Abstract
Increasing evidence suggests that patients with diabetes, particularly type 2 diabetes (T2D), are characterized by an increased risk of developing different types of cancer, so cancer could be proposed as a new T2D-related complication. On the other hand, cancer may also increase the risk of developing new-onset diabetes, mainly caused by anticancer therapies. Hyperinsulinemia, hyperglycemia, and chronic inflammation typical of T2D could represent possible mechanisms involved in cancer development in diabetic patients. MicroRNAs (miRNAs) are a subset of non-coding RNAs, ⁓22 nucleotides in length, which control the post-transcriptional regulation of gene expression through both translational repression and messenger RNA degradation. Of note, miRNAs have multiple target genes and alteration of their expression has been reported in multiple diseases, including T2D and cancer. Accordingly, specific miRNA-regulated pathways are involved in the pathogenesis of both conditions. In this review, a panel of experts from the Italian Association of Medical Oncology (AIOM), Italian Association of Medical Diabetologists (AMD), Italian Society of Diabetology (SID), Italian Society of Endocrinology (SIE), and Italian Society of Pharmacology (SIF) provide a critical view of the evidence about the involvement of miRNAs in the pathophysiology of both T2D and cancer, trying to identify the shared miRNA signature and pathways able to explain the strong correlation between the two conditions, as well as to envision new common pharmacological approaches.
Key words: type 2 diabetes, cancer, miRNAs, miRNA-based drugs
Highlights
-
•
Patients with T2D are characterized by an increased risk of developing different types of cancer.
-
•
Cancer may also increase the risk of developing new cases of diabetes.
-
•
MiRNAs are a subset of non-coding RNAs, which control the post-transcriptional regulation of gene expression.
-
•
Specific miRNA-regulated pathways are involved in the pathogenesis of both T2D and cancer.
-
•
In the near future, envisioning an miRNA-based drug able to treat both T2D and cancer may be possible.
Introduction
Diabetes mellitus (DM) involves a group of metabolic disorders characterized by chronically elevated glycemia. In its two main forms, DM is caused by almost complete immune-mediated β-cell destruction [type 1 diabetes mellitus (T1D)] or by reduced β-cell functional mass [type 2 diabetes mellitus (T2D)]. The number of people with DM has quadrupled in the past three decades, representing one of the fastest-growing health challenges of the 21st century.1,2 According to the International Diabetes Federation (IDF), ∼463 million adults between the ages of 20 and 79 years have DM, 90% being T2D.3
Increasing evidence suggests that patients with DM, particularly T2D, are characterized by an increased risk of developing different types of cancer:4 at the time of cancer diagnosis, ⁓18% of patients have pre-existing DM and the incidence of DM is approximately six times higher in cancer patients than in the general population.5 Accordingly, the increase in the incidence of cancer appears to parallel the increasing incidence of metabolic disorders, including T2D.6 On the other hand, cancer may also increase the risk of developing DM, mainly caused by anticancer therapies (i.e. corticosteroids, chemotherapy, radiotherapy, immunosuppressive therapies).4,7 T2D and cancer share many risk factors: obesity, sedentary lifestyle, unbalanced diet, cigarette smoking, and excessive alcohol consumption.8,9 Hyperinsulinemia, hyperglycemia, and chronic inflammation typical of T2D could be possible mechanisms involved in cancer development. Many cancer cells overexpress insulin receptors and therefore are more responsive to the mitogenic effects of insulin.9 In addition, since glucose excess is an important source of energy for cancer cells (‘Warburg effect’), hyperglycemia could promote tumor growth.8,9 Hyperglycemia could also cause oxidative stress and DNA damage, thus triggering or enhancing the tumorigenic process.9 Lastly, inflammation further promotes neoplastic transformation, cancer cell proliferation, and metastasization.9 On the other hand, tumor cachexia is often associated with glucose intolerance, insulin resistance, and inflammation that predispose to T2D development.7 Cancer-related stress can also induce hyperglycemia and worsen inflammation.7 Finally, DM can occur when cancer affects organs involved in glycemic homeostasis, such as the pancreas and liver.
MicroRNAs (miRNAs) are a subset of non-coding RNAs, ⁓22 nucleotides in length, that control the post-transcriptional regulation of gene expression through either translational repression or messenger RNA (mRNA) degradation.10 Importantly, it has been shown that an individual miRNA can control the expression of more than one target mRNA and that each mRNA may be regulated by multiple miRNAs.10 Growing evidence suggests that miRNAs operate in all organs and tissues, and play significant roles in various regulatory mechanisms, including cell differentiation, proliferation, apoptosis, and many others.10 Under various conditions, cells can also release miRNAs, either in free form or complexed with extracellular vesicles, that can be taken up by other cell types, thereby mediating cell-to-cell communication and coordinating multiple biological functions.11 Indeed, circulating miRNAs have been detected in different human biofluids including plasma, serum, urine, breast milk, cerebrospinal fluid, and saliva.12 It should be noted that recent research on circulating miRNAs highlights their usefulness as biomarkers of diseases; however, this aspect is beyond the scope of the current manuscript. Of note, miRNAs have multiple target genes and alteration of their expression has been reported in multiple diseases, including T2D and cancer.13 Interestingly, specific miRNA-regulated pathways are involved in the pathogenesis of both conditions.
In this review, a panel of experts from the Italian Association of Medical Oncology (AIOM), Italian Association of Medical Diabetologists (AMD), Italian Society of Diabetology (SID), Italian Society of Endocrinology (SIE), and Italian Society of Pharmacology (SIF) provide a critical view of the evidence about the involvement of miRNAs in the pathophysiology of both T2D and cancer, trying to identify the shared miRNA signature and pathways able to explain the strong correlation between the two conditions, as well as to envision new common pharmacological approaches.
The role of miRNAs in the pathogenesis of type 2 diabetes
In the past two decades, a growing number of evidence demonstrated the crucial role of miRNAs in the regulation of glucose homeostasis, insulin secretion, and action.14,15 The metabolic effects of miRNAs are exerted at various levels, both in the pancreatic islets and in peripheral tissues. Accordingly, miRNAs can regulate the two hallmarks of T2D pathogenesis: β-cell failure (Table 1) and peripheral insulin resistance (Table 2).
Table 1.
MiRNAs of importance for β-cell failure
| β-Cell failure | |
|---|---|
| miRNAs | Effects |
| miR-7 | Represses β-cell function,34 regulates GLP-1-mediated insulin release.49 |
| miR-9 | Represses β-cell function.37 |
| miR-15a | Regulates insulin synthesis.33 |
| miR-17-92 cluster | Regulates β-cell replication, restoration and adaptation, and GSIS.24 |
| miR-24 | Reduces insulin promoter activity and insulin mRNA levels and consequently insulin content.33,39 |
| miR-25 | Its overexpression leads to inhibition of insulin biosynthesis and increased β-cell apoptosis.27 |
| miR-26 | Reduces insulin promoter activity and insulin mRNA levels and consequently insulin content.33,39 |
| miR-29 family | Its overexpression negatively affects GSIS and β-cell proliferation.45 |
| miR-30d | Increases insulin gene expression.32 |
| miR-34a | Its overexpression results in sensitization to apoptosis and impaired nutrient-induced insulin secretion.26 |
| miR-92b | Its overexpression leads to inhibition of insulin biosynthesis and increased β-cell apoptosis.27 |
| miR-96 | Represses β-cell function.38 |
| miR-124a | Promotes β-cell dysfunction and death.43 |
| miR-125b-5p | Its down-regulation reduces insulin sensitivity and deregulates β-cell function.46 |
| miR-130a/b | Negatively correlated with insulin exocytosis.15 |
| miR-132 | Controls β-cell identity,48 proliferation and survival,51 regulates GLP-1-mediated insulin release.50 |
| miR-146 | Its overexpression results in sensitization to apoptosis and impaired nutrient-induced insulin secretion.26 |
| miR-148 | Reduces insulin promoter activity and insulin mRNA levels and consequently insulin content.33,39 |
| miR-152 | Negatively correlated with insulin exocytosis.15 |
| miR-153 | Its overexpression induces insulin secretion defects.28 |
| miR-181b | Regulates β-cell replication and GSIS.24 |
| miR-182 | Reduces insulin promoter activity and insulin mRNA levels and consequently insulin content.33,39 |
| miR-187 | Negatively correlated with GSIS.42 |
| miR-200 family | Represses β-cell function.35,166 |
| miR-204 | Blocks insulin production,47 promotes β-cell ER stress.52 |
| miR-212 | Regulates GLP-1-mediated insulin release.50 |
| miR-216a | Its knockout leads to a reduction in islet size, β-cell mass, and insulin levels.25 |
| miR-223 | Its overexpression drives the compensatory β-cell expansion and the maintenance of functional β-cell mass during metabolic stress.22 |
| miR-299-5p | Its reduction promotes β-cell dysfunction and loss.31 |
| miR-320a | Its overexpression initiates pancreatic islet dysfunction by increasing the ROS level, inhibiting proliferation, and inducing apoptosis.29 |
| miR-335 | Negatively correlated with insulin exocytosis.41 |
| miR-375 | Regulates β-cell adaptation to metabolic stress and insulin resistance19 and promotes GSIS33 |
| miR-383 | Its overexpression reverses β-cell apoptosis and oxidative stress.30 |
| miR-455 | Its overexpression drives the compensatory β-cell expansion and the maintenance of functional β-cell mass during metabolic stress.23 |
| miR-770-5p | Promotes β-cell dysfunction and death.44 |
ER, endoplasmic reticulum; GLP-1, glucagon-like peptide-1; GSIS, glucose-stimulated insulin secretion; miRNA, microRNA; mRNA, messenger RNA; ROS, reactive oxygen species.
Table 2.
MiRNAs of importance for insulin resistance
| Peripheral insulin resistance | |
|---|---|
| miRNAs | Effects |
| miR-19a | Regulates PI3K activity in endothelial cells.70 |
| miR-21 | Its overexpression improves the insulin-induced phosphorylation in AKT and the translocation of the GLUT4 in insulin-resistant adipocytes.87 |
| miR-27a | Its overexpression decreases glucose consumption and glucose uptake, and reduces the expression of GLUT4 and PI3K p85β.83 |
| miR-29 family | Controls PI3K-p85α subunit expression;67 its overexpression blunts insulin-stimulated AKT activation and glucose uptake in adipose tissue72 and causes a decrease in levels of GLUT4 which partially induces a decrease in insulin-dependent glucose uptake in skeletal muscle.84 |
| miR-30d | Its overexpression decreases glucose consumption and glucose uptake, and reduces the expression of GLUT4 and PI3K p85β.83 |
| miR-33 family | Controls AKT phosphorylation and insulin signaling pathways in the liver;73 increases IRS-2, PI3K levels, and AKT phosphorylation in liver, skeletal muscle, and subcutaneous adipocytes of IRS-1-deficient mice.74 |
| miR-96 | Decreases IRS-1 protein expression.64 |
| miR-103 | Affects insulin sensitivity.55,56 |
| miR-106b | Its overexpression decreases glucose consumption and glucose uptake, and reduces the expression of GLUT4 and PI3K p85β.83 |
| miR-107 | Affects insulin sensitivity.55,56 |
| miR-126 | Decreases IRS-1 protein expression,65 and controls PI3K-p85β subunit expression.68 |
| miR-128 family | Negatively regulates insulin signaling;60, 61, 62 its overexpression down-regulates INSR expression in adipocytes;62 its inhibition mitigates myocardial insulin resistance.63 |
| miR-133 family | Reduces the insulin-stimulated glucose uptake in adipose tissue and skeletal muscle.81,82 |
| miR-135 family | Reduces the expression of IRS-2;65 its overexpression reduces the expression of INSR and the glucose uptake in skeletal muscle.66 |
| miR-143 | Affects insulin sensitivity;53 its overexpression impairs insulin-stimulated AKT activation and glucose homeostasis;53,71 promotes GLUT4 protein expression in adipocytes.86 |
| miR-144 | Decreases IRS-1 protein expression.65 |
| miR-181a | Affects insulin sensitivity.54 |
| miR-194 | Its knockdown induces an increase in basal and insulin-stimulated glucose uptake and glycogen synthesis, mediated by increased insulin-induced AKT and GSK3β phosphorylation in skeletal muscle.85 |
| miR-199a-3p | Increases PI3K/AKT signaling pathway leading to reduced apoptosis and increased proliferation in endothelial cells.80 |
| miR-200a-5p | Promotes glucose uptake in the cardiomyocytes.89 |
| miR-205-5p | Increases AKT phosphorylation in hepatocytes and then decreases hepatocyte glucose production.75 |
| miR-223 | Promotes glucose uptake in the cardiomyocytes;88 its overexpression is associated with a reduction in GLUT4 protein content and insulin-stimulated glucose uptake in human differentiated adipocytes.90 |
| miR-320 | Boosts insulin resistance in adipose tissue and endothelial cell by regulating IGF-1/IGF-1R expression and inulin pathway,58 and controls PI3K-p85α subunit expression in the adipose tissue and skeletal muscle.58 |
| miR-338-3p | Its reduction induces insulin resistance.76 |
| miR-383 | Its reduction stimulates AKT signaling through the regulation of IGF-1R.59 |
| miR-384-5p | Controls PI3K-p110δ subunit expression.69 |
| miR-449a | Promotes insulin-stimulated PI3K and AKT phosphorylation in skeletal muscle.79 |
| miR-451 | Negatively regulates hepatic gluconeogenesis in an AKT-dependent way.78 |
| miR-499-5p | Its reduction induces insulin resistance.77 |
| miR-802 | Its reduction improves glucose tolerance and insulin action.57 |
AKT, protein kinase B; GLUT4, glucose transporter 4; GSK3β, glycogen synthase kinase-3β; IGF-1, insulin-like growth factor-1; IGF-1R, IGF-1 receptor; INSR, insulin receptor; IRS, INSR substrates; miRNA, microRNA.
