo lipogenesis and the transcription factor SREBP-1c. Diabetes Obes. Metab. 2010, 12, 83–92. [Google Scholar] [CrossRef]
  63. Wang, H.; Cao, Y.; Shu, L.; Zhu, Y.; Peng, Q.; Ran, L.; Wu, J.; Luo, Y.; Zuo, G.; Luo, J.; et al. Long non-coding RNA (lncRNA) H19 induces hepatic steatosis through activating MLXIPL and mTORC1 networks in hepatocytes. J. Cell. Mol. Med. 2020, 24, 1399–1412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Liu, J.; Tang, T.; Wang, G.D.; Liu, B. LncRNA-H19 promotes hepatic lipogenesis by directly regulating miR-130a/PPARγ axis in non-alcoholic fatty liver disease. Biosci. Rep. 2019, 39, BSR20181722. [Google Scholar] [CrossRef] [Green Version]
  65. Cheng, X.W.; Chen, Z.F.; Wan, Y.F.; Zhou, Q.; Wang, H.; Zhu, H.Q. Long non-coding RNA H19 suppression protects the endothelium against hyperglycemic-induced inflammation via inhibiting expression of miR-29b target gene vascular endothelial growth factor a through activation of the protein kinase B/endothelial nitric oxide synthase pathway. Front. Cell Dev. Biol. 2019, 7, 263. [Google Scholar] [PubMed]
  66. Daneshmoghadam, J.; Omidifar, A.; Akbari Dilmaghani, N.; Karimi, Z.; Emamgholipour, S.; Shanaki, M. The gene expression of long non-coding RNAs (lncRNAs): MEG3 and H19 in adipose tissues from obese women and its association with insulin resistance and obesity indices. J. Clin. Lab. Anal. 2021, 35, e23741. [Google Scholar] [CrossRef]
  67. Fawzy, M.S.; Abdelghany, A.A.; Toraih, E.A.; Mohamed, A.M. Circulating long noncoding RNAs H19 and GAS5 are associated with type 2 diabetes but not with diabetic retinopathy: A preliminary study. Bosn. J. Basic Med. Sci. 2020, 20, 365–371. [Google Scholar] [CrossRef] [Green Version]
  68. Tello-Flores, V.A.; Valladares-Salgado, A.; Ramírez-Vargas, M.A.; Cruz, M.; Del-Moral-Hernández, O.; Cahua-Pablo, J.; Ramírez, M.; Hernández-Sotelo, D.; Armenta-Solis, A.; Flores-Alfaro, E. Altered levels of MALAT1 and H19 derived from serum or serum exosomes associated with type-2 diabetes. Noncoding RNA Res. 2020, 5, 71–76. [Google Scholar] [CrossRef]
  69. Zhang, X.; Rice, K.; Wang, Y.; Chen, W.; Zhong, Y.; Nakayama, Y.; Zhou, Y.; Klibanski, A. Maternally expressed gene 3 (MEG3) noncoding ribonucleic acid: Isoform structure, expression, and functions. Endocrinology 2010, 151, 939–947. [Google Scholar] [CrossRef] [Green Version]
  70. Zhang, Y.; Fu, Y.; Zheng, Y.; Wen, Z.; Wang, C. Identification of differentially expressed mRNA and the Hub mRNAs modulated by lncRNA Meg3 as a competing endogenous RNA in brown adipose tissue of mice on a high-fat diet. Adipcyte 2020, 9, 346–358. [Google Scholar]
  71. Mondal, T.; Subhash, S.; Vaid, R.; Enroth, S.; Uday, S.; Reinius, B.; Mitra, S.; Mohammed, A.; James, A.R.; Hoberg, E.; et al. MEG3 long noncoding RNA regulates the TGF-β pathway genes through formation of RNA-DNA triplex structures. Nat. Commun. 2015, 6, 7743. [Google Scholar] [CrossRef] [Green Version]
  72. Tang, X.W.; Qin, Q.X. miR-335-5p induces insulin resistance and pancreatic islet β-cell secretion in gestational diabetes mellitus mice through VASH1-mediated TGF-β signaling pathway. J. Cell Physiol. 2019, 234, 6654–6666. [Google Scholar] [CrossRef] [PubMed]
  73. Hong, S.H.; Kang, M.; Lee, K.S.; Yu, K. High fat diet-induced TGF-β/Gbb signaling provokes insulin resistance through the tribbles expression. Sci. Rep. 2016, 6, 30265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Chen, D.L.; Shen, D.Y.; Han, C.K.; Tian, Y. LncRNA MEG3 aggravates palmitate-induced insulin resistance by regulating miR-185-5p/Egr2 axis in hepatic cells. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 5456–5467. [Google Scholar] [PubMed]
  75. Zhu, X.; Wu, Y.B.; Zhou, J.; Kang, D.M. Upregulation of lncRNA MEG3 promotes hepatic insulin resistance via increasing FoxO1 expression. Biochem. Biophys. Res. Commun. 2016, 469, 319–325. [Google Scholar] [CrossRef] [PubMed]
  76. Zhu, X.; Li, H.; Wu, Y.; Zhou, J.; Yang, G.; Wang, W. lncRNA MEG3 promotes hepatic insulin resistance by serving as a competing endogenous RNA of miR-214 to regulate ATF4 expression. Int. J. Mol. Med. 2019, 43, 345–357. [Google Scholar] [CrossRef] [PubMed]
  77. Zhu, X.; Li, H.; Wu, Y.; Zhou, J.; Yang, G.; Wang, W.; Kang, D.; Ye, S. CREB-upregulated lncRNA MEG3 promotes hepatic gluconeogenesis by regulating miR-302a-3p-CRTC2 axis. J. Cell. Biochem. 2019, 120, 4192–4202. [Google Scholar] [CrossRef] [PubMed]
  78. Cheng, X.; Shihabudeen Haider Ali, M.S.; Moran, M.; Viana, M.P.; Schlichte, S.L.; Zimmerman, M.C.; Khalimonchuk, O.; Feinberg, M.W.; Sun, X. Long non-coding RNA Meg3 deficiency impairs glucose homeostasis and insulin signaling by inducing cellular senescence of hepatic endothelium in obesity. Redox Biol. 2021, 40, 101863. [Google Scholar] [CrossRef]
