Skip to main content

The role of IGF2BP2, an m6A reader gene, in human metabolic diseases and cancers

Abstract

The human insulin-like growth factor 2 (IGF2) mRNA binding proteins 2 (IGF2BP2/IMP2) is an RNA-binding protein that regulates multiple biological processes. Previously, IGF2BP2 was thought to be a type 2 diabetes (T2D)-associated gene. Indeed IGF2BP2 modulates cellular metabolism in human metabolic diseases such as diabetes, obesity and fatty liver through post-transcriptional regulation of numerous genes in multiple cell types. Emerging evidence shows that IGF2BP2 is an N6-methyladenosine (m6A) reader that participates in the development and progression of cancers by communicating with different RNAs such as microRNAs (miRNAs), messenger RNAs (mRNAs) and long non-coding RNAs (lncRNAs). Additionally, IGF2BP2 is an independent prognostic factor for multiple cancer types. In this review, we summarize the current knowledge on IGF2BP2 with regard to diverse human metabolic diseases and its potential for cancer prognosis.

Introduction

The human insulin-like growth factor 2 (IGF2) mRNA binding proteins (IMP1-3 or IGF2BP1-3), first identified in 1999, attaches to the 5′ untranslated regions (5′ UTRs) of the translationally regulated IGF-II reader mRNA [1]. IGF2BP2, with a molecular mass of 66 kDa, has two N-terminal RNA-recognition motifs (RRMs) and four C-terminal human heterogeneous nuclear ribonucleoprotein (hnRNP)-K homology (KH) domains [1, 2] (Fig. 1a). Expression of IGFBP2 is generally maintained postnatally and participates in localization, stability and translation of RNAs [3].

Fig. 1
figure 1

a The structure of RRMs and KH domains. b Polymorphisms in the IGF2BP2. Boxes represent exons whereas the red color represents 5′ and 3′ untranslated regions of IGF2BP2. The ~ 80 T2D-associated SNPs are localized within the second intron of the IMP2 gene at chromosome 3

Recent genome-wide association studies (GWAS) have revealed that IGF2BP2 gene induces the development of type 2 diabetes (T2D) by disrupting insulin secretion [2]. Mechanistically, IGF2BP2 modulates cellular metabolism by post transcriptional regulation of several genes in numerous cell types and pathways [4]. In addition, dysregulation of IGF2BP2 is associated with progression of cancers and cancer stem cells [5]. Recently, IGF2BP2 has been shown to read N6-methyladenosine (m6A), the most abundant internal RNA modification in eukaryotic cells [6]. IGF2BP2 communicates with several RNAs such as microRNAs (miRNAs) [7], messenger RNAs (mRNAs) [8] and long non-coding RNAs (lncRNAs), where it regulates several biological processes [9]. M6A-RNA methylation refers to methylation of adenosine bases at position N6 in 3′UTRs near the stop codons but within the internal long exons [10, 11]. Modified IGF2BP2 participates in the development and progression of multiple metabolic disease and cancers, including diabetes [2], obesity [12], fatty liver [13], breast cancer [14], colorectal carcinoma [15], esophageal adenocarcinoma [16], glioma [17], hepatocellular carcinoma [18], lung cancer [19], pancreatic cancer [20] and many others.

In this review, we summarize the current evidence on the relationship between IGF2BP2 and metabolic disease, as well as the biological mechanisms underlying IGF2BP2 functions in cancers.

Expression of IGF2BP2 and metabolic disease

Murine models have uncovered the role of IGF2BP2 in metabolic diseases including diabetes, obesity, fatty liver and among others [4]. Based on the GWAS, a cluster of single nucleotide polymorphisms (SNPs) in the second intron of IGF2BP2 (Fig. 1b) have been implicated for T2D. The association between IGF2BP2 and human metabolic diseases stems in its post-transcriptional regulation of numerous genes in different cell types and pathway [4]. The sections that follow discuss in detail, the associations between IGF2BP2 and several metabolic diseases (Table 1).

Table 1 The roles of IGF2BP2 in various metabolic diseases and the associations between the expression of IGF2BP2 SNPs and the development several metabolic diseases as well as cancer in different populations

Expression of IGF2BP2 and diabetes

In 2007, Grarup, et al. [21] found no association between IGF2BP2 genetic variants and pancreatic-cell dysfunction in Danish population. Later on, the IGF2BP2 variant was found to decrease glucose-stimulated insulin secretion in the first but not the second phase of diabetes development [22]. Additionally, IGF2BP2 has also been implicated in the development of T2D/impaired glucose tolerance [23]. In Indian [24] and Chinese [25] populations, IGF2BP2 was found to be closely associated with T2D even after adjusting for age, sex and BMI. For European, Czech and Swedish populations, IGF2BP2 polymorphism has been associated with diabetic nephropathy in male patients with type 1 diabetes (T1D) [26]. Accordingly, we summarize how IGF2BP2 SNPs participate in the development of diabetes.