β-Cell failure
Recent studies have shown that certain miRNAs can regulate the expression of genes that are important for the maintenance of pancreatic β-cell homeostasis.14 Accordingly, miRNAs could exert important physiological roles in compensatory response of β-cells to stressful stimuli, regulation of insulin secretion mechanisms, maintenance of β-cell mass, and maturation process of β-cells.15,16
Compensatory response of β-cells to stressful stimuli
MiR-375 is the most highly expressed miRNA in pancreatic islets (10% of β-cell miRNAs are miR-375).17,18 It regulates β-cell adaptation to metabolic stress and insulin resistance;19 therefore, mice with global miR-375 knockout show progressive hyperglycemia with lower numbers of β-cells and impaired compensatory β-cell proliferation.20,21 Similarly, miR-223 and miR-455 expression is increased in islets of diabetic and obese mice, respectively, and drive the compensatory β-cell expansion and the maintenance of functional β-cell mass during metabolic stress.22,23 In addition, the miR-17-92 cluster regulates β-cell restoration and adaptation after streptozotocin treatment in mice.24 MiR-216a (a conserved, pancreas-specific miRNA) also plays important roles in pancreatic islet compensatory response, since its knockout in mice leads to a reduction in islet size, β-cell mass, and insulin levels.25 MiR-34a and miR-146 levels are increased in islets of diabetic db/db mice or in the MIN6 β-cell line exposed to palmitate and this increase results in sensitization to apoptosis and impaired nutrient-induced insulin secretion.26 Likewise, miR-25 and miR-92b expression is up-regulated by palmitate and pro-inflammatory cytokines, leading to inhibition of insulin biosynthesis and increased β-cell apoptosis.27 Similarly, the miR-153 expression level is increased in interleukin-1β-treated β-cells and induces insulin secretion defects.28 Furthermore, miR-320a is increased in the pancreatic β-cells from high-fat-diet (HFD)-treated mice and its overexpression in cultured β-cells initiates pancreatic islet dysfunction by increasing the reactive oxygen species level, inhibiting proliferation, and inducing apoptosis.29 On the contrary, miR-383 expression is reduced in T2D patients and its overexpression reversed cell apoptosis and oxidative stress in β-cell lines exposed to high glucose levels, and ameliorated hyperglycemia and pancreatic apoptosis in HFD-induced diabetic mice.30 Similarly, miR-299-5p expression is reduced by glucolipotoxicity in healthy human islets and this inhibition promotes β-cell dysfunction and loss.31
Regulation of insulin biosynthesis and secretion
Several miRNAs are able to regulate insulin biosynthesis and glucose-stimulated insulin secretion (GSIS). In particular, it has been demonstrated that miR-30d, miR-15a, and miR-375 play a constitutive role in insulin gene expression,32 insulin synthesis,33 and GSIS,33 respectively. On the contrary, miR-7, miR-9, miR-96, and miR-200 family (which consists of miR-200a/141 and miR-200b/miR-200c/miR-429 clusters, and account for up to two-thirds of all miRNAs in β-cells) appear to constitutively repress β-cell function, since their deletion or overexpression promotes or suppresses, respectively, insulin secretion.15,33, 34, 35, 36, 37, 38 Likewise, miR-24, miR-26, miR-148, and miR-182 reduce insulin promoter activity and insulin mRNA levels and consequently insulin content.33,39 In addition, miR-335, miR-152, and miR-130a/b are increased in pancreatic islets isolated from Goto-Kakizaki rats, a model of T2D with β-cell dysfunction,40 and their expression is negatively correlated with insulin exocytosis.15,41 Similarly, miR-187, miR-124a, and miR-770-5p expression is increased in human T2D islets and negatively correlated with β-cell function.42, 43, 44 Similarly, the transient overexpression of miR-29a negatively affects GSIS in MIN6 cells.45 On the contrary, miR-125b-5p is down-regulated in β-cells of diabetic mice and this results in reduced insulin sensitivity and deregulated β-cell function.46 Interestingly, it has been demonstrated that miR-204 is able to reduce the expression of MafA, a key transcription factor controlling insulin expression, thus blocking insulin production.47 MafA expression is also indirectly and positively influenced by miR-132.48 Finally, it has been recently demonstrated that miR-7, miR-132, and miR-212 expression in β-cells is regulated by glucagon-like peptide-1 (GLP-1) and they play a functional role in the regulation of GLP-1-mediated insulin release.49,50
Regulation of β-cell mass
It has been demonstrated that several miRNAs are able to constitutively control β-cell mass. It is the case of miR-132 that controls pancreatic β-cell proliferation and survival through phosphatase and tensin homolog (PTEN)/protein kinase B (AKT)/forkhead box O3 (FOXO3) signaling.51 On the contrary, miR-7, miR-9, miR-96, and miR-200 family deletion or overexpression promotes or suppresses, respectively, β-cell survival.15,33, 34, 35, 36, 37, 38 Similarly, the transient overexpression of miR-29a negatively affects MIN6 cell proliferation.45 Likewise, miR-124a and miR-770-5p expression is increased in patients with T2D and promotes β-cell death.43,44 In addition, many miRNAs can also affect the maturity state of β-cells: for example, the miR-29 family, miR-204, and miR-129 are up-regulated, whereas the miR-17-92 cluster, miR-181b, and miR-215 are suppressed, in young, immature, proliferating islets compared to adult, mature, non-proliferating islets.15 In particular, the miR-17-92 cluster and miR-181b are able to regulate both β-cell replication and GSIS.24 It has been demonstrated that miR-204 activates the unfolded protein response signaling, promoting endoplasmic reticulum stress and apoptosis in β-cell lines and human islets.52
Insulin resistance
MiRNAs also participate in the mechanisms underlying insulin resistance in peripheral tissues such as the liver, adipose tissue, skeletal muscle, and others.14 Specifically, miR-143,53 miR-181a,54 miR-103, and miR-10755,56 have all been shown to affect insulin sensitivity, and more recently, miR-802 has been shown to increase with obesity and its reduction improves glucose tolerance and insulin action.57
Several studies have demonstrated that miRNAs can affect each step of insulin and insulin-like growth factor (IGF)-(1/2) signaling: expression of insulin receptor (INSR), IGF receptor (IGF-R), and INSR substrates (IRS), intracellular pathways, glucose transporter 4 (GLUT4) translocation, and glucose uptake.
INSR, IGF-R, and IRS expression
MiR-320 plays an important role in boosting insulin resistance in adipose tissue and endothelial cells by regulating IGF-1/IGF-1R expression and insulin pathways.58 Similarly, the reduction of miR-383 expression stimulates AKT signaling through the regulation of IGF-1R.59 The miR-128a/b negatively regulates intracellular mediators that are associated with the insulin signaling protein cascades including INSR and IRS-1 at both mRNA and protein levels.60,61 Accordingly, obesity promotes the activation of miR-128 that reduces INSR expression in adipocytes.62 Interestingly, miR-128-3p is overexpressed in the myocardium of mice manifesting severe cardiac dysfunction, together with IRS-1 degradation and insulin resistance, while its inhibition mitigates myocardial insulin resistance.63 Likewise, miR-96, miR-126, and miR-144 are supposed to target IRS-1 and their expression causes a significant decrease in IRS-1 protein expression and consequent damage to insulin signaling.64,65 On the other hand, miR-135a significantly reduces the expression of IRS-2 at both mRNA and protein levels.65 Accordingly, murine skeletal muscle C2C12 cells overexpressing miR-135 show reduced expression of INSR and glucose uptake, similar to the insulin-resistant cell lines.66
Phosphatidylinositol 3-kinase/AKT signaling
Several miRNAs can regulate phosphatidylinositol 3-kinase (PI3K) activity. In particular, miR-320, miR-128a/b, and miR-29 control PI3K-p85α subunit expression in the adipose tissue and skeletal muscle,58,60,67 while miR-126 regulates the subunit p85β,68 and miR-384-5p regulates the catalytic subunit p110δ.69 MiR-19a also regulates PI3K in endothelial cells under shear stress.70 Furthermore, the activation of AKT can be modulated by miR-143: the overexpression of miR-143 in mice impairs insulin-stimulated AKT activation and glucose homeostasis.53,71 In addition, miR-143 expression is increased in mouse models of obesity.53,71
In the adipose tissue cell-line 3T3-L1, the overexpression of miR-29 blunts insulin-stimulated AKT activation and glucose uptake.72 Similarly, the miR-33 family (miR-33a and miR-33b) controls AKT phosphorylation and insulin signaling pathways in the liver.73 In addition, IRS-1-deficient mice show reduced levels of miR-33, which compensates for insulin resistance by increasing IRS-2, PI3K levels, and AKT phosphorylation in the liver, skeletal muscle, and subcutaneous adipocytes.74 Interestingly, miR-205-5p gain of function increases AKT phosphorylation in hepatocytes and then decreases hepatocyte glucose production and lowers glucose levels in mice.75 Furthermore, in the liver of the db/db, HFD-fed, and tumor necrosis factor-α-treated mice, miR-338-3p and miR-499-5p levels are reduced and this induces insulin resistance, as indicated by altered glucose tolerance and insulin tolerance, and impairs AKT-mediated glycogen synthesis.76,77 On the contrary, miR-451 is elevated in the liver of diabetic mice and negatively regulates hepatic gluconeogenesis in an AKT-dependent way, thus alleviating hyperglycemia.78 Moreover, in C2C12 skeletal muscle cells, miR-449a promotes insulin-mediated PI3K/AKT signaling.79 MiR-199a-3p levels are reduced in peripheral blood from patients with T2D compared to healthy subjects and this may be associated with vascular endothelial cell injury, since transfection with miR-199a-3p mimics activates the PI3K/AKT signaling pathway in human umbilical vein endothelial cells, leading to reduced apoptosis and increased proliferation.80
GLUT4 translocation and glucose uptake
Finally, several miRNAs can affect glucose uptake by modulating GLUT4 translocation in adipose tissue and skeletal muscle.33 For instance, the miR-133 family (miR-133a-1, miR-133a-2, and miR-133b) reduces insulin-stimulated glucose uptake in both adipose tissue and skeletal muscle.81,82 Similarly, the overexpression of miR-106b, miR-27a, and miR-30d in L6 cells decreases glucose consumption and glucose uptake and reduces the expression of GLUT4 and PI3K p85β.83 Likewise, overexpression of miR-29a causes a decrease in levels of GLUT4 which partially induces a decrease in insulin-dependent glucose uptake in C2C12 cells.84 Also, miR-194 knockdown in L6 skeletal muscle cells induces an increase in basal and insulin-stimulated glucose uptake and glycogen synthesis, mediated by increased insulin-induced AKT and glycogen synthase kinase-3β phosphorylation.85
Interestingly, miR-194 expression is significantly reduced by 25%-50% in humans with pre-DM and established DM, as an adaptive response to facilitate tissue glucose uptake and metabolism in the face of insulin resistance.85 On the other hand, miR-143 promotes GLUT4 protein expression in adipocytes.86 In insulin-resistant adipocytes, the overexpression of miR-21 improves the insulin-induced AKT phosphorylation and GLUT4 translocation.87 Additionally, GLUT4 is also targeted by miR-223 and miR-200a-5p that promote glucose uptake in the cardiomyocytes.88,89 Differently, overexpression of miR-223 in human differentiated adipocytes is associated with a reduction in GLUT4 protein content and insulin-stimulated glucose uptake.90
Are MiRNAs involved in the pathogenesis of diabetes mellitus also involved in the pathogenesis of cancer?
MiRNAs are known as critical regulators of gene expression and, in cancer, they play a role in oncogenesis, metastasis, and resistance to various therapies. MiRNAs can be classified as oncogenes (oncomiRs), tumor-suppressor genes, pro-metastatic (metastamiRs), and metastasis suppressors.91 The importance of miRNAs in cancer, in general, is highlighted by the observation that half of all miRNA genes are located in cancer-associated genomic regions or fragile sites, which are frequently altered in cancer.92,93 Interestingly, many of the miRNAs involved in the pathogenesis of T2D ("The role of miRNAs in the pathogenesis of type 2 diabetes" section, Tables 1 and 2) also participate in multiple biological processes related to cancer onset and progression. Here we focused on miRNAs recognized as intervening in the main tumorigenic processes, and therefore playing a role in the pathogenesis of multiple types of cancer, while excluding miRNAs that are involved only in specific cancers. The major genes or pathways targeted by these miRNAs are also summarized in Table 3.
Table 3.