  79. Liu, H.Z.; Wang, Q.Y.; Zhang, Y.; Qi, D.T.; Li, M.W.; Guo, W.Q.; Ma, Y.H.; Wang, L.Y.; Chen, Y.; Gao, C.Y. Pioglitazone up-regulates long non-coding RNA MEG3 to protect endothelial progenitor cells via increasing HDAC7 expression in metabolic syndrome. Biomed. Pharmacother. 2016, 78, 101–109. [Google Scholar] [CrossRef] [PubMed]
  80. Naveed, A.; Fortini, E.; Li, R.; Fox, A.H. Long non-coding rnas and nuclear body formation and function. In Molecular Biology of Long Non-Coding RNAs; Khalil, A., Ed.; Springer: Cham, Switzerland, 2019; pp. 65–84. [Google Scholar]
  81. Deng, W.; Fan, C.; Shen, R.; Wu, Y.; Du, R.; Teng, J. Long noncoding MIAT acting as a ceRNA to sponge microRNA-204-5p to participate in cerebral microvascular endothelial cell injury after cerebral ischemia through regulating HMGB1. J. Cell. Physiol. 2020, 235, 4571–4586. [Google Scholar] [CrossRef] [PubMed]
  82. Sun, C.; Huang, L.; Li, Z.; Leng, K.; Xu, Y.; Jiang, X.; Cui, Y. Long non-coding RNA MIAT in development and disease: A new player in an old game. J. Biomed. Sci. 2018, 25, 23. [Google Scholar] [CrossRef] [PubMed]
  83. Azat, M.; Huojiahemaiti, X.; Gao, R.; Peng, P. Long noncoding RNA MIAT: A potential role in the diagnosis and mediation of acute myocardial infarction. Mol. Med. Rep. 2019, 20, 5216–5222. [Google Scholar] [CrossRef]
  84. Ishii, N.; Ozaki, K.; Sato, H.; Mizuno, H.; Susumu, S.; Takahashi, A.; Miyamoto, Y.; Ikegawa, S.; Kamatani, N.; Hori, M.; et al. Identification of a novel non-coding RNA, MIAT, that confers risk of myocardial infarction. J. Hum. Genet. 2006, 51, 1087–1099. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Qu, Y.; Xiao, H.; Xiao, W.; Xiong, Z.; Hu, W.; Gao, Y.; Ru, Z.; Wang, C.; Bao, L.; Wang, K.; et al. Upregulation of MIAT regulates LOXL2 expression by competitively binding miR-29c in clear cell renal cell carcinoma. Cell. Physiol. Biochem. 2018, 48, 1075–1087. [Google Scholar] [CrossRef] [PubMed]
  86. Dongiovanni, P.; Meroni, M.; Baselli, G.A.; Bassani, G.A.; Rametta, R.; Pietrelli, A.; Maggioni, M.; Facciotti, F.; Trunzo, V.; Badiali, S.; et al. Insulin resistance promotes Lysyl Oxidase Like 2 induction and fibrosis accumulation in non-alcoholic fatty liver disease. Clin. Sci. 2017, 131, 1301–1315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Yan, B.; Yao, J.; Liu, J.Y.; Li, X.M.; Wang, X.Q.; Li, Y.J.; Tao, Z.F.; Song, Y.C.; Chen, Q.; Jiang, Q. lncRNA-MIAT regulates microvascular dysfunction by functioning as a competing endogenous RNA. Circ. Res. 2015, 116, 1143–1156. [Google Scholar] [CrossRef] [PubMed]
  88. Ying, W.; Tseng, A.; Chang, R.C.; Wang, H.; Lin, Y.L.; Kanameni, S.; Brehm, T.; Morin, A.; Jones, B.; Splawn, T.; et al. miR-150 regulates obesity-associated insulin resistance by controlling B cell functions. Sci. Rep. 2016, 6, 20176. [Google Scholar] [CrossRef]
  89. 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, 517–523. [Google Scholar] [CrossRef] [PubMed]
  90. Chen, S.; Chen, H.; Yu, C.; Lu, R.; Song, T.; Wang, X.; Tang, W.; Gao, Y. Long noncoding RNA myocardial infarction associated transcript promotes the development of thoracic aortic by targeting microRNA-145 via the PI3K/Akt signaling pathway. J. Cell. Biochem. 2019, 120, 14405–14413. [Google Scholar] [CrossRef] [PubMed]
  91. Huang, X.; Gao, Y.; Qin, J.; Lu, S. lncRNA MIAT promotes proliferation and invasion of HCC cells via sponging miR-214. Am. J. Physiol. Gastrointest Liver Physiol. 2018, 314, G559–G565. [Google Scholar] [CrossRef]
  92. Cheng, F.; Yuan, G.; He, J.; Shao, Y.; Zhang, J.; Guo, X. Aberrant expression of miR-214 is associated with obesity-induced insulin resistance as a biomarker and therapeutic. Diagn. Pathol. 2020, 15, 18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Calderari, S.; Diawara, M.R.; Garaud, A.; Gauguier, D. Biological roles of microRNAs in the control of insulin secretion and action. Physiol. Genom. 2017, 49, 1–10. [Google Scholar] [CrossRef] [Green Version]
  94. Yan, C.; Li, J.; Feng, S.; Li, Y.; Tan, L. Long noncoding RNA Gomafu upregulates Foxo1 expression to promote hepatic insulin resistance by sponging miR-139-5p. Cell Death Dis. 2018, 9, 289. [Google Scholar] [CrossRef] [PubMed]
  95. Liu, C.; Wu, H.T.; Zhu, N.; Shi, Y.N.; Liu, Z.; Ao, B.X.; Liao, D.F.; Zheng, X.L.; Qin, L. Steroid receptor RNA activator: Biologic function and role in disease. Clin. Chim. Acta 2016, 459, 137–146. [Google Scholar] [CrossRef] [PubMed]
  96. Colley, S.M.; Leedman, P.J. Steroid Receptor RNA Activator—A nuclear receptor coregulator with multiple partners: Insights and challenges. Biochimie 2011, 93, 1966–1972. [Google Scholar] [CrossRef]