Expression of IGF2BP2 rs4402960 and rs1470579 in diabetes

IGF2BP2 rs4402960 and rs1470579 are the most common SNPs in diabetes. Research shows that expressions of IGF2BP2 rs4402960 gene variant in Chinese Han [27,28,29,30,31], Japanese [32], Asian [27, 33], Icelandic [34], Greek-Cypriot [35], Czechs [26], Germania [33, 36], Lebanese [37], Arabian [38], Tunisian [39], Moroccan [38] and Indian population [40] increase the risk for T2D. IGF2BP2 rs4402960 is also associated with lower fasting insulin level and impaired β-cell function, both associated with obesity [34]. Meanwhile, wild C IGF2BP2 rs4402960 allele protects against T2D in Chinese Han individuals. In addition, the therapeutic efficacy of repaglinide is enhanced in Chinese T2D patients with IGF2BP2 rs4402960 polymorphism [41]. The effect of pioglitazone on postprandial plasma glucose, glycated hemoglobin, serum triglycerides and high-density lipoprotein cholesterol is in Chinese individuals with rs4402960 polymorphism [30]. Moreover, IGF2BP2 rs4402960 is strongly associated with the development of gestational diabetes mellitus (GDM), besides being a potential diagnostic marker for GDM as well [42]. However, no association has been found between IGF2BP2 rs4402960 polymorphism and the risk of developing GDM in Polish [43] and Chinese [44] population, but it influence the length of gestation period and health (based on Apgar scores) of newborns in these populations.

On the other hand, IGF2BP2 rs1470579 is also associated with T2D in Lebanese [45], Chinese Han [29, 30] and Iranian populations [46]. In addition, IGF2BP2 rs1470579 polymorphism reduces the therapeutic efficacy of repaglinide in T2D patients in Chinese population [41]. The effect of pioglitazone against PPG, TG and HDL-C is also lower in Chinese patients with rs1470579 gene variant [30].

However, other researches failed to replicate the confirmed rs4402960 and rs1470579 susceptibility variants in French Caucasians [47], Indian [48, 49], Chinese Han [50] and Russian populations [51]. A global meta-analysis of 35 studies encompassing 175,965 subjects on the association between IGF2BP2 rs4402960 and rs1470579 and T2D revealed that even though these polymorphisms increases the risk of developing T2D, the associations vary among ethnic populations [52].

Other IGF2BP2 SNPs in diabetes

IGF2BP2 rs11705701 has been associated with low body fat, which contributes to insulin resistance and consequently T2D risk in Mexican American population [53]. IGF2BP2 rs11705701 has also been associated with higher risk of T2D in Russian population. Meanwhile, allele A of rs11705701 has been linked with low levels of short isoform (p58) but high levels of the long isoform (p66) of IGF2BP2 protein in adipose tissue of non-obese individuals [51]. Additionally, IGF2BP2 rs11705701 has been strongly associated with prediabetes in female patients [54]. Although no association has been found between IGF2BP2 rs11705701 and the risk of developing GDM in Polish population, it lengthens the gestation and improves the health (based on Apgar scores) of newborns in this population [43]. On the other hand, rs9826022, a rare mutation in the 3′downstream region of IGF2BP2, is closely associated with T2D [47]. Besides, IGF2BP2 rs11927381 and rs7640539 are all associated with the risk of developing T2D among Chinese Han population [55]. Meanwhile, rs6777038, rs16860234 and rs7651090 of IGF2BP2 are closely linked with glutamic acid decarboxylase (GAD) antibody- negative diabetes [56].

The mechanisms underlying IGF2BP2 in diabetic nephropathy

Diabetic nephropathy (DN) is one of the most serious microvascular complications that increases the risk of death of T2D patients [57]. Recently, it is has been found that the role of IGF2BP2 in DN depends on interlinked communication with several other genes, miRNAs and lncRNAs (Fig. 2). Laminin-β2 (lamb2), the key laminin subunit, participates in maintaining normal basement membrane structure and function of the glomerular [58]. Intriguingly, IGF2BP2 regulates expression of lamb2 by directly targeting lamb2 mRNA in actin cytoskeleton [59]. IGF2 also regulates the regeneration and survival of podocytes [60, 61]. For instance, Jing, et al. [62] found that antisense of insulin-like growth factor-2 receptor non-coding RNA (AIRN) regulates the translation of IGF2 and lamb2 by binding to IGF2BP2, thus maintaining normal podocyte viability and glomerular barrier function, preventing DN. As such, AIRN is potentially a new therapeutic target against diabetic nephropathy in individuals with low lamb2 levels.