Major genes/pathways targeted by selected miRNAs in cancer
| miRNAs | Major genes/pathways targeted in cancer |
|---|---|
| miR-7 | Targets XRCC2 expression and EGFR signaling pathway.95,167 |
| miR-9 | Modulates the PI3K/AKT, JAK/STAT, Notch1, Wnt/β-catenin, Ras, and ERK signaling pathways.98 |
| miR-15a | Targets the oncogenes BCL2, MCL1, CCND1, and Wnt3A.99 |
| miR-17-92 cluster | Targets tumor-suppressive (PTEN, RB2, and TGF-β) and oncogenic (c-myc, Notch, and Sonic Hedgehog) signaling pathways.102,103 |
| miR-21 | Modulates PTEN, PDCD4, RECK, MAPK/ERK, PI3K/AKT, STAT3, and BCL2 gene expression.106 |
| miR-24 | Increases the oncogenes (c-myc, BCL2, HIF1) gene expression and reduces the tumor-suppressor (p21 and p53) protein expression.108 |
| miR-25 | Regulates E2F1 expression.168 |
| miR-26 | Targets caspase-3, caspase-9, and poly (ADP-ribose) polymerase.111 |
| miR-27a | Increases PI3K/AKT/GSK3β, Wnt/β-catenin, Ras/MEK/ERK, and c-myc signaling pathways, while inhibiting TGF-β pathway.112 |
| miR-34 | Activates p53; inhibits vimentin and fibronectin expression; promotes E-cadherin expression; controls Wnt, TGF-β/Smad, and Notch signaling pathways.115, 116, 117 |
| miR-96 | Targets the structural proteins (involved in cell migration and adhesion) and the FOXO transcription factors.119 |
| miR-125b | Suppresses the expression of BCL-2 family genes.169 |
| miR-126 | Reduces the PI3K, K-Ras, VEGF signaling pathways.123 |
| miR-132 | Modulates the PI3K, TGF-β, and Hippo signaling pathways; reduces the expression of oncogenes Ras, AKT, and mTOR.125 |
| miR-135 | Targets AKT and Wnt signaling pathways, and APC, FOXO1, FOXN1, RECK, some matrix metalloproteinases gene expression.126 |
| miR-144 | Modulates the Src/AKT/ERK, Notch, JAK2/STAT3, EGFR, and PTEN/PI3K/AKT signaling pathways.127 |
| miR-153 | Reduces PTEN expression.130 |
| miR-181 | Regulates the MAPK, ERK, Wnt, TGF-β, EGFR/Ras, PI3K/AKT, p53 signaling pathways.132 |
| miR-212 | Modulates the Wnt/β-catenin, Hedgehog, and Hippo/YAP signaling pathways.136 |
| miR-320 | Suppresses E-cadherin level and increases N-cadherin and vimentin expression by directly targeting FOXM1; regulates the Wnt, PI3K/AKT, TGF-β/Smad, CDK6, STAT3, and E2F1 signaling pathways.139 |
| miR-335 | Targets the Hedgehog, the PI3K/AKT/mTOR, and the Hippo/YAP signaling pathways.140 |
| miR-375 | Targets the AEG-1, YAP1, IGF-1R, PDK1, Wnt, NF-κB, Notch, and TGF-β/Smad signaling pathways.141,142 |
| miR-449a | Targets the TAK1, Notch, NF-κB/p65/VEGF, RB-E2F, MAPKs, Wnt/β-catenin, p53, and androgen receptor signaling pathways.143 |
| miR-802 | Modulates the regulation of the Wnt, PI3K/AKT, ERK, and Hedgehog signaling pathways.144 |
AEG-1, astrocyte elevated gene 1; AKT, protein kinase B; APC, adenomatous polyposis coli; BCL2, B-cell lymphoma 2; CCND1, cyclin D1; CDK6, cyclin dependent kinase 6; EGFR, epidermal growth factor receptor; ERK, extracellular receptor kinase; FOX, forkhead box; GSK3β, glycogen synthase kinase-3β; HIF1, hypoxia-inducible factor 1; IGF-1R, insulin-like growth factor 1 receptor; JAK, Janus kinase; MAPK, mitogen-activated protein kinase; MCL1, induced myeloid leukemia cell differentiation protein; miRNA, microRNA; mTOR, mammalian target of rapamycin; NF-κB, nuclear factor-kappa B; PDCD4, programmed cell death protein 4; PDK1, phosphoinositide-dependent kinase-1; PI3K, phosphatidylinositol 3-kinase; PTEN, phosphatase and tensin homolog; Ras, rat sarcoma virus; RB, retinoblastoma-like protein; RECK, reversion-inducing cysteine-rich protein with Kazal motifs; Smad, small mother against decapentaplegic; Src, steroid receptor coactivator; STAT, signal transducer activator of transcription; TAK1, TGF-β activated kinase 1; TGF-β, transforming growth factor-β; VEGF, vascular endothelial growth factor; XRCC2, X-ray repair complementing defective repair gene; YAP, yes-associated protein.
miR-7
MiR-7 is one of the most conserved and oldest miRNAs and is engaged in numerous signaling circuits involved in differentiation, regulation of proliferation, apoptosis, and migration. Its dominant activity is tumor suppression by inhibition of cell proliferation and survival. Indeed, in most tumors, its expression is reduced, and in some cancer types, it acts as an oncomiR (Table 4).94,95 Of note, its depletion affects the expression of various proteins involved in apoptosis, epidermal growth factor receptor signaling pathways, and multidrug resistance.95 In most cases, its depletion limits the effectiveness of anticancer therapies, while its restoration sensitizes cells to the administered drugs. Therefore, miR-7 might be considered a potential adjuvant agent, which can increase the efficiency of standard chemotherapeutics.95
Table 4.
MiRNAs differentially expressed in diabetic and oncological patients, and profoundly involved in the pathogenesis of both type 2 diabetes (T2D) and cancer
| miRNAs | Expression in T2D | Expression in cancer |
|---|---|---|
| hsa-miR-7 | Increased170 | Reduced in breast, lung, ovarian, prostate, gastric, and skin cancers, as well as in glioblastoma, hepatocellular carcinoma, melanoma, and leukemia.94,95 |
| hsa-miR-9 | Increased171 | Aberrant expression in breast, cervical, colorectal, lung, gastric, bladder, and prostate cancers, as well as in glioblastoma, squamous cell carcinoma of skin and oral cavity, hematological malignancies, and uveal melanoma.96,97 |
| hsa-miR-15a | Reduced171, 172, 173 | Reduced in chronic lymphocytic leukemia, colorectal, bladder, and prostate cancers, as well as in human brain glioma, hepatoma, and different types of lymphoma or leukemia.100,101 |
| miR-17-92 cluster (in particular hsa-miR-17, hsa-miR-18a, hsa-miR-19a, hsa-miR-20a) | Further studies required | Increased in lung cancer and human B-cell lymphomas.104,105 |
| hsa-miR-21 | Reduced87,171,173 | Increased in gliomas, breast, and colorectal cancers.106 |
| hsa-miR-24 | Reduced173 | Reduced in breast, colorectal, and non-small-cell lung cancers, as well as in hepatocellular, nasopharyngeal, laryngeal squamous cell, and esophageal squamous cell carcinomas.107 |
| hsa-miR-27a | Increased83,171,174 | Increased in ovarian, prostate, and gastric cancers, colorectal carcinoma, and esophageal tumors.112 |
| hsa-miR-29 | Increased15,45,72,84,171,174 | Reduced in breast, ovary, gastric, colon, lung, pancreatic, prostate, cervix, and oral cancers, as well as in hepatocellular carcinoma, glioblastoma, glioma, melanoma, leukemia, lymphoma, osteosarcoma, rhabdomyosarcoma, and neuroblastoma.113 |
| hsa-miR-34 | Increased26,171,173 | Dysregulated in colorectal, prostate, breast, lung, and liver cancers, as well as in osteosarcoma and hematological neoplasms.115 |
| hsa-miR-106b | Further studies required | Increased in breast, prostate, lung, gastric, and colorectal cancers, as well as in hepatocellular and esophageal squamous cell carcinomas.120 |
| hsa-miR-125b | Reduced46 | Reduced in non-small-cell lung cancer, anaplastic thyroid cancer, bladder, ovarian, breast, gallbladder, colorectal, and endometrioid-endometrial cancers, as well as in hepatocellular and esophageal squamous cell carcinomas, melanoma, osteosarcoma, chondrosarcoma, multiple myeloma, and Ewing’s sarcoma.121 Increased in nasopharyngeal carcinoma, retinoblastoma, glioblastoma, poorly differentiated non-small-cell lung cancer, acute lymphoblastic leukemia, acute myeloid leukemia, and gastric cancer.121 |
| hsa-miR-126 | Reduced173 | Lost in breast, lung, gastric, colorectal, cervix, bladder, oral, and prostate cancers.123,124 |
| hsa-miR-132 | Further studies required | Reduced in hepatocellular carcinoma, osteosarcoma, breast, colorectal, gastric, lung, prostate, pancreatic, and ovarian cancers.125 |
| hsa-miR-135 | Increased66 | Reduced in prostate, renal, gallbladder, and nasopharyngeal cancers, as well as in glioma, impacting the enhancement of cell proliferation and aggressive behavior.126 Increased in bladder, oral, colorectal, and liver cancers.126 Dual role in in breast, gastric, lung, and pancreatic cancers, as well as in head and neck squamous cell carcinomas.126 |
| hsa-miR-144 | Further studies required | Reduced in lung, gastric, colorectal, thyroid, cervical, ovarian, prostate, bladder, esophageal, pancreatic, and breast cancers, as well as in osteosarcoma, glioblastoma, cholangiocarcinoma, hepatocellular, renal cell, head and neck squamous cell, and oral squamous cell carcinomas, leukemia, and lymphomas.127,128 |
| hsa-miR-181 | Further studies required | Aberrant expression in different solid tumors and hematological malignancies.132 |
| hsa-miR-205 | Reduced75 | Reduced in breast, liver, prostate, skin, renal, and colorectal cancers, as well as in glioblastoma.137 Increased in cervical, ovarian, endometrial, and lung cancers.137 |
| hsa-miR-375 | Further studies required | Reduced in hepatocellular carcinoma, gastric, esophagus, head and neck, and other cancers.142 |
| hsa-miR-449a | Reduced79 | Reduced in gastric, lung, breast, bladder, liver, cancer, prostate, ovarian, and endometrial cancers, as well as in glioma, neuroblastoma, and retinoblastoma.143 |
| hsa-miR-802 | Increased57 | Reduced in gastric, colorectal, breast, cervical, and epithelial ovarian cancers, as well as in melanoma and tongue, oral, esophageal carcinoma, and laryngeal squamous cell carcinomas.144 Increased in hepatocellular carcinoma, bladder urothelial cancer, osteosarcoma, and cholesteatoma tissue cells.144 |
miRNA, microRNA.
miR-9
MiR-9 plays a prominent role in tumorigenesis and its aberrant expression has been observed in many human cancer types (Table 4).96,97 Of note, the role of miR-9 in cancer is dependent on cancer type, so miR-9 serves as a tumor-suppressor miRNA in some cancers and has an oncogenic role in others.98 MiR-9 controls different biological processes, such as self-renewal, proliferation, and differentiation, and has been shown to play important regulatory roles in cancer biology regulating processes such as tumor initiation (apoptosis/proliferation), progression (angiogenesis/metastasization), and chemosensitivity.97,98 Specifically, miR-9 expression affects many biochemical pathways commonly deregulated in human cancers (Table 3).
miR-15a
MiR-15a is known to act as a tumor suppressor. Its expression inhibits cell proliferation, promotes apoptosis of cancer cells, and suppresses tumorigenicity [tumor survival, proliferation, migration, angiogenesis, invasion, and epithelial–mesenchymal transition (EMT)] both in vitro and in vivo, by targeting multiple oncogenes (Table 3).99 MiR-15a is regulated by some important oncogenes or tumor suppressors (e.g. c-myc, p53), contributing to cancer initiation and insensitivity to chemotherapy.100 MiR-15a expression is frequently reduced in many types of cancer (Table 4), and restoring its level has helped to enhance apoptosis, delay cell cycle of the cancer cells, and enhance chemosensitivity of tumor.100,101
miR-17-92 cluster (miR-17, miR-18a, miR-19a, miR-20a)
Overexpression of the miR-17-92 cluster is a key oncogenic event in various cancer types, as its members target tumor-suppressive proteins and pathways (Table 3).102,103 In particular, the miR-17-92 cluster is remarkably increased in lung cancer, especially in the most aggressive form, small-cell lung cancer, and also enhances lung cancer cell growth.104 Elevated expression of the miR-17-92 cluster was also found in several human B-cell lymphomas.105 Of note, several studies have revealed the influence of oncogenic signaling pathways (Table 3) in the transcriptional regulation of miR-17-92 cluster in cancer.102
miR-21
MiR-21 is one of the first oncomiRs found to be up-regulated in a variety of cancers (Table 4).106 Some of its target genes are associated with cell proliferation, migration, invasion, and apoptosis (Table 3).106 As a result, miR-21 has been proposed as a plausible diagnostic and prognostic biomarker, as well as a therapeutic target for several types of cancer.
miR-24
MiR-24 targets and regulates numerous genes in various cancer types (Table 4).107 In particular, it enhances the expression of several oncogenes (Table 3), while reducing the expression of several tumor-suppressor proteins (Table 3).108 MiR-24 also regulates cell cycle and is associated with numerous cancer hallmarks such as apoptosis, proliferation, metastasis, invasion, angiogenesis, autophagy, and drug resistance.108
miR-25 and miR-26
Similarly, miR-25 altered expression has been reported in many human malignant tumors.109 It seems capable of regulating, via different targets, some of the most important pathways, including proliferation, invasion, differentiation, apoptosis, and autophagy.109 Interestingly, it functions as both an oncogene and in some cases a tumor suppressor, depending on the type of cancer it is involved in.109 Furthermore, the aberrant expression of miR-26 exists in various kinds of tumor tissues, where it participates in the regulation of tumor cell metabolism, proliferation, differentiation, apoptosis or autophagy, invasion, and metastasis through targeting gene expression at the post-transcriptional level.110 It functions as either tumor suppressor or oncogene in different types of tumors.110 For instance, miR-26b has been repeatedly found to decrease in colorectal specimens and cancer cell lines, whereas its overexpression is linked to the inhibition of colorectal cancer growth that is probably subsequent to enhanced levels of key apoptosis-related proteins (Table 3).111
miR-27a
MiR-27a is able to promote tumor proliferation and invasion (Table 3).112 It has also been reported to be associated with chemotherapy resistance in several cancers, although the mechanisms remain unclear.112 Accordingly, its expression is increased in several tumors (Table 4).112
miR-29 family
The miR-29 family is involved in important biological functions, such as cell growth, programmed death, differentiation, and proliferation, thereby playing a direct function in cancer progression.113 Indeed, its expression is reduced in most cancers, which may be a sign of its role in inhibiting cancerous cells (Table 4).113 MiR-29 intervenes in key processes, such as apoptosis, proliferation, metastasization, angiogenesis, EMT, and immunomodulation.114
miR-34
MiR-34 has been reported to be dysregulated in various human cancers (Table 4) and, at the same time, is also the first miRNA that has proven to be directly regulated by the tumor suppressor p53115 and to induce p53 activation.116 Thus, the miR-34 family is known to inhibit tumorigenesis. A large quantity of experimental data showed that miR-34 could influence EMT and then cell metastasis and invasion (Table 3).115,117 In addition, miR-34a also appears to be involved in the regulation of apoptosis in colorectal cancer cell lines.118
miR-96
MiR-96 regulates cell motility, tumor initiation, progression, and invasion as an oncogene or tumor suppressor, mainly by targeting the structural proteins (involved in cell migration and adhesion) and the FOXO transcription factors, which are mainly considered tumor suppressors during tumor progression, regulating the expression of genes involved in cell proliferation, apoptosis, differentiation, and cell cycle regulation.119
miR-106b
MiR-106b overexpression has been identified in multiple tumor types (Table 4) and controls cell proliferation, migration, invasion, and metastasization.120 Moreover, miR-106b overexpression is associated with aggressive clinical and pathological features (Table 3).