  97. Xu, B.; Gerin, I.; Miao, H.; Vu-Phan, D.; Johnson, C.N.; Xu, R.; Chen, X.W.; Cawthorn, W.P.; MacDougald, O.A.; Koenig, R.J. Multiple roles for the non-coding RNA SRA in regulation of adipogenesis and insulin sensitivity. PLoS ONE 2010, 5, e14199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Sheng, L.; Ye, L.; Zhang, D.; Cawthorn, W.P.; Xu, B. New Insights into the long non-coding RNA SRA: Physiological functions and mechanisms of action. Front. Med. 2018, 5, 244. [Google Scholar] [CrossRef] [PubMed]
  99. Liu, S.; Sheng, L.; Miao, H.; Saunders, T.L.; MacDougald, O.A.; Koenig, R.J.; Xu, B. SRA gene knockout protects against diet-induced obesity and improves glucose tolerance. J. Biol. Chem. 2014, 289, 13000–13009. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Chen, G.; Yu, D.; Nian, X.; Liu, J.; Koenig, R.J.; Xu, B.; Sheng, L. LncRNA SRA promotes hepatic steatosis through repressing the expression of adipose triglyceride lipase (ATGL). Sci. Rep. 2016, 6, 35531. [Google Scholar] [CrossRef] [PubMed]
  101. Rinn, J.L.; Kertesz, M.; Wang, J.K.; Squazzo, S.L.; Xu, X.; Brugmann, S.A.; Goodnough, L.H.; Helms, J.A.; Farnham, P.J.; Segal, E.; et al. Functional demarcation of active and silent chromatin domains in human HOX loci by noncoding RNAs. Cell 2007, 129, 1311–1323. [Google Scholar] [CrossRef] [Green Version]
  102. Tsai, M.C.; Manor, O.; Wan, Y.; Mosammaparast, N.; Wang, J.K.; Lan, F.; Shi, Y.; Segal, E.; Chang, H.Y. Long noncoding RNA as modular scaffold of histone modification complexes. Science 2010, 329, 689–693. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Wu, L.; Murat, P.; Matak-Vinkovic, D.; Murrell, A.; Balasubramanian, S. Binding interactions between long noncoding RNA HOTAIR and PRC2 proteins. Biochemistry 2013, 52, 9519–9527. [Google Scholar] [CrossRef] [PubMed]
  104. Cantile, M.; Di Bonito, M.; Tracey De Bellis, M.; Botti, G. Functional interaction among lncRNA HOTAIR and microRNAs in cancer and other human diseases. Cancers 2021, 13, 570. [Google Scholar] [CrossRef] [PubMed]
  105. Li, M.; Guo, Y.; Wang, X.J.; Duan, B.H.; Li, L. HOTAIR participates in hepatic insulin resistance via regulating SIRT1. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 7883–7890. [Google Scholar]
  106. Shaker, O.G.; Abdelaleem, O.O.; Mahmoud, R.H.; Abdelghaffar, N.K.; Ahmed, T.I.; Said, O.M.; Zaki, O.M. Diagnostic and prognostic role of serum miR-20b, miR-17-3p, HOTAIR, and MALAT1 in diabetic retinopathy. IUBMB Life 2019, 71, 310–320. [Google Scholar] [CrossRef]
  107. Majumder, S.; Hadden, M.J.; Thieme, K.; Batchu, S.N.; Niveditha, D.; Chowdhury, S.; Yerra, V.G.; Advani, S.L.; Bowskill, B.B.; Liu, Y.; et al. Dysregulated expression but redundant function of the long non-coding RNA HOTAIR in diabetic kidney disease. Diabetologia 2019, 62, 2129–2142. [Google Scholar] [CrossRef] [PubMed]
  108. Biswas, S.; Feng, B.; Chen, S.; Liu, J.; Aref-Eshghi, E.; Gonder, J.; Ngo, V.; Sadikovic, B.; Chakrabarti, S. The long non-coding RNA HOTAIR is a critical epigenetic mediator of angiogenesis in diabetic retinopathy. Investig. Ophthalmol. Vis. Sci. 2021, 62, 20. [Google Scholar] [CrossRef] [PubMed]
  109. Gao, L.; Wang, X.; Guo, S.; Xiao, L.; Liang, C.; Wang, Z.; Li, Y.; Liu, Y.; Yao, R.; Zhang, Y. LncRNA HOTAIR functions as a competing endogenous RNA to upregulate SIRT1 by sponging miR-34a in diabetic cardiomyopathy. J. Cell. Physiol. 2019, 234, 4944–4958. [Google Scholar] [CrossRef]
  110. Qi, K.; Zhong, J. LncRNA HOTAIR improves diabetic cardiomyopathy by increasing viability of cardiomyocytes through activation of the PI3K/Akt pathway. Exp. Ther. Med. 2018, 16, 4817–4823. [Google Scholar] [CrossRef] [Green Version]
  111. Coccia, E.M.; Cicala, C.; Charlesworth, A.; Ciccarelli, C.; Rossi, G.B.; Philipson, L.; Sorrentino, V. Regulation and expression of a growth arrest-specific gene (gas5) during growth, differentiation, and development. Mol. Cell. Biol. 1992, 12, 3514–3521. [Google Scholar] [CrossRef] [Green Version]
  112. Raho, G.; Barone, V.; Rossi, D.; Philipson, L.; Sorrentino, V. The gas 5 gene shows four alternative splicing patterns without coding for a protein. Gene 2000, 256, 13–17. [Google Scholar] [CrossRef]
  113. Liang, J.; Wen, J.; Huang, Z.; Chen, X.P.; Zhang, B.X.; Chu, L. Small nucleolar RNAS: Insight into their function in cancer. Front. Oncol. 2019, 9, 587. [Google Scholar] [CrossRef] [Green Version]
  114. Jin, F.; Wang, N.; Zhu, Y.; You, L.; Wang, L.; De, W.; Tang, W. Downregulation of long noncoding RNA Gas5 affects cell cycle and insulin secretion in mouse pancreatic β cells. Cell. Physiol. Biochem. 2017, 43, 2062–2073. [Google Scholar] [CrossRef] [PubMed]