Fig. 2
figure 2

The role of IGF2BP2 in DN. IGF2BP2, regulated by AIRN and miR-let-7b/HMGA2, promotes proliferation of podocytes and functioning of the glomerular by targeting lamb2 and IGF2

IGF2BP2 in fatty liver and steatohepatitis and the specific mechanisms

IGF2BP2 up-regulates the expression of fatty acid elongase 6 (ELOVL6), which catalyzes the elongation of C16 fatty acids to C18, contributing to the development of human nonalcoholic steatohepatitis [63]. Besides, the activation of ELOVL6 is regulated by sterol regulatory element binding transcription factor 1. IGF2BP2 promotes the development of steatohepatitis-associated cirrhosis in de-differentiated cells by accelerating ductular reaction [64]. Dai et al. [12] found that IGF2BP2−/− mice were highly resistant to diet-induced obesity and fatty liver disease, and showed greater glucose tolerance and insulin sensitivity. IGF2BP2 inhibits the translation of the untranslated Ucp1 bearing mRNAs and binds to mitochondrial components. However, Regué et al. [13] reported that specific IGF2BP2-hepatocyte knockout results in greater accumulation of triglycerides in the liver. This suggests that the expression of IGF2BP2, which encodes carnitine palmi toyltransferase 1A (CPT1A) and peroxisome proliferator-activated receptor, disrupts fatty acid oxidation, thus promoting accumulation of the fatty acids in the liver.

IGF2BP2 and cancers

In recent years, over-expression of IGF2BP2 in multiple human cancers has been associated with poorer prognosis of the disease. For instance, over-expression of IGF2BP2 confers shorter survival and poor prognosis of patients with acute myelocytic leukemia (AML) [65], breast cancer [14], esophageal carcinoma [16], low-grade gliomas [17], hepatocellular carcinoma (HCC) [66], head and neck squamous cell carcinoma (HNSCC) [67], pancreatic ductal adenocarcinoma (PDAC) [20, 68,69,70] and gallbladder carcinoma (GBC) [71]. Herein, we summarize the specific roles of IGF2BP2 in multiple cancers and provide a comprehensive view of IGF2BP2 (Fig. 2; Table 2).

Table 2 The expression, clinical significance and biological functions of IGF2BP2 in different cancer types

IGF2BP2 in different cancers

IGF2BP2 in breast cancer

IGF2BP2 rs4402960 increases the risk of developing breast cancer in female Chinese Hans [72]. Compared to luminal or apocrine subtypes, IGF2BP2 is over-expressed in basal-like breast cancer tissues [14]. Liu et al. [73] further reported that IGF2BP2 is over-expressed in breast cancer tissues, where it up-regulates auto-immune response. Consequently, over-expression of IGF2BP2 is not only a potential biomarker for developing breast cancer but also a novel diagnostic factor for the same disease. Meanwhile, Ccn6, secreted by normal breast epithelium, can suppress the expression of IGF2BP2 protein in cancerous breast tissues, thus modulating the growth of the tumor. Ccn6/Wisp3 knockdown up-regulated the expression of IGF2BP2 in mice, who developed mammary carcinomas characterized by spindle and squamous differentiation, validated hallmarks of metaplastic breast carcinomas [74] (Fig. 3).

Fig. 3
figure 3

The role of IGF2BP2 in regulating multiple human cancer parameters. IGF2BP2 regulates proliferation, apoptosis, cell cycle, migration, invasion, cell viability, MDV, EMT, glycolysis, autophagy, self-renewal, tumor growth etc., of multiple cancer types such AML, breast cancer, colorectal cancer, ERMS, glioma, hepatocellular carcinoma, lung cancer, pancreatic cancer and GBC. IGF2BP2 performs its modulatory functions by interacting with miRNAs, lncRNAs, mRNAs and other m6A-related genes

IGF2BP2 in pancreatic cancer

IGF2BP2 is also over-expressed in PDAC [68, 69]. Meanwhile, Glucose transporter 1 (GLUT1) is an integral membrane protein consisting of 12 transmembrane helices and an intracellular domain which promotes aerobic glycolysis and proliferation of cancer cells [75,76,77]. IGF2BP2 also promotes aerobic glycolysis and proliferation of PDAC cells by directly binding to and stabilizing GLUT1 mRNA [68]. Further correlation analyses have revealed that over-expression of IGF2BP2 inhibits the expression of apoptosis (B-cell lymphoma-extra large) and ubiquitination (E3 ubiquitin ligase Smurf1 and F-Box protein 45) associated genes. Expression of IGF2BP2 also promotes tumor progression by inducing epithelial-mesenchymal transition (EMT).