miR-125b
MiR-125b expression has been found to be both increased as an oncogene in various cancers and reduced as a tumor suppressor in others (Table 4).121 At present, many genes have been confirmed as targets of miR-125b, covering a variety of biological signaling pathways and affecting the formation of many malignant phenotypes, such as proliferation, differentiation, migration, apoptosis, cell cycle, and drug resistance in different cancers.121,122
miR-126
Several studies have revealed the role of miR-126 as suppressor of tumor formation in various types of cancer, impairing cancer progression through the down-regulation of signaling pathways that control tumor cell proliferation, migration, invasion, survival, and especially angiogenesis (Table 3).123 Accordingly, miR-126 was reported to be lost in various human cancers (Table 4).123,124
miR-132
Decreases in miR-132 level in various types of malignancies (Table 4) have been reported, indicating its function as a tumor suppressor.125 MiR-132 is involved in cell proliferation, migration, and invasion, affecting cell cycle pathways, or reducing oncogene expression (Table 3).125
miR-135
MiR-135 is involved in the pathoetiology of several neoplastic conditions, with both tumor-suppressive and oncogenic roles.126 Indeed, its expression is reduced or increased depending on the type of tumor in which it is involved (Table 4).126 Studies in breast, gastric, lung, and pancreatic cancers, as well as head and neck squamous cell carcinomas, have reported dual roles for miR-135126 (Table 3).
miR-144
MiR-144 has been identified as a tumor suppressor and its expression is reduced in many types of solid and hematological malignancies (Table 4).127,128 Increasing evidence supports a crucial role for miR-144 in modulating physiopathologic processes, such as proliferation, apoptosis, invasion, migration, and angiogenesis in different tumor cells (Table 3).127 Besides these functions, miR-144 can also affect drug sensitivity, cancer treatment, and patient prognosis.127
miR-148/-152 family
The expression pattern of miR-148/-152 family members has been found to be reduced in various cancer cells, indicating a tumor-suppressive role for these miRNAs, regulating various pathways including malignant transformation and tumor initiation, growth, angiogenesis, migration, and invasion.129
miR-153
Previous studies have indicated that miR-153 functions as a tumor suppressor and as an oncogenic regulator. It is implicated in cancer pathophysiological processes, including proliferation, apoptosis, invasion, EMT and metastasis, angiogenesis, and chemo/radiotherapy resistance.130,131 In this line, miR-153 suppresses tumor growth in epithelial cancer, glioblastoma, and leukemia. In contrast, miR-153 stimulates cell proliferation in prostate cancer cells.130
miR-181
The miR-181 family regulates many relevant biological processes such as cell proliferation, apoptosis, autophagy, angiogenesis, EMT, mitochondrial function, and immune response. Importantly, several studies have shown aberrant expression of these miRNAs in different solid tumors and hematological malignancies, where they act either as tumor suppressors or oncomiRs,132 regulating several cancer-related pathways (Table 3).132
miR-200 family
The miR-200 family contributes to maintaining the epithelial phenotype by repressing the expression of factors that favor the process of EMT, a key hallmark of oncogenic transformation.133 It also promotes resistance to chemotherapeutic drugs as well as radiotherapy during anticancer therapy.134
miR-204 and miR-212
MiR-204 has a dual function as a tumor-suppressive gene and/or an oncomiR in different cancers, by promoting apoptosis, conferring resistance of cancer cells to chemotherapy, and suppressing cancer stem cell self-renewal and EMT.135 Similarly, miR-212 serves as an oncogene or a tumor suppressor by influencing different targets or pathways during the development and progression of cancer136 (Table 3).
miR-205
MiR-205 is a highly conserved miRNA that can act as both an oncomiR and a tumor suppressor, although most reports confirm its emerging role as a tumor suppressor in many cancers.137 Its expression can be both augmented and reduced in cancer (Table 4).137 Accordingly, in specific cell types, miR-205 facilitates tumor initiation and proliferation acting as an oncogene; in others, it inhibits cell proliferation, invasion, and EMT, thus playing a tumor-suppressive role.138 Interestingly, restoration of miR-205 makes cells more sensitive to drug treatments and mitigates drug resistance.137
miR-320
The miR-320 family is one of many tumor-suppressor families which has been demonstrated to be related to the repression of EMT and metastasization, cell proliferation, apoptosis, and drug resistance (Table 3).139
miR-335
The expression level of miR-335 in tissues and cancer cells varies according to cancer type. It may serve as an oncogene or a tumor suppressor by regulating different targets or pathways in tumor initiation, development, and metastasis (Table 3). Furthermore, miR-335 also influences tumor microenvironment and drug sensitivity.140
miR-375
MiR-375 is known to be involved in tumor cell proliferation, migration, and drug resistance. Previous studies have shown that miR-375 affects EMT of human tumor cells via some key transcription factors and signaling pathways (Table 3) and is vital for the development of cancer.141 MiR-375 has been found to be significantly reduced in multiple types of cancer (Table 4), and suppresses core hallmarks of cancer by targeting several important oncogenes (Table 3).142 Accordingly, reduced expression of miR-375 in tissue or circulation may indicate the presence of a neoplasia as well as a poor prognosis of many malignant cancers.142
miR-449a
MiR-449a has been reported to inhibit tumor growth, invasion, and metastasis, and to promote apoptosis and differentiation (Table 3).143 This miRNA has a low expression level in several cancer cell lines and tumor patients (Table 4).143
miR-802
Lastly, current studies have found that miR-802 can target and regulate genes in different tumors (Table 3).144 MiR-802 is abnormally expressed in many tumors (Table 4).144
Other miRNAs
Other miRNAs involved in the processes of tumor initiation, progression, and invasion are miR-107,145 miR-187,146 miR-223,147 miR-338,148 miR-383,149 and miR-451.150
MiRNAs in type 2 diabetes and cancer: common pathways
With T2D and cancer independently representing two leading causes of death, the association between these two conditions further aggravates the socioeconomic burden on global public health. Beyond the epidemiological basis, the underlying molecular mechanisms mediating DM–cancer association are not yet fully understood. In this context, the role of specific miRNAs in both T2D and various cancer types individually, and the overlap between disease-specific regulatory miRNA networks, may be important to identify a conserved miRNA signature common to both diseases. In particular, in the previous sections, we have reported that 40 miRNAs are profoundly involved in the pathogenesis of both T2D and cancer.
To evaluate functions of identified miRNAs, we carried out target prediction and functional annotation using the miRSystem database, which is a web-based system that integrates seven well-known miRNA target gene prediction programs: DIANA-microT web server v5.0 (http://www.microrna.gr/webServer), miRanda, miRBridge, PicTar, PITA, rna22, and TargetScan and two experimentally validated databases, TarBase and miRecords (http://mirsystem.cgm.ntu.edu.tw/).151 The miRSystem can identify the biological functions/pathways regulated by miRNAs based on the enriched functions of their target genes.151 The analysis parameters in the miRSystem were set as follows: hit frequency = 5; observed-to-expected (O/E) ratio = 2; minimal size of genes annotated by the ontology term for testing >50; pathways matched should be from Kyoto Encyclopedia of Genes and Genomes (KEGG), Biocarta, Pathway Interaction Database, and REACTOME databases.151 As shown in Table 5, 28 of the queried miRNAs participate in pathways involved in cancer (KEGG database, 124 targeted mRNAs in the pathway). At the same time, 25 of the queried miRNAs participate in pathways involved in DM (REACTOME database, 38 targeted mRNAs in the pathway). The main pathways potentially involved in both DM and cancer included (Table 5): (i) mitogen-activated protein kinase (MAPK) signaling pathway (KEGG), (ii) INSR signaling cascade (KEGG), (iii) IRS-mediated signaling (REACTOME), (iv) IRS-related events (REACTOME), (v) signaling by INSR (REACTOME), (vi) insulin signaling pathway (KEGG), (vii) mammalian target of rapamycin (mTOR) signaling pathway (KEGG), (viii) PI3K cascade (REACTOME), (ix) phosphatidylinositol signaling system (KEGG), (x) apoptosis (KEGG), and (xi) caspase cascade in apoptosis (Pathway Interaction Database). Twenty-five of the 40 queried miRNAs were found to be involved in both DM and cancer pathways (Figure 1). This supports the hypothesis that these miRNAs should be taken into consideration to envision new pharmacological approaches common to both diseases (see "MiRNAs: a new molecular target for the simultaneous treatment of diabetes mellitus and cancer?" section). A more detailed analysis shows that insulin signaling is the most represented pathway, confirming the hypothesis that hyperinsulinemia and hyperglycemia could be possible shared mechanisms involved in both T2D and cancer development. Accordingly, as stated above, many cancer cells overexpress insulin receptors and therefore are more responsive to the mitogenic effects of insulin,9 as well as hyperglycemia could cause oxidative stress and DNA damage and promote tumor growth.8,9
Table 5.
Selected pathways involved in diabetes mellitus and cancer pathogenesis
| Category | Term | Total gene of the term | Union target in the term | Union miRNAs in the term | Score |
|---|---|---|---|---|---|
| KEGG | PATHWAYS_IN_CANCER | 325 | 124 | 28 | 2.839 |
| KEGG | MAPK_SIGNALING_PATHWAY | 272 | 101 | 27 | 2.452 |
| PATHWAY_INTERACTION_DATABASE | DIRECT_P53_EFFECTORS | 137 | 50 | 25 | 2.155 |
| KEGG | TGF-BETA_SIGNALING_PATHWAY | 84 | 39 | 25 | 1.929 |
| PATHWAY_INTERACTION_DATABASE | E2F_TRANSCRIPTION_FACTOR_NETWORK | 73 | 33 | 26 | 1.908 |
| KEGG | WNT_SIGNALING_PATHWAY | 150 | 63 | 26 | 1.720 |
| KEGG | CELL_CYCLE | 124 | 45 | 26 | 1.615 |
| REACTOME | INSULIN_RECEPTOR_SIGNALING_CASCADE | 86 | 35 | 24 | 1.582 |
| PATHWAY_INTERACTION_DATABASE | REGULATION_OF_RETINOBLASTOMA_PROTEIN | 64 | 32 | 24 | 1.543 |
| REACTOME | IRS-MEDIATED_SIGNALING | 81 | 32 | 24 | 1.459 |
| REACTOME | IRS-RELATED_EVENTS | 81 | 32 | 24 | 1.459 |
| REACTOME | SIGNALING_BY_INSULIN_RECEPTOR | 109 | 37 | 25 | 1.408 |
| KEGG | INSULIN_SIGNALING_PATHWAY | 137 | 45 | 24 | 1.397 |
| KEGG | MTOR_SIGNALING_PATHWAY | 52 | 21 | 23 | 1.390 |
| REACTOME | SIGNALING_BY_INTERLEUKINS | 106 | 40 | 25 | 1.314 |
| REACTOME | CELL_CYCLE_MITOTIC | 330 | 67 | 24 | 1.304 |
| KEGG | P53_SIGNALING_PATHWAY | 68 | 30 | 20 | 1.288 |
| REACTOME | PI3K_CASCADE | 70 | 26 | 23 | 1.275 |
| KEGG | VEGF_SIGNALING_PATHWAY | 76 | 26 | 24 | 1.218 |
| PATHWAY_INTERACTION_DATABASE | INTEGRINS_IN_ANGIOGENESIS | 74 | 27 | 23 | 1.207 |
| PATHWAY_INTERACTION_DATABASE | HIF-1-ALPHA_TRANSCRIPTION_FACTOR_NETWORK | 65 | 27 | 26 | 1.201 |
| PATHWAY_INTERACTION_DATABASE | NOTCH_SIGNALING_PATHWAY | 59 | 25 | 22 | 1.072 |
| PATHWAY_INTERACTION_DATABASE | VALIDATED_TARGETS_OF_C-MYC_TRANSCRIPTIONAL_REPRESSION | 63 | 26 | 24 | 1.014 |
| KEGG | B_CELL_RECEPTOR_SIGNALING_PATHWAY | 75 | 20 | 22 | 0.904 |
| KEGG | PHOSPHATIDYLINOSITOL_SIGNALING_SYSTEM | 78 | 23 | 20 | 0.895 |
| KEGG | JAK-STAT_SIGNALING_PATHWAY | 155 | 32 | 22 | 0.875 |
| REACTOME | INTEGRATION_OF_ENERGY_METABOLISM | 125 | 33 | 25 | 0.872 |
| REACTOME | REGULATION_OF_INSULIN_SECRETION | 98 | 26 | 25 | 0.823 |
| KEGG | APOPTOSIS | 88 | 24 | 22 | 0.794 |
| KEGG | PANCREATIC_SECRETION | 103 | 26 | 23 | 0.786 |
| PATHWAY_INTERACTION_DATABASE | VALIDATED_TARGETS_OF_C-MYC_TRANSCRIPTIONAL_ACTIVATION | 81 | 23 | 24 | 0.764 |
| PATHWAY_INTERACTION_DATABASE | CASPASE_CASCADE_IN_APOPTOSIS | 56 | 16 | 19 | 0.662 |
| REACTOME | DIABETES_PATHWAYS | 229 | 38 | 25 | 0.644 |
| KEGG | INOSITOL_PHOSPHATE_METABOLISM | 57 | 15 | 16 | 0.579 |
| KEGG | CELL_ADHESION_MOLECULES_(CAMS) | 133 | 26 | 20 | 0.557 |
| PATHWAY_INTERACTION_DATABASE | P53_PATHWAY | 58 | 20 | 18 | 0.526 |
| KEGG | HEDGEHOG_SIGNALING_PATHWAY | 56 | 22 | 16 | 0.504 |
KEGG, Kyoto Encyclopedia of Genes and Genomes; miRNA, microRNA.