  115. Lin, H.; Xing, W.; Li, Y.; Xie, Y.; Tang, X.; Zhang, Q. Downregulation of serum long noncoding RNA GAS5 may contribute to insulin resistance in PCOS patients. Gynecol. Endocrinol. 2018, 34, 784–788. [Google Scholar] [CrossRef]
  116. Ma, M.; Duan, R.; Shen, L.; Liu, M.; Ji, Y.; Zhou, H.; Li, C.; Liang, T.; Li, X.; Guo, L. The lncRNA Gm15622 stimulates SREBP-1c expression and hepatic lipid accumulation by sponging the miR-742-3p in mice. J. Lipid Res. 2020, 61, 1052–1064. [Google Scholar] [CrossRef] [Green Version]
  117. Wang, Y.; Hu, Y.; Sun, C.; Zhuo, S.; He, Z.; Wang, H.; Yan, M.; Liu, J.; Luan, Y.; Dai, C.; et al. Down-regulation of Risa improves insulin sensitivity by enhancing autophagy. FASEB J. 2016, 30, 3133–3145. [Google Scholar] [CrossRef] [Green Version]
  118. Huang, J.F.; Guo, Y.J.; Zhao, C.X.; Yuan, S.X.; Wang, Y.; Tang, G.N.; Zhou, W.P.; Sun, S.H. Hepatitis B virus X protein (HBx)-related long noncoding RNA (lncRNA) down-regulated expression by HBx (Dreh) inhibits hepatocellular carcinoma metastasis by targeting the intermediate filament protein vimentin. Hepatology 2013, 57, 1882–1892. [Google Scholar] [CrossRef]
  119. Villalva-Pérez, J.M.; Ramírez-Vargas, M.A.; Serafín-Fabían, J.I.; Ramírez, M.; Elena Moreno-Godínez, M.; Espinoza-Rojo, M.; Flores-Alfaro, E. Characterization of Huh7 cells after the induction of insulin resistance and post-treatment with metformin. Cytotechnology 2020, 72, 499–511. [Google Scholar] [CrossRef]
  120. Takahashi, N.; Kimura, A.P.; Ohmura, K.; Naito, S.; Yoshida, M.; Ieko, M. Knockdown of long noncoding RNA dreh facilitates cell surface GLUT4 expression and glucose uptake through the involvement of vimentin in 3T3-L1 adipocytes. Gene 2020, 735, 144404. [Google Scholar] [CrossRef]
  121. Takahashi, N.; Kimura, A.P.; Otsuka, K.; Ohmura, K.; Naito, S.; Yoshida, M.; Ieko, M. Dreh, a long noncoding RNA repressed by metformin, regulates glucose transport in C2C12 skeletal muscle cells. Life Sci. 2019, 236, 116906. [Google Scholar] [CrossRef] [PubMed]
  122. Shu, L.; Hou, G.; Zhao, H.; Huang, W.; Song, G.; Ma, H. Resveratrol improves high-fat diet-induced insulin resistance in mice by downregulating the lncRNA NONMMUT008655.2. Am. J. Transl. Res. 2020, 12, 1–18. [Google Scholar]
  123. Gao, H.; Kerr, A.; Jiao, H.; Hon, C.C.; Rydén, M.; Dahlman, I.; Arner, P. Long non-coding RNAs associated with metabolic traits in human white adipose tissue. EBioMedicine 2018, 30, 248–260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Li, P.; Ruan, X.; Yang, L.; Kiesewetter, K.; Zhao, Y.; Luo, H.; Chen, Y.; Gucek, M.; Zhu, J.; Cao, H. A liver-enriched long non-coding RNA, lncLSTR, regulates systemic lipid metabolism in mice. Cell Metab. 2015, 21, 455–467. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Thomas, A.A.; Feng, B.; Chakrabarti, S. ANRIL: A regulator of VEGF in diabetic retinopathy. Investig. Ophthalmol. Vis. Sci. 2017, 58, 470–480. [Google Scholar] [CrossRef]
  126. Lin, M.; Mao, Z.J. lncRNA-mRNA competing endogenous RNA network in IR-hepG2 cells ameliorated by APBBR decreasing ROS levels: A systematic analysis. PeerJ 2020, 8, e8604. [Google Scholar] [CrossRef] [PubMed]
  127. Han, M.; You, L.; Wu, Y.; Gu, N.; Wang, Y.; Feng, X.; Xiang, L.; Chen, Y.; Zeng, Y.; Zhong, T. RNA-sequencing analysis reveals the potential contribution of lncRNAs in palmitic acid-induced insulin resistance of skeletal muscle cells. Biosci. Rep. 2020, 40, BSR20192523. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Li, X.; Zhao, Z.; Gao, C.; Rao, L.; Hao, P.; Jian, D.; Li, W.; Tang, H.; Li, M. The diagnostic value of whole blood lncRNA ENST00000550337.1 for pre-diabetes and type 2 diabetes mellitus. Exp. Clin. Endocrinol. Diabetes 2017, 125, 377–383. [Google Scholar] [CrossRef]
  129. Sun, J.; Ruan, Y.; Wang, M.; Chen, R.; Yu, N.; Sun, L.; Liu, T.; Chen, H. Differentially expressed circulating LncRNAs and mRNA identified by microarray analysis in obese patients. Sci. Rep. 2016, 6, 35421. [Google Scholar] [CrossRef] [Green Version]
  130. Wang, J.; Yang, W.; Chen, Z.; Chen, J.; Meng, Y.; Feng, B.; Sun, L.; Dou, L.; Li, J.; Cui, Q.; et al. Long noncoding RNA lncSHGL recruits hnRNPA1 to suppress hepatic gluconeogenesis and lipogenesis. Diabetes 2018, 67, 581–593. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Role of metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) in the molecular mechanisms of insulin resistance. The downregulation of MALAT1 affects the modulation of polypyrimidine tract-binding protein 1 (PTBP1), demonstrating that it contributes to the dysfunction of pancreatic β-cells and the development of type 2 diabetes (T2D). In states of hyperglycemia and high levels of free fatty acids (FFAs), MALAT1 is overexpressed through the activation of the PDX1/MALAT1/PTBP1 axis and indirectly promotes the overexpression of tumor necrosis factor-alpha (TNF-α), interleukins (IL)-6, and IL-1β when bound to polycomb repressive complex 2 (PRC2), inhibiting the expression of anti-inflammatory genes. Glycated albumin induces monocyte chemoattractant protein-1 (MCP-1) expression, which depends on the MALAT1/miR-124 axis. MALAT1 directly interacts with SREBP-1c, inhibiting its ubiquitination and increasing its activity, which leads to the expression of its target lipogenic genes, thus inducing intracellular lipid accumulation and IR. Increased transcription of proinflammatory and lipogenic genes leads to the activation of threonine/serine kinases c-Jun-N-terminal kinases (JNK) and phosphokinase C (PKC), which are associated with the negative regulation of insulin signaling.
Figure 1. Role of metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) in the molecular mechanisms of insulin resistance. The downregulation of MALAT1 affects the modulation of polypyrimidine tract-binding protein 1 (PTBP1), demonstrating that it contributes to the dysfunction of pancreatic β-cells and the development of type 2 diabetes (T2D). In states of hyperglycemia and high levels of free fatty acids (FFAs), MALAT1 is overexpressed through the activation of the PDX1/MALAT1/PTBP1 axis and indirectly promotes the overexpression of tumor necrosis factor-alpha (TNF-α), interleukins (IL)-6, and IL-1β when bound to polycomb repressive complex 2 (PRC2), inhibiting the expression of anti-inflammatory genes. Glycated albumin induces monocyte chemoattractant protein-1 (MCP-1) expression, which depends on the MALAT1/miR-124 axis. MALAT1 directly interacts with SREBP-1c, inhibiting its ubiquitination and increasing its activity, which leads to the expression of its target lipogenic genes, thus inducing intracellular lipid accumulation and IR. Increased transcription of proinflammatory and lipogenic genes leads to the activation of threonine/serine kinases c-Jun-N-terminal kinases (JNK) and phosphokinase C (PKC), which are associated with the negative regulation of insulin signaling.
Ijms 22 07256 g001
Figure 2. Imprinted maternally expressed transcript (H19) alterations and their molecular mechanisms. (a) The decrease in folates due to hyperglycemia leads to hypomethylation (Me) of the H19 promoter, increasing its transcription. H19 inactivates S-adenosylhomocysteine hydrolase (SAHH) and inhibits hepatocyte nuclear factor 4 alpha (HNF4A) promoter methylation. (b) The glucagon-stimulated upregulation of H19 via the cyclic adenosine monophosphate/ protein kinase A (cAMP/PKA) axis induces nuclear translocation of HNF4A and activates the transcription of gluconeogenic genes glucose-6-phosphatase catalytic (G6PC) and phosphoenolpyruvate carboxykinase (PCK). (c) Inhibition of miR-29b by H19 increases vascular endothelial growth factor-A (VEGFA) expression, leading to decreased activation of protein kinase B (AKT) and nitric oxide synthase (eNOS), increasing reactive oxygen species (ROS) production and proinflammatory cytokines. (d) High levels of free fatty acids (FFAs) promote H19 overexpression, which induces increased mTOR complex 1 (mTORC) signaling, nuclear translocation of MLX-interacting protein-like (MLXIPL) and element-binding transcription factor 1c (SREBP-1c), and the interaction of polypyrimidine tract-binding protein 1 (PTBP1) with SREBP-1c, which in turn induces lipid accumulation. (e) The binding of H19 to miR-130a induces the nuclear translocation of proliferator-activated receptor gamma (PPARγ), which activates the transcription of lipogenic genes such as acetyl-CoA carboxylase 1 (ACC1), fatty acid synthase (FAS), and cytochrome C oxidase (SCO1), promoting the intracellular accumulation of lipids. (f) Hyperinsulinemia induces the activation of far upstream element-binding protein (KSRP), which forms a complex with primiR Let7, promoting its maturation to miR-Let7, which binds H19, insulin receptor (INSR), and dual-specificity phosphatase 27 (DUSP27), degrading them. (g) The decrease in DUSP27 reduces the activation of cAMP-activated protein kinase (AMPK) and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), which inhibits glucose uptake and mitochondrial biogenesis. (h) H19 downregulation induces the binding of p53 to the forkhead box O1 (FoxO1) promoter, increasing its transcription. (I) The insulin signaling pathway is deregulated, decreasing the phosphorylation of INSR and AKT by phosphatidylinositol 3-kinase (PI3K), which induces an increase in mTORC and FoxO1, promoting the translocation of FoxO1 to the nucleus and the transcription of gluconeogenic genes.