Figure 1.
We identified the main microRNAs (miRNAs) involved in the pathophysiology of type 2 diabetes, for which a direct role has been shown in mediating pancreatic β-cell dysfunction and death and the onset of peripheral insulin resistance (green circles), the two hallmarks of type 2 diabetes pathogenesis. Among them, we have identified miRNAs also involved in cancer pathogenesis (miRNAs in bold in the pink boxes). We have then carried out target prediction and functional annotation using the miRSystem database, identifying 25 miRNAs (blue circle) that participate in pathways involved in both type 2 diabetes and cancer (olive green box). IRS, insulin receptor substrates; MAPK, mitogen-activated protein kinase; mTOR, mammalian target of rapamycin; PI3K, phosphatidylinositol 3-kinase.
In addition, although the possible use of miRNAs as a biomarker is beyond the scope of the current manuscript, in Table 4 we have also summarized the evidence from literature on how most of these miRNAs are differentially expressed in both diabetic and oncological patients. Interestingly, many miRNAs are either down- or up-regulated in both pathological conditions (Table 4), suggesting their possible use as common biomarkers. Targeted studies are needed to understand whether the expression of these miRNAs in patients with both diseases is further modified compared to patients with diabetes or cancer alone. In addition, it should be noted that various regulatory mechanisms (including genetic polymorphisms, methylation of miRNA promoters, interactions with RNA-binding proteins or other coding/non-coding RNAs) control miRNA expression, activity, and/or bioavailability,152 making it challenging to decipher the precise role of miRNAs as a biomarker in specific pathophysiological context.
MiRNAs: a new molecular target for the simultaneous treatment of diabetes mellitus and cancer?
MiRNA-based therapeutics is a growing field and advances have been made for a variety of disease states, including metabolic disorders and cancer (reviewed in Bajan and Hutvagner153). Dysregulation of miRNA expression in human diseases may provide opportunities for the development of miRNA-based therapies that utilize mimics to increase levels of an miRNA, or miRNA inhibitors (antagomiRs) to down-regulate or block miRNA expression and activity, depending on the type of alteration observed (hypoexpression or hyperexpression).154 Unfortunately, there are still many challenges to the application of miRNA-based therapies, such as in vivo stability, incorrect distribution to the tissue sites of interest, breakdown or overload of endogenous RNA machinery, and in cases of viral vector delivery, toxicity and immunogenicity.155 In addition, one miRNA can be both hyper- and hypoexpressed according to the disease or its stage, making it difficult to be used as a precise target.156 Lastly, the possibility of off-target effects (as yet undiscovered mRNA targets) cannot be excluded and should be carefully considered since they may cause severe unanticipated responses.156 Accordingly, several miRNA-targeted gene therapies have been terminated or withdrawn from clinical trials because of serious adverse effects. For example, the filrst miRNA replacement therapy clinical trial, which tested the efficacy of miR-34 (MRX34) in cancer, was terminated at phase I due to severe immune-related side-effects.157 On the other hand, miRNAs, due to their ability to target numerous transcripts, often in the same biological process, have the potential to regulate an entire signaling pathway known to be misregulated in a pathogenic state, thereby increasing their desirability in clinical applications.153
To date, no miRNA-based antidiabetes and/or anticancer therapies have been approved by the Food and Drug Administration (FDA). Even though numerous miRNAs have been implicated in the pathogenesis of T2D, to the best of our knowledge, no miRNA-based therapy for the treatment of T2D has yet been envisioned or has entered a pre-clinical trial program. Conversely, several potential anticancer therapies have reached phase I and phase II clinical trials (reviewed in He et al.158), although none of them include the 25 miRNAs we identified in the "MiRNAs in type 2 diabetes and cancer: common pathways" section.
In the context of cancer, two studies described the possibility to co-deliver miR-7 and anticancer drugs loaded in nanovehicles, in order to sensitize cancer cells to the chemotherapeutics.159,160 However, this possibility must undergo further investigation. On the other hand, the use of intravenous delivery of anti-miR-17-92 for the treatment of allograft medulloblastoma tumor has been tested in immune-compromised mice, resulting in the blockage of tumor growth.161 Nevertheless, some issues regarding selective anti-miR delivery to cancer cells and its side-effects in animal models still require clarification.102 Although there are no drugs that directly target miR-27, it has been demonstrated that GT-094 (a nitric oxide-releasing nonsteroidal anti-inflammatory drug) in colon cancer cell lines,162 arsenic trioxide (a widely used anticancer drug in leukemia),163 and liraglutide164 in breast cancer cell lines are able to inhibit cancer proliferation through the reduction of miRNA-27a expression. Similarly, it has been demonstrated that several anticancer agents (e.g. 4-phenylbutyric acid, 5-aza-2′-deoxycytidine, olea europaea leaf extract, mifepristone) could induce the apoptosis of cancer cells (including breast cancer and glioblastoma cell lines) through the up-regulation of miR-153.131 Several in vitro studies have shown that the up-regulation of miR-144-3p expression can increase the susceptibility of different cancer cell lines (gastric cancer cells, hepatocellular carcinoma cell lines, glioma and glioblastoma cells, non-small-cell lung cancer, cervical cancer cells, anaplastic thyroid carcinoma cell lines) to chemotherapy and radiotherapy.127 Finally, an miR-383 mimic has been found to suppress proliferation and enhance chemosensitivity in ovarian cancer cells.165
Interestingly, a more detailed analysis of miRNAs in Table 4 shows that several miRNAs are down-regulated (i.e. miR-15a, miR-24, miR-125b, miR-126, miR-205, miR-449a) or up-regulated (i.e. miR-27a, miR-135, miR-802) in both diabetic and oncological patients. Accordingly, their hyperexpression or inhibition, respectively, could represent promising targets to simultaneously counteract T2D and cancer development.
Conclusions and future perspectives
Increasing evidence suggests that patients with DM, particularly T2D, are characterized by an increased risk of developing different types of cancer,4,5 to the point that cancer could be proposed as a new T2D-related complication. On the other hand, cancer may also increase the risk of new cases of DM, mainly caused by the use of anticancer therapies (i.e. corticosteroids, chemotherapy, radiotherapy, immunosuppressive therapies).4,7 Hyperinsulinemia, hyperglycemia, and chronic inflammation typical of T2D could represent possible mechanisms involved in cancer development in diabetic patients.
MiRNAs are a subset of non-coding RNAs, ⁓22 nucleotides in length, that control post-transcriptional regulation of gene expression through both translational repression or mRNA degradation.10 Of note, miRNAs have multiple target genes and alteration of their expression has been reported in multiple diseases, including T2D and cancer.13 Accordingly, specific miRNA-regulated pathways are involved in the pathogenesis of both conditions.
In this review, we have identified the main miRNAs involved in the pathophysiology of T2D ("The role of miRNAs in the pathogenesis of type 2 diabetes" section), for which a direct role has been shown in mediating pancreatic β-cell failure (Table 1) and the onset of peripheral insulin resistance (Table 2), the two hallmarks of T2D pathogenesis (Figure 1). Among them, we have tried to identify miRNAs also involved in cancer pathogenesis, finally focusing our attention on 40 miRNAs profoundly involved in the pathogenesis of both diabetes and cancer (Figure 1). Some of them were also down- or up-regulated in both pathological conditions (Table 4). Interestingly, in cancer, these miRNAs play a role in cell proliferation and survival, autophagy, self-renewal, differentiation, angiogenesis, EMT, invasion, and sensitivity to various anticancer therapies ("Are miRNAs involved in the pathogenesis of diabetes mellitus also involved in the pathogenesis of cancer?" section). We then carried out target prediction and functional annotation using the miRSystem database, identifying 25 miRNAs that participate in pathways involved in both T2D and cancer ("MiRNAs in type 2 diabetes and cancer: common pathways" section, Table 5, and Figure 1). This supports the hypothesis that these miRNAs could represent a possible signature for the early diagnosis of T2D and cancer, and should be taken into consideration to envision new pharmacological approaches common to both diseases. Interestingly, insulin signaling was the most represented pathway, confirming the hypothesis that its impairment should be considered as a possible common mechanism in the pathogenesis of both T2D and cancer.
MiRNA-based drugs are the latest frontier in nucleic acid therapeutics, although there are still many challenges to their clinical application.153, 154, 155, 156 To date, no miRNA-based antidiabetes and/or anticancer therapies have been approved by the FDA. Several potential anticancer therapies have reached phase I and phase II clinical trials (reviewed in He et al.158); however, they do not include the 25 miRNAs we identified in "MiRNAs in type 2 diabetes and cancer: common pathways" section.
In conclusion, we believe that in the near future it may be possible to envision an miRNA-based drug able to treat both T2D and cancer, thus targeting two of the major causes of death of the 21st century, which are closely related to each other.
Acknowledgments
Funding
None declared.
Disclosure
The authors have declared no conflicts of interest.
References
- 1.Cho N.H., Shaw J.E., Karuranga S., et al. IDF diabetes atlas: global estimates of diabetes prevalence for 2017 and projections for 2045. Diabetes Res Clin Pract. 2018;138:271–281. doi: 10.1016/j.diabres.2018.02.023. [DOI] [PubMed] [Google Scholar]
- 2.Saeedi P., Petersohn I., Salpea P., et al. Global and regional diabetes prevalence estimates for 2019 and projections for 2030 and 2045: results from the International Diabetes Federation Diabetes Atlas, 9th edition. Diabetes Res Clin Pract. 2019;157 doi: 10.1016/j.diabres.2019.107843. [DOI] [PubMed] [Google Scholar]
- 3.International Diabetes Federation (IDF). IDF Diabetes Atlas. 9th ed; 2019. https://www.diabetesatlas.org/en/ Available at.