Figure 2. Imprinted maternally expressed transcript (H19) alterations and their molecular mechanisms. (a) The decrease in folates due to hyperglycemia leads to hypomethylation (Me) of the H19 promoter, increasing its transcription. H19 inactivates S-adenosylhomocysteine hydrolase (SAHH) and inhibits hepatocyte nuclear factor 4 alpha (HNF4A) promoter methylation. (b) The glucagon-stimulated upregulation of H19 via the cyclic adenosine monophosphate/ protein kinase A (cAMP/PKA) axis induces nuclear translocation of HNF4A and activates the transcription of gluconeogenic genes glucose-6-phosphatase catalytic (G6PC) and phosphoenolpyruvate carboxykinase (PCK). (c) Inhibition of miR-29b by H19 increases vascular endothelial growth factor-A (VEGFA) expression, leading to decreased activation of protein kinase B (AKT) and nitric oxide synthase (eNOS), increasing reactive oxygen species (ROS) production and proinflammatory cytokines. (d) High levels of free fatty acids (FFAs) promote H19 overexpression, which induces increased mTOR complex 1 (mTORC) signaling, nuclear translocation of MLX-interacting protein-like (MLXIPL) and element-binding transcription factor 1c (SREBP-1c), and the interaction of polypyrimidine tract-binding protein 1 (PTBP1) with SREBP-1c, which in turn induces lipid accumulation. (e) The binding of H19 to miR-130a induces the nuclear translocation of proliferator-activated receptor gamma (PPARγ), which activates the transcription of lipogenic genes such as acetyl-CoA carboxylase 1 (ACC1), fatty acid synthase (FAS), and cytochrome C oxidase (SCO1), promoting the intracellular accumulation of lipids. (f) Hyperinsulinemia induces the activation of far upstream element-binding protein (KSRP), which forms a complex with primiR Let7, promoting its maturation to miR-Let7, which binds H19, insulin receptor (INSR), and dual-specificity phosphatase 27 (DUSP27), degrading them. (g) The decrease in DUSP27 reduces the activation of cAMP-activated protein kinase (AMPK) and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), which inhibits glucose uptake and mitochondrial biogenesis. (h) H19 downregulation induces the binding of p53 to the forkhead box O1 (FoxO1) promoter, increasing its transcription. (I) The insulin signaling pathway is deregulated, decreasing the phosphorylation of INSR and AKT by phosphatidylinositol 3-kinase (PI3K), which induces an increase in mTORC and FoxO1, promoting the translocation of FoxO1 to the nucleus and the transcription of gluconeogenic genes.
Ijms 22 07256 g002
Figure 3. Molecular effects of maternally expressed gene 3 (MEG3) and myocardial infarction-associated transcript (MIAT) on insulin resistance. (a) High-fat diet (HFD)-induced cAMP response element-binding protein (CREB) activation promotes MEG3 expression. (b) MEG3 overexpression inhibits miR-214, promoting activating transcription factor 4 (ATF4) expression, activating forkhead box O1(FoxO1), and increasing fatty acid synthase (FAS), leading to a lipotoxic environment. (c) MEG3 inhibits the action of miR-302a-3p, which leads to the activation of the CREB-CREB-regulated transcriptional coactivator (CRTC2) complex, increasing their interaction with ATF4 and leading to the expression of gluconeogenic proteins. (d) MEG3 expression inhibits miR-185-5p, leading to early growth response protein-2 (EGR2) overexpression. (e) Inhibition of miR-140-5p by MEG3 promotes cellular senescence. (f) MEG3 overexpression increases proliferator-activated receptor gamma (PPARγ) expression and FAS activation, leading to the production of fatty acids and lipotoxic effects. Furthermore, (1) the overexpression of MIAT prevents the inhibition of FoxO1 by miR-139-5p, promoting FAS expression and raising the levels of FFAs. (2) MIAT inhibits miR-145, thereby inhibiting the phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) pathway, which leads to a reduction in insulin sensitivity. (3) The inhibition of miR-29c by MIAT facilitates the expression and action of lysyl oxidase-like 2 (LOXL2), promoting a lipotoxic environment. (4) After the inhibition of miR-150-5p by MIAT, the levels of tumor necrosis factor-alpha (TNF-α), and interleukins 6, and 1β increase.
Figure 3. Molecular effects of maternally expressed gene 3 (MEG3) and myocardial infarction-associated transcript (MIAT) on insulin resistance. (a) High-fat diet (HFD)-induced cAMP response element-binding protein (CREB) activation promotes MEG3 expression. (b) MEG3 overexpression inhibits miR-214, promoting activating transcription factor 4 (ATF4) expression, activating forkhead box O1(FoxO1), and increasing fatty acid synthase (FAS), leading to a lipotoxic environment. (c) MEG3 inhibits the action of miR-302a-3p, which leads to the activation of the CREB-CREB-regulated transcriptional coactivator (CRTC2) complex, increasing their interaction with ATF4 and leading to the expression of gluconeogenic proteins. (d) MEG3 expression inhibits miR-185-5p, leading to early growth response protein-2 (EGR2) overexpression. (e) Inhibition of miR-140-5p by MEG3 promotes cellular senescence. (f) MEG3 overexpression increases proliferator-activated receptor gamma (PPARγ) expression and FAS activation, leading to the production of fatty acids and lipotoxic effects. Furthermore, (1) the overexpression of MIAT prevents the inhibition of FoxO1 by miR-139-5p, promoting FAS expression and raising the levels of FFAs. (2) MIAT inhibits miR-145, thereby inhibiting the phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) pathway, which leads to a reduction in insulin sensitivity. (3) The inhibition of miR-29c by MIAT facilitates the expression and action of lysyl oxidase-like 2 (LOXL2), promoting a lipotoxic environment. (4) After the inhibition of miR-150-5p by MIAT, the levels of tumor necrosis factor-alpha (TNF-α), and interleukins 6, and 1β increase.
Ijms 22 07256 g003
Figure 4. lncRNAs and transcription factors related to the development of IR. (a) Lipogenic activity is associated with elevated levels of H19, MALAT1, MEG3, and MIAT and their interaction with nuclear proteins, such as element-binding transcription factor 1c (SREBP-1c), proliferator-activated receptor gamma PPARγ, and forkhead box O1 (FoxO1), and in consequence, the expression of lipogenic genes is promoted, e.g., acetyl-CoA carboxylase 1 (ACC1), fatty acid synthase (FAS), and cytochrome C oxidase (SCO1), as well as other proteins such as lysyl oxidase-like 2 (LOXL2) by MIAT, which leads to the accumulation of lipids. (b) MEG3 upregulation activates CREB-regulated transcriptional coactivator (CRTC2), which in turn activates (cAMP response element-binding protein) CREB by binding to activating transcription factor 4 (ATF4) and FoxO1; in addition, hepatocyte nuclear factor 4 alpha (HNF4A) and FoxO1 are activated by H19 overexpression, which induces gluconeogenesis. In contrast, H19 downregulation promotes the transcription of FoxO1 and the gluconeogenic genes. (c) MALAT1 overexpression induces the expression of zeste polycomb homolog 2 (EZH2)/polycomb repressive complex 2 (PRC2), monocyte chemoattractant protein-1 (MCP-1), and serum amyloid antigen 3 (SAA3), which in turn promote the elevation of interleukins 6 and 1β, and tumor necrosis factor-alpha (TNF-α). Additionally, the expression of H19 induces the dephosphorylation of protein kinase B (AKT) and nitric oxide synthase (eNOS), and adenine dinucleotide phosphate oxidase (NADPH) and TNF-α levels increase as a result of the expression of c-Jun-N-terminal kinases (JNK) due to MIAT. (d) MEG3 downregulation promotes miR-140-5p expression, which is related to cellular senescence. Overexpression of MALAT1 activates polypyrimidine tract-binding protein 1 (PTBPI), which is involved in pancreatic β-cell dysfunction. MIAT downregulation affects the phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) pathway signaling, decreasing insulin sensitivity and cell viability.
Figure 4. lncRNAs and transcription factors related to the development of IR. (a) Lipogenic activity is associated with elevated levels of H19, MALAT1, MEG3, and MIAT and their interaction with nuclear proteins, such as element-binding transcription factor 1c (SREBP-1c), proliferator-activated receptor gamma PPARγ, and forkhead box O1 (FoxO1), and in consequence, the expression of lipogenic genes is promoted, e.g., acetyl-CoA carboxylase 1 (ACC1), fatty acid synthase (FAS), and cytochrome C oxidase (SCO1), as well as other proteins such as lysyl oxidase-like 2 (LOXL2) by MIAT, which leads to the accumulation of lipids. (b) MEG3 upregulation activates CREB-regulated transcriptional coactivator (CRTC2), which in turn activates (cAMP response element-binding protein) CREB by binding to activating transcription factor 4 (ATF4) and FoxO1; in addition, hepatocyte nuclear factor 4 alpha (HNF4A) and FoxO1 are activated by H19 overexpression, which induces gluconeogenesis. In contrast, H19 downregulation promotes the transcription of FoxO1 and the gluconeogenic genes. (c) MALAT1 overexpression induces the expression of zeste polycomb homolog 2 (EZH2)/polycomb repressive complex 2 (PRC2), monocyte chemoattractant protein-1 (MCP-1), and serum amyloid antigen 3 (SAA3), which in turn promote the elevation of interleukins 6 and 1β, and tumor necrosis factor-alpha (TNF-α). Additionally, the expression of H19 induces the dephosphorylation of protein kinase B (AKT) and nitric oxide synthase (eNOS), and adenine dinucleotide phosphate oxidase (NADPH) and TNF-α levels increase as a result of the expression of c-Jun-N-terminal kinases (JNK) due to MIAT. (d) MEG3 downregulation promotes miR-140-5p expression, which is related to cellular senescence. Overexpression of MALAT1 activates polypyrimidine tract-binding protein 1 (PTBPI), which is involved in pancreatic β-cell dysfunction. MIAT downregulation affects the phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) pathway signaling, decreasing insulin sensitivity and cell viability.
Ijms 22 07256 g004
Table 1. Roles of other lncRNAs in molecular mechanisms associated with insulin resistance and related diseases.
Table 1. Roles of other lncRNAs in molecular mechanisms associated with insulin resistance and related diseases.
LncRNAsPathologyModelTissueMain TargetRegulating during IRReference
NONMMUT008655.2IRHigh-fat-diet-induced IR miceLiverRegulation of SOC-
S3 via miR-133c, miR-3569-5p, miR-504-3p, and miR-7076-5p
Upregulated in IR.
Resveratrol improves insulin sensitivity by downregulating this lncRNA.