- 4.Chowdhury T.A., Jacob P. Challenges in the management of people with diabetes and cancer. Diabet Med. 2019;36(7):795–802. doi: 10.1111/dme.13919. [DOI] [PubMed] [Google Scholar]
- 5.Hershey D.S. Importance of glycemic control in cancer patients with diabetes: treatment through end of life. Asia Pac J Oncol Nurs. 2017;4(4):313. doi: 10.4103/apjon.apjon_40_17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Natalicchio A., Faggiano A., Zatelli M.C., et al. Metabolic disorders and gastroenteropancreatic-neuroendocrine tumors (GEP-NETs): how do they influence each other? An Italian Association of Medical Oncology (AIOM)/Italian Association of Medical Diabetologists (AMD)/Italian Society of Endocrinology (SIE)/Italian Society of Pharmacology (SIF) multidisciplinary consensus position paper. Crit Rev Oncol Hematol. 2022;169 doi: 10.1016/j.critrevonc.2021.103572. [DOI] [PubMed] [Google Scholar]
- 7.Hwangbo Y., Kang D., Kang M., et al. Incidence of diabetes after cancer development: a Korean national cohort study. JAMA Oncol. 2018;4(8):1099–1105. doi: 10.1001/jamaoncol.2018.1684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Giovannucci E., Harlan D.M., Archer M.C., et al. Diabetes and cancer: a consensus report. Diabetes Care. 2010;33:1674–1685. doi: 10.2337/dc10-0666. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Cignarelli A., Genchi V.A., Caruso I., et al. Diabetes and cancer: pathophysiological fundamentals of a ‘dangerous affair.’. Diabetes Res Clin Pract. 2018;143:378–388. doi: 10.1016/j.diabres.2018.04.002. [DOI] [PubMed] [Google Scholar]
- 10.Cai Y., Yu X., Hu S., Yu J. A brief review on the mechanisms of miRNA regulation. Genomics Proteomics Bioinformatics. 2009;7(4):147–154. doi: 10.1016/S1672-0229(08)60044-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Vasu S., Kumano K., Darden C.M., Rahman I., Lawrence M.C., Naziruddin B. MicroRNA signatures as future biomarkers for diagnosis of diabetes states. Cells. 2019;8(12):1533. doi: 10.3390/cells8121533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Weber J.A., Baxter D.H., Zhang S., et al. The microRNA spectrum in 12 body fluids. Clin Chem. 2010;56(11):1733–1741. doi: 10.1373/clinchem.2010.147405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Rupaimoole R., Slack F.J. MicroRNA therapeutics: towards a new era for the management of cancer and other diseases. Nat Rev Drug Discov. 2017;16(3):203–221. doi: 10.1038/nrd.2016.246. [DOI] [PubMed] [Google Scholar]
- 14.He X., Kuang G., Wu Y., Ou C. Emerging roles of exosomal miRNAs in diabetes mellitus. Clin Transl Med. 2021;11(6):e468. doi: 10.1002/ctm2.468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Vienberg S., Geiger J., Madsen S., Dalgaard L.T. MicroRNAs in metabolism. Acta Physiol (Oxf) 2017;219(2):346–361. doi: 10.1111/apha.12681. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Regazzi R. MicroRNAs as therapeutic targets for the treatment of diabetes mellitus and its complications. Expert Opin Ther Targets. 2018;22(2):153–160. doi: 10.1080/14728222.2018.1420168. [DOI] [PubMed] [Google Scholar]
- 17.Poy M.N., Eliasson L., Krutzfeldt J., et al. A pancreatic islet-specific microRNA regulates insulin secretion. Nature. 2004;432(7014):226–230. doi: 10.1038/nature03076. [DOI] [PubMed] [Google Scholar]
- 18.van de Bunt M., Gaulton K.J., Parts L., et al. The miRNA profile of human pancreatic islets and beta-cells and relationship to type 2 diabetes pathogenesis. PLoS One. 2013;8(1) doi: 10.1371/journal.pone.0055272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Lynn F.C., Skewes-Cox P., Kosaka Y., McManus M.T., Harfe B.D., German M.S. MicroRNA expression is required for pancreatic islet cell genesis in the mouse. Diabetes. 2007;56(12):2938–2945. doi: 10.2337/db07-0175. [DOI] [PubMed] [Google Scholar]
- 20.Poy M.N., Hausser J., Trajkovski M., et al. miR-375 maintains normal pancreatic alpha- and beta-cell mass. Proc Natl Acad Sci U S A. 2009;106(14):5813–5818. doi: 10.1073/pnas.0810550106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Salunkhe V.A., Esguerra J.L.S., Ofori J.K., et al. Modulation of microRNA-375 expression alters voltage-gated Na(+) channel properties and exocytosis in insulin-secreting cells. Acta Physiol (Oxf) 2015;213(4):882–892. doi: 10.1111/apha.12460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Li Y., Deng S., Peng J., et al. MicroRNA-223 is essential for maintaining functional β-cell mass during diabetes through inhibiting both FOXO1 and SOX6 pathways. J Biol Chem. 2019;294(27):10438–10448. doi: 10.1074/jbc.RA119.007755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Hu Q., Mu J., Liu Y., et al. Obesity-induced miR-455 upregulation promotes adaptive pancreatic β-cell proliferation through the CPEB1/CDKN1B pathway. Diabetes. 2022;71(3):394–411. doi: 10.2337/db21-0134. [DOI] [PubMed] [Google Scholar]
- 24.Wan S., Zhang J., Chen X., et al. MicroRNA-17-92 regulates beta-cell restoration after streptozotocin treatment. Front Endocrinol (Lausanne) 2020;11:9. doi: 10.3389/fendo.2020.00009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Erener S., Ellis C.E., Ramzy A., et al. Deletion of pancreas-specific miR-216a reduces beta-cell mass and inhibits pancreatic cancer progression in mice. Cell Rep Med. 2021;2(11) doi: 10.1016/j.xcrm.2021.100434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Lovis P., Roggli E., Laybutt D.R., et al. Alterations in microRNA expression contribute to fatty acid-induced pancreatic beta-cell dysfunction. Diabetes. 2008;57(10):2728–2736. doi: 10.2337/db07-1252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Shen Z., Yu Y., Yang Y., et al. miR-25 and miR-92b regulate insulin biosynthesis and pancreatic β-cell apoptosis. Endocrine. 2022;76:526–535. doi: 10.1007/s12020-022-03016-9. [DOI] [PubMed] [Google Scholar]
- 28.Sun Y., Zhou S., Shi Y., et al. Inhibition of miR-153, an IL-1β-responsive miRNA, prevents beta cell failure and inflammation-associated diabetes. Metabolism. 2020;111 doi: 10.1016/j.metabol.2020.154335. [DOI] [PubMed] [Google Scholar]
- 29.Du H., Yin Z., Zhao Y., et al. miR-320a induces pancreatic β cells dysfunction in diabetes by inhibiting MafF. Mol Ther Nucleic Acids. 2021;26:444–457. doi: 10.1016/j.omtn.2021.08.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Cheng X., Huang Y., Yang P., Bu L. miR-383 ameliorates high glucose-induced β-cells apoptosis and hyperglycemia in high-fat induced diabetic mice. Life Sci. 2020;263 doi: 10.1016/j.lfs.2020.118571. [DOI] [PubMed] [Google Scholar]
- 31.Huang Q., You W., Li Y., et al. Glucolipotoxicity-inhibited miR-299-5p regulates pancreatic β-cell function and survival. Diabetes. 2018;67(11):2280–2292. doi: 10.2337/db18-0223. [DOI] [PubMed] [Google Scholar]
- 32.Tang X., Muniappan L., Tang G., Özcan S. Identification of glucose-regulated miRNAs from pancreatic {beta} cells reveals a role for miR-30d in insulin transcription. RNA. 2009;15(2):287–293. doi: 10.1261/rna.1211209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Chakraborty C., Doss C.G.P., Bandyopadhyay S., Agoramoorthy G. Influence of miRNA in insulin signaling pathway and insulin resistance: micro-molecules with a major role in type-2 diabetes. Wiley Interdiscip Rev RNA. 2014;5(5):697–712. doi: 10.1002/wrna.1240. [DOI] [PubMed] [Google Scholar]
- 34.Latreille M., Hausser J., Stützer I., et al. MicroRNA-7a regulates pancreatic β cell function. J Clin Invest. 2014;124(6):2722–2735. doi: 10.1172/JCI73066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Belgardt B.F., Ahmed K., Spranger M., et al. The microRNA-200 family regulates pancreatic beta cell survival in type 2 diabetes. Nat Med. 2015;21(6):619–627. doi: 10.1038/nm.3862. [DOI] [PubMed] [Google Scholar]
- 36.Eliasson L., Regazzi R. Micro(RNA) management and mismanagement of the islet. J Mol Biol. 2020;432(5):1419–1428. doi: 10.1016/j.jmb.2019.09.017. [DOI] [PubMed] [Google Scholar]
- 37.Plaisance V., Abderrahmani A., Perret-Menoud V., Jacquemin P., Lemaigre F., Regazzi R. MicroRNA-9 controls the expression of granuphilin/Slp4 and the secretory response of insulin-producing cells. J Biol Chem. 2006;281(37):26932–26942. doi: 10.1074/jbc.M601225200. [DOI] [PubMed] [Google Scholar]
- 38.Lovis P., Gattesco S., Regazzi R. Regulation of the expression of components of the exocytotic machinery of insulin-secreting cells by microRNAs. Biol Chem. 2008;389(3):305–312. doi: 10.1515/BC.2008.026. [DOI] [PubMed] [Google Scholar]
- 39.Melkman-Zehavi T., Oren R., Kredo-Russo S., et al. miRNAs control insulin content in pancreatic β-cells via downregulation of transcriptional repressors. EMBO J. 2011;30(5):835–845. doi: 10.1038/emboj.2010.361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Esguerra J.L.S., Bolmeson C., Cilio C.M., Eliasson L. Differential glucose-regulation of microRNAs in pancreatic islets of non-obese type 2 diabetes model Goto-Kakizaki rat. PLoS One. 2011;6(4) doi: 10.1371/journal.pone.0018613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Salunkhe V.A., Ofori J.K., Gandasi N.R., et al. MiR-335 overexpression impairs insulin secretion through defective priming of insulin vesicles. Physiol Rep. 2017;5(21) doi: 10.14814/phy2.13493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Locke J.M., Da Silva Xavier G., Dawe H.R., Rutter G.A., Harries L.W. Increased expression of miR-187 in human islets from individuals with type 2 diabetes is associated with reduced glucose-stimulated insulin secretion. Diabetologia. 2014;57(1):122–128. doi: 10.1007/s00125-013-3089-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Sebastiani G., Po A., Miele E., et al. MicroRNA-124a is hyperexpressed in type 2 diabetic human pancreatic islets and negatively regulates insulin secretion. Acta Diabetol. 2015;52(3):523–530. doi: 10.1007/s00592-014-0675-y. [DOI] [PubMed] [Google Scholar]
- 44.Wang M., Wei J., Ji T., Zang K. miRNA-770-5p expression is upregulated in patients with type 2 diabetes and miRNA-770-5p knockdown protects pancreatic β-cell function via targeting BAG5 expression. Exp Ther Med. 2021;22(1):664. doi: 10.3892/etm.2021.10096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Duan J., Qian X.L., Li J., et al. miR-29a negatively affects glucose-stimulated insulin secretion and MIN6 cell proliferation via Cdc42/β-catenin signaling. Int J Endocrinol. 2019;2019 doi: 10.1155/2019/5219782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Yu C.Y., Yang C.Y., Rui Z.L. MicroRNA-125b-5p improves pancreatic β-cell function through inhibiting JNK signaling pathway by targeting DACT1 in mice with type 2 diabetes mellitus. Life Sci. 2019;224:67–75. doi: 10.1016/j.lfs.2019.01.031. [DOI] [PubMed] [Google Scholar]
- 47.Xu G., Chen J., Jing G., Shalev A. Thioredoxin-interacting protein regulates insulin transcription through microRNA-204. Nat Med. 2013;19(9):1141–1146. doi: 10.1038/nm.3287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Nesca V., Guay C., Jacovetti C., et al. Identification of particular groups of microRNAs that positively or negatively impact on beta cell function in obese models of type 2 diabetes. Diabetologia. 2013;56(10):2203–2212. doi: 10.1007/s00125-013-2993-y. [DOI] [PubMed] [Google Scholar]
- 49.Matarese A., Gambardella J., Lombardi A., Wang X., Santulli G. miR-7 regulates GLP-1-mediated insulin release by targeting β-arrestin 1. Cells. 2020;9(7):1621. doi: 10.3390/cells9071621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Shang J., Li J., Keller M.P., et al. Induction of miR-132 and miR-212 expression by glucagon-like peptide 1 (GLP-1) in rodent and human pancreatic β-cells. Mol Endocrinol. 2015;29(9):1243–1253. doi: 10.1210/me.2014-1335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Mziaut H., Henniger G., Ganss K., et al. MiR-132 controls pancreatic beta cell proliferation and survival through Pten/Akt/Foxo3 signaling. Mol Metab. 2020;31:150–162. doi: 10.1016/j.molmet.2019.11.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Xu G., Chen J., Jing G., Grayson T.B., Shalev A. miR-204 targets PERK and regulates UPR signaling and β-cell apoptosis. Mol Endocrinol. 2016;30(8):917–924. doi: 10.1210/me.2016-1056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Jordan S.D., Krüger M., Willmes D.M., et al. Obesity-induced overexpression of miRNA-143 inhibits insulin-stimulated AKT activation and impairs glucose metabolism. Nat Cell Biol. 2011;13(4):434–448. doi: 10.1038/ncb2211. [DOI] [PubMed] [Google Scholar]
- 54.Zhou B., Li C., Qi W., et al. Downregulation of miR-181a upregulates sirtuin-1 (SIRT1) and improves hepatic insulin sensitivity. Diabetologia. 2012;55(7):2032–2043. doi: 10.1007/s00125-012-2539-8. [DOI] [PubMed] [Google Scholar]
- 55.Trajkovski M., Hausser J., Soutschek J., et al. MicroRNAs 103 and 107 regulate insulin sensitivity. Nature. 2011;474(7353):649–653. doi: 10.1038/nature10112. [DOI] [PubMed] [Google Scholar]
- 56.Wilfred B.R., Wang W.X., Nelson P.T. Energizing miRNA research: a review of the role of miRNAs in lipid metabolism, with a prediction that miR-103/107 regulates human metabolic pathways. Mol Genet Metab. 2007;91(3):209–217. doi: 10.1016/j.ymgme.2007.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Kornfeld J.W., Baitzel C., Könner A.C., et al. Obesity-induced overexpression of miR-802 impairs glucose metabolism through silencing of Hnf1b. Nature. 2013;494(7435):111–115. doi: 10.1038/nature11793. [DOI] [PubMed] [Google Scholar]
- 58.Ling H.Y., Ou H.S., Feng S.D., et al. Changes in microRNA (miR) profile and effects of miR-320 in insulin-resistant 3T3-L1 adipocytes. Clin Exp Pharmacol Physiol. 2009;36(9):e32–e39. doi: 10.1111/j.1440-1681.2009.05207.x. [DOI] [PubMed] [Google Scholar]