[122]
ASMER-1 (ENSG00000235609.4)
ASMER-2 (CATG00000111229.1)
Obesity and IRHumanWhite adipose PPARγSilencing of ASMER-1 and ASMER-2 decreases the expression of adipogenic genes and reduces the triglyceride accumulation in adipocytes.[123]
lncLSTRWithoutMiceLiverTDP-43/Cyp8b1Depletion of lncLSTR leads to reduced triglyceride levels.[124]
ANRIL (CDKN2B-AS1)DiabetesHRECs cells exposed to high levels of glucose and diabetic miceRetinal PRC2/VEGFHigh levels of glucose promote the overexpression of ANRIL. ANRIL inhibition promotes glucose-induced VEGF overexpression. [125]
CTD-2517M22.14Hepatic IRIR-HepG2 cellsLivermiR-346; miR-1306-5p; miR-18a-3p; miR-1914-5p_R_1/SCDThese lncRNAs positively regulate MAPK, linked to inflammation and IR. The reduction in ROS with a subsequent decrease in the relative expression of all lncRNAs might ameliorate inflammation.[126]
CTD-2600O9.2miR-486-3p/DUSP5
CTD-3014M21.1miR-324-5p, miR-2478_L-1/SCD
LAMA5-AS1miR-18a-3p; miR-516b-5p; miR-763-p3/SCD
HOXB-AS3PC-5p-36422_6/SCD
RP5-1057I20.5miR-23b-5p, miR-185-3p, miR-642a-5p/SCD
MIR4435-2HGmiR-149-3p_L; miR-1304-3p; miR-452-5p/PLK3
ENSMUST00000160839IRPalmitic acid-induced IR C2C12 cellsMyotubesPDK-4PDK4 upregulation has been related to IR and was positively correlated with ENSMUST00000160839.[127]
ENST00000550337.1Pre-diabetes and T2DHumanWhole bloodUndescribedENST00000550337.1 could be a potential diagnostic biomarker for pre-diabetes and T2D.[35,128]
PLUTO
CDKN2BAS1
LincRNA-p21
XIST
PANDA
NBR2
LET
SALRNA1
THRIL
T2DHumanPBMCUndescribedThe majority of the lncRNAs were positively correlated with poor glycemic control, IR, transcriptional markers of senescence, inflammation, and HDAC3 and negatively associated with telomere length.[35]
p5549ObesityHumanWhole blood (in addition, 4 blood components) and adipose tissueTDGThe 3 lncRNAs were negatively correlated with BMI, waist circumference, and waist-to-hip ratio. Only the circulating level of p19461 was negatively associated with IR. TDG is involved in DNA repair. CHCHD3 regulation could protect mitochondrial function, accelerating fat oxidation and reducing obesity.[129]
p21015Undescribed
p19461CHCHD3
lncSHGLObesity and IRHigh-fat-diet-induced IR
mice
Primary mouse hepatocyteshnRNPA1/CALM/AktlncSHGL induced Akt activation via lncSHGL/hnRNPA1/CALM pathway. Downregulation of lncSHGL promotes IR and fatty liver.[130]
IR: Insulin resistance; SOCS3: Suppressor of cytokine signaling 3; PPARγ: Peroxisome proliferator-activated receptor gamma; TDP-43: Transactive response DNA binding protein 43; Cyp8b1: Cytochrome P450 Family 8 Subfamily B Member 1; HRECs: Human retinal endothelial cells; PRC2: Polycomb repressive complex 2; miR: micro-RNA; VEGF: Vascular endothelial growth factor; SCD: Stearoyl-CoA desaturase; DUSP5: Dual specificity phosphatase 5; PLK3: Polo-like kinase 3; ROS: Reactive oxygen species; PDk4: Pyruvate Dehydrogenase Kinase 4; PBMC: Peripheral blood mononuclear cells; HDAC3: Histone deacetylase 3; TDG: Thymine-DNA glycosylase; CHCHD3: Coiled-Coil-Helix-Coiled-Coil-Helix Domain-Containing Protein 3; hnRNPA1: Heterogeneous Nuclear Ribonucleoprotein A1; CALM: Calmodulin; Akt: Protein kinase B.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Tello-Flores, V.A.; Beltrán-Anaya, F.O.; Ramírez-Vargas, M.A.; Esteban-Casales, B.E.; Navarro-Tito, N.; Alarcón-Romero, L.d.C.; Luciano-Villa, C.A.; Ramírez, M.; del Moral-Hernández, Ó.; Flores-Alfaro, E. Role of Long Non-Coding RNAs and the Molecular Mechanisms Involved in Insulin Resistance. Int. J. Mol. Sci. 2021, 22, 7256. https://doi.org/10.3390/ijms22147256

AMA Style

Tello-Flores VA, Beltrán-Anaya FO, Ramírez-Vargas MA, Esteban-Casales BE, Navarro-Tito N, Alarcón-Romero LdC, Luciano-Villa CA, Ramírez M, del Moral-Hernández Ó, Flores-Alfaro E. Role of Long Non-Coding RNAs and the Molecular Mechanisms Involved in Insulin Resistance. International Journal of Molecular Sciences. 2021; 22(14):7256. https://doi.org/10.3390/ijms22147256

Chicago/Turabian Style

Tello-Flores, Vianet Argelia, Fredy Omar Beltrán-Anaya, Marco Antonio Ramírez-Vargas, Brenda Ely Esteban-Casales, Napoleón Navarro-Tito, Luz del Carmen Alarcón-Romero, Carlos Aldair Luciano-Villa, Mónica Ramírez, Óscar del Moral-Hernández, and Eugenia Flores-Alfaro. 2021. "Role of Long Non-Coding RNAs and the Molecular Mechanisms Involved in Insulin Resistance" International Journal of Molecular Sciences 22, no. 14: 7256. https://doi.org/10.3390/ijms22147256

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

Article Metrics

Back to TopTop