- 59.He Z., Cen D., Luo X., et al. Downregulation of miR-383 promotes glioma cell invasion by targeting insulin-like growth factor 1 receptor. Med Oncol. 2013;30(2):557. doi: 10.1007/s12032-013-0557-0. [DOI] [PubMed] [Google Scholar]
- 60.Motohashi N., Alexander M.S., Shimizu-Motohashi Y., Myers J.A., Kawahara G., Kunkel L.M. Regulation of IRS1/Akt insulin signaling by microRNA-128a during myogenesis. J Cell Sci. 2013;126(Pt 12):2678–2691. doi: 10.1242/jcs.119966. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Landgraf P., Rusu M., Sheridan R., et al. A mammalian microRNA expression atlas based on small RNA library sequencing. Cell. 2007;129(7):1401–1414. doi: 10.1016/j.cell.2007.04.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Arcidiacono B., Chiefari E., Foryst-Ludwig A., et al. Obesity-related hypoxia via miR-128 decreases insulin-receptor expression in human and mouse adipose tissue promoting systemic insulin resistance. EBioMedicine. 2020;59 doi: 10.1016/j.ebiom.2020.102912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Ruiz-Velasco A., Zi M., Hille S.S., et al. Targeting mir128-3p alleviates myocardial insulin resistance and prevents ischemia-induced heart failure. Elife. 2020;9 doi: 10.7554/eLife.54298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Jeong H.J., Park S.Y., Yang W.M., Lee W. The induction of miR-96 by mitochondrial dysfunction causes impaired glycogen synthesis through translational repression of IRS-1 in SK-Hep1 cells. Biochem Biophys Res Commun. 2013;434(3):503–508. doi: 10.1016/j.bbrc.2013.03.104. [DOI] [PubMed] [Google Scholar]
- 65.Ryu H.S., Park S.Y., Ma D., Zhang J., Lee W. The induction of microRNA targeting IRS-1 is involved in the development of insulin resistance under conditions of mitochondrial dysfunction in hepatocytes. PLoS One. 2011;6(3) doi: 10.1371/journal.pone.0017343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Honardoost M., Arefan E., Soleimani M., Soudi S., Sarookhani M.R. Development of insulin resistance through induction of miRNA-135 in C2C12 cells. Cell J. 2016;18(3):353–361. doi: 10.22074/cellj.2016.4563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Park S.Y., Lee J.H., Ha M., Nam J.W., Kim V.N. miR-29 miRNAs activate p53 by targeting p85 alpha and CDC42. Nat Struct Mol Biol. 2009;16(1):23–29. doi: 10.1038/nsmb.1533. [DOI] [PubMed] [Google Scholar]
- 68.Guo C., Sah J.F., Beard L., Willson J.K.V., Markowitz S.D., Guda K. The noncoding RNA, miR-126, suppresses the growth of neoplastic cells by targeting phosphatidylinositol 3-kinase signaling and is frequently lost in colon cancers. Genes Chromosomes Cancer. 2008;47(11):939–946. doi: 10.1002/gcc.20596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Bao Y., Lin C., Ren J., Liu J. MicroRNA-384-5p regulates ischemia-induced cardioprotection by targeting phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit delta (PI3K p110δ) Apoptosis. 2013;18(3):260–270. doi: 10.1007/s10495-013-0802-1. [DOI] [PubMed] [Google Scholar]
- 70.He J., Li Y., Yang X., et al. The feedback regulation of PI3K-miR-19a, and MAPK-miR-23b/27b in endothelial cells under shear stress. Molecules. 2012;18(1):1–13. doi: 10.3390/molecules18010001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Takanabe R., Ono K., Abe Y., et al. Up-regulated expression of microRNA-143 in association with obesity in adipose tissue of mice fed high-fat diet. Biochem Biophys Res Commun. 2008;376(4):728–732. doi: 10.1016/j.bbrc.2008.09.050. [DOI] [PubMed] [Google Scholar]
- 72.He A., Zhu L., Gupta N., Chang Y., Fang F. Overexpression of micro ribonucleic acid 29, highly up-regulated in diabetic rats, leads to insulin resistance in 3T3-L1 adipocytes. Mol Endocrinol. 2007;21(11):2785–2794. doi: 10.1210/me.2007-0167. [DOI] [PubMed] [Google Scholar]
- 73.Dávalos A., Goedeke L., Smibert P., et al. miR-33a/b contribute to the regulation of fatty acid metabolism and insulin signaling. Proc Natl Acad Sci U S A. 2011;108(22):9232–9237. doi: 10.1073/pnas.1102281108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Tang C.Y., Man X.F., Guo Y., et al. IRS-2 partially compensates for the insulin signal defects in IRS-1-/- mice mediated by miR-33. Mol Cells. 2017;40(2):123–132. doi: 10.14348/molcells.2017.2228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Langlet F., Tarbier M., Haeusler R.A., et al. MicroRNA-205-5p is a modulator of insulin sensitivity that inhibits FOXO function. Mol Metab. 2018;17:49–60. doi: 10.1016/j.molmet.2018.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Dou L., Wang S., Sun L., et al. Mir-338-3p mediates Tnf-A-induced hepatic insulin resistance by targeting PP4r1 to regulate PP4 expression. Cell Physiol Biochem. 2017;41(6):2419–2431. doi: 10.1159/000475912. [DOI] [PubMed] [Google Scholar]
- 77.Wang L., Zhang N., Pan H.P., Wang Z., Cao Z.Y. MiR-499-5p contributes to hepatic insulin resistance by suppressing PTEN. Cell Physiol Biochem. 2015;36(6):2357–2365. doi: 10.1159/000430198. [DOI] [PubMed] [Google Scholar]
- 78.Zhuo S., Yang M., Zhao Y., et al. MicroRNA-451 negatively regulates hepatic glucose production and glucose homeostasis by targeting glycerol kinase-mediated gluconeogenesis. Diabetes. 2016;65(11):3276–3288. doi: 10.2337/db16-0166. [DOI] [PubMed] [Google Scholar]
- 79.Poddar S., Kesharwani D., Datta M. miR-449a regulates insulin signalling by targeting the Notch ligand, Jag1 in skeletal muscle cells. Cell Commun Signal. 2019;17(1):84. doi: 10.1186/s12964-019-0394-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Wang H., Wang Z., Tang Q. Reduced expression of microRNA-199a-3p is associated with vascular endothelial cell injury induced by type 2 diabetes mellitus. Exp Ther Med. 2018;16(4):3639–3645. doi: 10.3892/etm.2018.6655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Ivey K.N., Muth A., Arnold J., et al. MicroRNA regulation of cell lineages in mouse and human embryonic stem cells. Cell Stem Cell. 2008;2(3):219–229. doi: 10.1016/j.stem.2008.01.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Horie T., Ono K., Nishi H., et al. MicroRNA-133 regulates the expression of GLUT4 by targeting KLF15 and is involved in metabolic control in cardiac myocytes. Biochem Biophys Res Commun. 2009;389(2):315–320. doi: 10.1016/j.bbrc.2009.08.136. [DOI] [PubMed] [Google Scholar]
- 83.Zhou T., Meng X., Che H., et al. Regulation of insulin resistance by multiple MiRNAs via targeting the GLUT4 signalling pathway. Cell Physiol Biochem. 2016;38(5):2063–2078. doi: 10.1159/000445565. [DOI] [PubMed] [Google Scholar]
- 84.Zhou Y., Gu P., Shi W., et al. MicroRNA-29a induces insulin resistance by targeting PPARδ in skeletal muscle cells. Int J Mol Med. 2016;37(4):931–938. doi: 10.3892/ijmm.2016.2499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Latouche C., Natoli A., Reddy-Luthmoodoo M., Heywood S.E., Armitage J.A., Kingwell B.A. MicroRNA-194 modulates glucose metabolism and its skeletal muscle expression is reduced in diabetes. PLoS One. 2016;11(5) doi: 10.1371/journal.pone.0155108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Esau C., Kang X., Peralta E., et al. MicroRNA-143 regulates adipocyte differentiation. J Biol Chem. 2004;279(50):52361–52365. doi: 10.1074/jbc.C400438200. [DOI] [PubMed] [Google Scholar]
- 87.Ling H.Y., Hu B., Hu X.B., et al. MiRNA-21 reverses high glucose and high insulin induced insulin resistance in 3T3-L1 adipocytes through targeting phosphatase and tensin homologue. Exp Clin Endocrinol Diabetes. 2012;120(9):553–559. doi: 10.1055/s-0032-1311644. [DOI] [PubMed] [Google Scholar]
- 88.Lu H., Buchan R.J., Cook S.A. MicroRNA-223 regulates Glut4 expression and cardiomyocyte glucose metabolism. Cardiovasc Res. 2010;86(3):410–420. doi: 10.1093/cvr/cvq010. [DOI] [PubMed] [Google Scholar]
- 89.Yang T., Liu T., Cao C., Xu S. miR-200a-5p augments cardiomyocyte hypertrophy induced by glucose metabolism disorder via the regulation of selenoproteins. J Cell Physiol. 2019;234(4):4095–4103. doi: 10.1002/jcp.27206. [DOI] [PubMed] [Google Scholar]
- 90.Chuang T.Y., Wu H.L., Chen C.C., et al. MicroRNA-223 expression is upregulated in insulin resistant human adipose tissue. J Diabetes Res. 2015;2015 doi: 10.1155/2015/943659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Vishnoi A., Rani S. MiRNA biogenesis and regulation of diseases: an overview. Methods Mol Biol. 2017;1509:1–10. doi: 10.1007/978-1-4939-6524-3_1. [DOI] [PubMed] [Google Scholar]
- 92.Calin G.A., Sevignani C., Dumitru C.D., et al. Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers. Proc Natl Acad Sci U S A. 2004;101(9):2999–3004. doi: 10.1073/pnas.0307323101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Sevignani C., Calin G.A., Siracusa L.D., Croce C.M. Mammalian microRNAs: a small world for fine-tuning gene expression. Mamm Genome. 2006;17(3):189–202. doi: 10.1007/s00335-005-0066-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Korać P., Antica M., Matulić M. MiR-7 in cancer development. Biomedicines. 2021;9(3):325. doi: 10.3390/biomedicines9030325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Gajda E., Grzanka M., Godlewska M., Gawel D. The role of miRNA-7 in the biology of cancer and modulation of drug resistance. Pharmaceuticals (Basel) 2021;14(2):1–24. doi: 10.3390/ph14020149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Khafaei M., Rezaie E., Mohammadi A., et al. miR-9: from function to therapeutic potential in cancer. J Cell Physiol. 2019;234(9):14651–14665. doi: 10.1002/jcp.28210. [DOI] [PubMed] [Google Scholar]
- 97.Nowek K., Wiemer E.A.C., Jongen-Lavrencic M. The versatile nature of miR-9/9∗ in human cancer. Oncotarget. 2018;9(29):20838–20854. doi: 10.18632/oncotarget.24889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Tapeh B.E.G., Mosayyebi B., Mansoori B., Gravand Z.A., Sarvnaz H., Mohammadi A. Emerging molecular functions of microRNA-9: cancer pathology and therapeutic implications. Anticancer Agents Med Chem. 2021;21(17):2304–2314. doi: 10.2174/1871520621666210217094839. [DOI] [PubMed] [Google Scholar]
- 99.Aqeilan R.I., Calin G.A., Croce C.M. miR-15a and miR-16-1 in cancer: discovery, function and future perspectives. Cell Death Differ. 2010;17(2):215–220. doi: 10.1038/cdd.2009.69. [DOI] [PubMed] [Google Scholar]
- 100.Huang E., Liu R., Chu Y. miRNA-15a/16: as tumor suppressors and more. Future Oncol. 2015;11(16):2351–2363. doi: 10.2217/fon.15.101. [DOI] [PubMed] [Google Scholar]
- 101.Calin G.A., Dumitru C.D., Shimizu M., et al. Frequent deletions and down-regulation of micro-RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc Natl Acad Sci U S A. 2002;99(24):15524–15529. doi: 10.1073/pnas.242606799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Fuziwara C.S., Kimura E.T. Insights into regulation of the miR-17-92 cluster of miRNAs in cancer. Front Med (Lausanne) 2015;2:64. doi: 10.3389/fmed.2015.00064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Lewis B.P., Shih I.H., Jones-Rhoades M.W., Bartel D.P., Burge C.B. Prediction of mammalian microRNA targets. Cell. 2003;115(7):787–798. doi: 10.1016/s0092-8674(03)01018-3. [DOI] [PubMed] [Google Scholar]
- 104.Hayashita Y., Osada H., Tatematsu Y., et al. A polycistronic microRNA cluster, miR-17-92, is overexpressed in human lung cancers and enhances cell proliferation. Cancer Res. 2005;65(21):9628–9632. doi: 10.1158/0008-5472.CAN-05-2352. [DOI] [PubMed] [Google Scholar]
- 105.Olive V., Jiang I., He L. mir-17-92, a cluster of miRNAs in the midst of the cancer network. Int J Biochem Cell Biol. 2010;42(8):1348–1354. doi: 10.1016/j.biocel.2010.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Bautista-Sánchez D., Arriaga-Canon C., Pedroza-Torres A., et al. The promising role of miR-21 as a cancer biomarker and its importance in RNA-based therapeutics. Mol Ther Nucleic Acids. 2020;20:409–420. doi: 10.1016/j.omtn.2020.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Wang S., Liu N., Tang Q., Sheng H., Long S., Wu W. MicroRNA-24 in cancer: a double side medal with opposite properties. Front Oncol. 2020;10 doi: 10.3389/fonc.2020.553714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Mukherjee S., Shelar B., Krishna S. Versatile role of miR-24/24-1∗/24-2∗ expression in cancer and other human diseases. Am J Transl Res. 2022;14(1):20–54. [PMC free article] [PubMed] [Google Scholar]
- 109.Caiazza C., Mallardo M. The roles of miR-25 and its targeted genes in development of human cancer. MicroRNA. 2016;5(2):113–119. doi: 10.2174/2211536605666160905093429. [DOI] [PubMed] [Google Scholar]
- 110.Li C., Li Y., Lu Y., et al. miR-26 family and its target genes in tumorigenesis and development. Crit Rev Oncol Hematol. 2021;157 doi: 10.1016/j.critrevonc.2020.103124. [DOI] [PubMed] [Google Scholar]
- 111.Wang B., Lu F.-Y., Shi R.-H., et al. MiR-26b regulates 5-FU-resistance in human colorectal cancer via down-regulation of Pgp. Am J Cancer Res. 2018;8(12):2518–2527. [PMC free article] [PubMed] [Google Scholar]
- 112.Zhang J., Cao Z., Yang G., You L., Zhang T., Zhao Y. MicroRNA-27a (miR-27a) in solid tumors: a review based on mechanisms and clinical observations. Front Oncol. 2019;9:893. doi: 10.3389/fonc.2019.00893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Alizadeh M., Safarzadeh A., Beyranvand F., et al. The potential role of miR-29 in health and cancer diagnosis, prognosis, and therapy. J Cell Physiol. 2019;234(11):19280–19297. doi: 10.1002/jcp.28607. [DOI] [PubMed] [Google Scholar]
- 114.Kwon J.J., Factora T.D., Dey S., Kota J. A systematic review of miR-29 in cancer. Mol Ther Oncolytics. 2018;12:173–194. doi: 10.1016/j.omto.2018.12.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Zhang L., Liao Y., Tang L. MicroRNA-34 family: a potential tumor suppressor and therapeutic candidate in cancer. J Exp Clin Cancer Res. 2019;38(1):53. doi: 10.1186/s13046-019-1059-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Navarro F., Lieberman J. miR-34 and p53: new insights into a complex functional relationship. PLoS One. 2015;10(7) doi: 10.1371/journal.pone.0132767. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Yu F., Jiao Y., Zhu Y., et al. MicroRNA 34c gene down-regulation via DNA methylation promotes self-renewal and epithelial-mesenchymal transition in breast tumor-initiating cells. J Biol Chem. 2012;287(1):465–473. doi: 10.1074/jbc.M111.280768. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Yamakuchi M., Ferlito M., Lowenstein C.J. miR-34a repression of SIRT1 regulates apoptosis. Proc Natl Acad Sci U S A. 2008;105(36):13421–13426. doi: 10.1073/pnas.0801613105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Rahimi H.R., Mojarrad M., Moghbeli M. MicroRNA-96: a therapeutic and diagnostic tumor marker. Iran J Basic Med Sci. 2022;25(1):3–13. doi: 10.22038/IJBMS.2021.59604.13226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Sagar S.K. miR-106b as an emerging therapeutic target in cancer. Genes Dis. 2021;9(4):889–899. doi: 10.1016/j.gendis.2021.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Wang Y., Zeng G., Jiang Y. The emerging roles of miR-125b in cancers. Cancer Manag Res. 2020;12:1079–1088. doi: 10.2147/CMAR.S232388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Wang J.K., Wang Z., Li G. MicroRNA-125 in immunity and cancer. Cancer Lett. 2019;454:134–145. doi: 10.1016/j.canlet.2019.04.015. [DOI] [PubMed] [Google Scholar]
- 123.Meister J., Schmidt M.H.H. miR-126 and miR-126∗: new players in cancer. ScientificWorldJournal. 2010;10:2090–2100. doi: 10.1100/tsw.2010.198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Ebrahimi F., Gopalan V., Smith R.A., Lam A.K.Y. miR-126 in human cancers: clinical roles and current perspectives. Exp Mol Pathol. 2014;96(1):98–107. doi: 10.1016/j.yexmp.2013.12.004. [DOI] [PubMed] [Google Scholar]
- 125.Rafat M., Moraghebi M., Afsa M., Malekzadeh K. The outstanding role of miR-132-3p in carcinogenesis of solid tumors. Hum Cell. 2021;34(4):1051–1065. doi: 10.1007/s13577-021-00544-w. [DOI] [PubMed] [Google Scholar]
- 126.Kadkhoda S., Eslami S., Mahmud Hussen B., Ghafouri-Fard S. A review on the importance of miRNA-135 in human diseases. Front Genet. 2022;13 doi: 10.3389/fgene.2022.973585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Zhou M., Wu Y., Li H., Zha X. MicroRNA-144: a novel biological marker and potential therapeutic target in human solid cancers. J Cancer. 2020;11(22):6716–6726. doi: 10.7150/jca.46293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Kooshkaki O., Rezaei Z., Rahmati M., et al. MiR-144: a new possible therapeutic target and diagnostic/prognostic tool in cancers. Int J Mol Sci. 2020;21(7):2578. doi: 10.3390/ijms21072578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Friedrich M., Pracht K., Mashreghi M.F., Jäck H.M., Radbruch A., Seliger B. The role of the miR-148/-152 family in physiology and disease. Eur J Immunol. 2017;47(12):2026–2038. doi: 10.1002/eji.201747132. [DOI] [PubMed] [Google Scholar]
- 130.Alamdari-Palangi V., Jaberi K.R., Jaberi A.R., et al. The role of miR-153 and related upstream/downstream pathways in cancers: from a potential biomarker to treatment of tumor resistance and a therapeutic target. Med Oncol. 2022;39(5):62. doi: 10.1007/s12032-022-01653-8. [DOI] [PubMed] [Google Scholar]
- 131.Yousefnia S. A comprehensive review on miR-153: mechanistic and controversial roles of miR-153 in tumorigenicity of cancer cells. Front Oncol. 2022;12 doi: 10.3389/fonc.2022.985897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Indrieri A., Carrella S., Carotenuto P., Banfi S., Franco B. The pervasive role of the miR-181 family in development, neurodegeneration, and cancer. Int J Mol Sci. 2020;21(6):2092. doi: 10.3390/ijms21062092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Cavallari I., Ciccarese F., Sharova E., et al. The miR-200 family of microRNAs: fine tuners of epithelial-mesenchymal transition and circulating cancer biomarkers. Cancers (Basel) 2021;13(23):5874. doi: 10.3390/cancers13235874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Kozak J., Jonak K., Maciejewski R. The function of miR-200 family in oxidative stress response evoked in cancer chemotherapy and radiotherapy. Biomed Pharmacother. 2020;125 doi: 10.1016/j.biopha.2020.110037. [DOI] [PubMed] [Google Scholar]
- 135.Li T., Pan H., Li R. The dual regulatory role of miR-204 in cancer. Tumour Biol. 2016;37(9):11667–11677. doi: 10.1007/s13277-016-5144-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Chen W., Song J., Bian H., et al. The functions and targets of miR-212 as a potential biomarker of cancer diagnosis and therapy. J Cell Mol Med. 2020;24(4):2392–2401. doi: 10.1111/jcmm.14966. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Chauhan N., Dhasmana A., Jaggi M., Chauhan S.C., Yallapu M.M. miR-205: a potential biomedicine for cancer therapy. Cells. 2020;9(9):1957. doi: 10.3390/cells9091957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Ferrari E., Gandellini P. Unveiling the ups and downs of miR-205 in physiology and cancer: transcriptional and post-transcriptional mechanisms. Cell Death Dis. 2020;11(11):980. doi: 10.1038/s41419-020-03192-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Liang Y., Li S., Tang L. MicroRNA 320, an anti-oncogene target miRNA for cancer therapy. Biomedicines. 2021;9(6):591. doi: 10.3390/biomedicines9060591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Ye L., Wang F., Wu H., et al. Functions and targets of miR-335 in cancer. Onco Targets Ther. 2021;14:3335–3349. doi: 10.2147/OTT.S305098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Wei J., Lu Y., Wang R., et al. MicroRNA-375: potential cancer suppressor and therapeutic drug. Biosci Rep. 2021;41(9) doi: 10.1042/BSR20211494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Yan J.W., Lin J.S., He X.X. The emerging role of miR-375 in cancer. Int J Cancer. 2014;135(5):1011–1018. doi: 10.1002/ijc.28563. [DOI] [PubMed] [Google Scholar]
- 143.Yong-Ming H., Ai-Jun J., Xiao-Yue X., Jian-Wei L., Chen Y., Ye C. miR-449a: a potential therapeutic agent for cancer. Anticancer Drugs. 2017;28(10):1067–1078. doi: 10.1097/CAD.0000000000000555. [DOI] [PubMed] [Google Scholar]
- 144.Gao T., Zou M., Shen T., Duan S. Dysfunction of miR-802 in tumors. J Clin Lab Anal. 2021;35(11) doi: 10.1002/jcla.23989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Jiang Z.P., Zhou T.B. Role of miR-107 and its signaling pathways in diseases. J Recept Signal Transduct Res. 2014;34(5):338–341. doi: 10.3109/10799893.2014.896383. [DOI] [PubMed] [Google Scholar]
- 146.Peng W., Sha H., Sun X., et al. Role and mechanism of miR-187 in human cancer. Am J Transl Res. 2020;12(9):4873–4884. [PMC free article] [PubMed] [Google Scholar]
- 147.Favero A., Segatto I., Perin T., Belletti B. The many facets of miR-223 in cancer: oncosuppressor, oncogenic driver, therapeutic target, and biomarker of response. Wiley Interdiscip Rev RNA. 2021;12(6):e1659. doi: 10.1002/wrna.1659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Moghbeli M. Molecular interactions of miR-338 during tumor progression and metastasis. Cell Mol Biol Lett. 2021;26(1):13. doi: 10.1186/s11658-021-00257-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Yi Q., Xie W., Sun W., Sun W., Liao Y. A concise review of microRNA-383: exploring the insights of its function in tumorigenesis. J Cancer. 2022;13(1):313–324. doi: 10.7150/jca.64846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Bai H., Wu S. miR-451: a novel biomarker and potential therapeutic target for cancer. Onco Targets Ther. 2019;12:11069–11082. doi: 10.2147/OTT.S230963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Lu T.P., Lee C.Y., Tsai M.H., et al. miRSystem: an integrated system for characterizing enriched functions and pathways of microRNA targets. PLoS One. 2012;7(8) doi: 10.1371/journal.pone.0042390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.de Sousa M.C., Gjorgjieva M., Dolicka D., Sobolewski C., Foti M. Deciphering miRNAs’ action through miRNA editing. Int J Mol Sci. 2019;20(24):6249. doi: 10.3390/ijms20246249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Bajan S., Hutvagner G. RNA-based therapeutics: from antisense oligonucleotides to miRNAs. Cells. 2020;9(1):137. doi: 10.3390/cells9010137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Ling H., Fabbri M., Calin G.A. MicroRNAs and other non-coding RNAs as targets for anticancer drug development. Nat Rev Drug Discov. 2013;12(11):847–865. doi: 10.1038/nrd4140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Zhang Y., Wang Z., Gemeinhart R.A. Progress in microRNA delivery. J Control Release. 2013;172(3):962–974. doi: 10.1016/j.jconrel.2013.09.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Condrat C.E., Thompson D.C., Barbu M.G., et al. miRNAs as biomarkers in disease: latest findings regarding their role in diagnosis and prognosis. Cells. 2020;9(2):276. doi: 10.3390/cells9020276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Hong D.S., Kang Y.K., Borad M., et al. Phase 1 study of MRX34, a liposomal miR-34a mimic, in patients with advanced solid tumours. Br J Cancer. 2020;122(11):1630–1637. doi: 10.1038/s41416-020-0802-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.He B., Zhao Z., Cai Q., et al. miRNA-based biomarkers, therapies, and resistance in cancer. Int J Biol Sci. 2020;16(14):2628–2647. doi: 10.7150/ijbs.47203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Gajda E., Godlewska M., Mariak Z., Nazaruk E., Gawel D. Combinatory treatment with miR-7-5p and drug-loaded cubosomes effectively impairs cancer cells. Int J Mol Sci. 2020;21(14):1–19. doi: 10.3390/ijms21145039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Cui X., Sun Y., Shen M., et al. Enhanced chemotherapeutic efficacy of paclitaxel nanoparticles co-delivered with microRNA-7 by inhibiting paclitaxel-induced EGFR/ERK pathway activation for ovarian cancer therapy. ACS Appl Mater Interfaces. 2018;10(9):7821–7831. doi: 10.1021/acsami.7b19183. [DOI] [PubMed] [Google Scholar]
- 161.Murphy B.L., Obad S., Bihannic L., et al. Silencing of the miR-17∼92 cluster family inhibits medulloblastoma progression. Cancer Res. 2013;73(23):7068–7078. doi: 10.1158/0008-5472.CAN-13-0927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Pathi S.S., Jutooru I., Chadalapaka G., et al. GT-094, a NO-NSAID, inhibits colon cancer cell growth by activation of a reactive oxygen species-microRNA-27a: ZBTB10-specificity protein pathway. Mol Cancer Res. 2011;9(2):195–205. doi: 10.1158/1541-7786.MCR-10-0363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Toden S., Okugawa Y., Buhrmann C., et al. Novel evidence for curcumin and boswellic acid-induced chemoprevention through regulation of miR-34a and miR-27a in colorectal cancer. Cancer Prev Res (Phila) 2015;8(5):431–443. doi: 10.1158/1940-6207.CAPR-14-0354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Zhao W., Zhang X., Zhou Z., et al. Liraglutide inhibits the proliferation and promotes the apoptosis of MCF-7 human breast cancer cells through downregulation of microRNA-27a expression. Mol Med Rep. 2018;17(4):5202–5212. doi: 10.3892/mmr.2018.8475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Jiang J., Xie C., Liu Y., Shi Q., Chen Y. Up-regulation of miR-383-5p suppresses proliferation and enhances chemosensitivity in ovarian cancer cells by targeting TRIM27. Biomed Pharmacother. 2019;109:595–601. doi: 10.1016/j.biopha.2018.10.148. [DOI] [PubMed] [Google Scholar]
- 166.Ofori J.K., Karagiannopoulos A., Nagao M., et al. Human islet microRNA-200c is elevated in type 2 diabetes and targets the transcription factor ETV5 to reduce insulin secretion. Diabetes. 2022;71(2):275–284. doi: 10.2337/db21-0077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Xu K., Chen Z., Qin C., Song X. miR-7 inhibits colorectal cancer cell proliferation and induces apoptosis by targeting XRCC2. Onco Targets Ther. 2014;7:325–332. doi: 10.2147/OTT.S59364. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.O’Donnell K.A., Wentzel E.A., Zeller K.I., Dang C.V., Mendell J.T. c-Myc-regulated microRNAs modulate E2F1 expression. Nature. 2005;435(7043):839–843. doi: 10.1038/nature03677. [DOI] [PubMed] [Google Scholar]
- 169.Gong J., Zhang J.P., Li B., et al. MicroRNA-125b promotes apoptosis by regulating the expression of Mcl-1, Bcl-w and IL-6R. Oncogene. 2013;32(25):3071–3079. doi: 10.1038/onc.2012.318. [DOI] [PubMed] [Google Scholar]
- 170.Wan S., Wang J., Wang J., et al. Increased serum miR-7 is a promising biomarker for type 2 diabetes mellitus and its microvascular complications. Diabetes Res Clin Pract. 2017;130:171–179. doi: 10.1016/j.diabres.2017.06.005. [DOI] [PubMed] [Google Scholar]
- 171.Mahjoob G., Ahmadi Y., Fatima Rajani H., Khanbabaei N., Abolhasani S. Circulating microRNAs as predictive biomarkers of coronary artery diseases in type 2 diabetes patients. J Clin Lab Anal. 2022;36(5) doi: 10.1002/jcla.24380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Zampetaki A., Kiechl S., Drozdov I., et al. Plasma microRNA profiling reveals loss of endothelial miR-126 and other microRNAs in type 2 diabetes. Circ Res. 2010;107(6):810–817. doi: 10.1161/CIRCRESAHA.110.226357. [DOI] [PubMed] [Google Scholar]
- 173.Grieco G.E., Besharat Z.M., Licata G., et al. Circulating microRNAs as clinically useful biomarkers for type 2 diabetes mellitus: miRNomics from bench to bedside. Transl Res. 2022;247:137–157. doi: 10.1016/j.trsl.2022.03.008. [DOI] [PubMed] [Google Scholar]
- 174.Villard A., Marchand L., Thivolet C., Rome S. Diagnostic value of cell-free circulating microRNAs for obesity and type 2 diabetes: a meta-analysis. J Mol Biomark Diagn. 2015;6(6):251. doi: 10.4172/2155-9929.1000251. [DOI] [PMC free article] [PubMed] [Google Scholar]