IGF2BP2 in esophageal cancer

Multivariate logistic analyses have demonstrated that IGF2BP2 rs1470579 and rs4402960 polymorphisms increase the risk of developing esophageal squamous-cell carcinoma [78]. In contrast, a separate research showed that IGF2BP2 rs1470579 phenotype decreases the risk of developing esophagogastric junction adenocarcinoma in Eastern Chinese Han population [79]. Elsewhere, IGF2BP2 polymorphism has been found to increase the risk of developing human esophageal adenocarcinoma and Barret’s esophageal tissue. It also promotes growth, proliferation, metabolism and inflammation of cancer tissues [16].

IGF2BP2 in other cancers

Stratified analyses and haplotype analysis revealed that in Eastern Chinese Han population, IGF2BP2 rs1470579 and rs4402960 polymorphism decreased the risk of developing NSCLC among females < 60 years and non-alcohol drinker [19]. However, IGF2BP2 rs4402960 and rs6769511 phenotypes strongly predict (positive) response of metastatic gastric cancer patients to chemotherapy [80].

Over-expression of IGF2BP2 in AML patients negatively correlates with expression of CCAAT/enhancer binding protein α, a positive prognostic factor. Conversely, over-expression of IGF2BP2 positively correlates with the expression of poor prognostic factors including mutated FMS-like tyrosine kinase 3 and isocitrate dehydrogenase 1 [65]. Intriguingly, AML cells continue to grow in IGF2BP2 knockdown subjects. Expression of IGF2BP2 has also been shown to be up-regulated in colorectal cancer (CRC) tissues, promoting proliferation and survival of the cancer cells [81]. Analysis of The Cancer Genome Atlas (TCGA) data combined with immunohistochemical (IHC) tests [67] revealed that IGF2BP2 is over-expressed in HNSCC tissues, promoting scavenging and degradation, synthesis and metabolism and growth of tumor cells. In addition, over-expression of IGF2BP2 is a risk factor for poor prognosis of HNSCC patients. Besides, IGF2BP2 is frequently up-regulated in GBC, and has been shown to promote growth of xenograft tumors in mice. Moreover, over-expression of IGF2BP2 promotes the production of reactive oxygen species and expression of small GTPase Ras-related C3 botulinum toxin substrate 1 in GBC [71]. In addition, high mobility group AT-hook 2 (HMGA2), a DNA-binding protein, is often reactivated in various cancers. Expression of HMGA2 enhances metastasis and is associated with poor prognosis of cancers [82,83,84]. Interestingly, HMGA2 regulates IGF2BP2, a noble rhabdomyosarcoma (ERMS) protein key in survival and growth of cells. IGF2BP2 binds NRAS mRNA, regulating the expression of NRAS protein [85]. RPSAP52, an antisense transcribed pseudogene of HMGA2, promotes proliferation of sarcoma and self-renewal pathways by cross-linking with IGF2BP2 [86]. Except for HMGA2, IGF2BP2 markedly promotes functions of IGF and proliferation of cancer cells by binding and stabilizing HMGA1 [8].

Mechanism underlying IGF2BP2 regulation of cancers

Mechanistically, IGF2BP2 modulates proliferation, migration, invasion, metastasis and apoptosis of cancer cells by regulation transcription of miRNAs, lncRNAs and other m6A-related genes [7, 9, 15].

IGF2BP2 with miRNAs in cancers

miRNAs are a group of endogenous, highly conserved, noncoding RNAs (18–25 nts in length) that regulate gene expression both transcriptionally and post-transcriptionally [87,88,89,90]. Accumulating evidence has demonstrated remarkable relationship between the expression patterns of miRNAs and IGF2BP2 and development as well as progression of tumors. For instance, miR-1193 is often down-regulated in breast cancer tissues and culture cell lines. However, over-expression of miR-1193 inhibits proliferation and invasion of breast cancer cells by binding the 3′UTR region of IGF2BP2 mRNA, activating ERK and PI3K/Akt signaling pathways [7]. The expression of IGF2BP2 is also up-regulated in CRC tissues, where it promotes proliferation and survival of the cancer cells by post-transcriptionally inhibiting miR-195-mediated degradation of rubisco assembly factor 1 [81]. Additionally, miR-188 is down-regulated in glioma cells and tissues, its over-expression inhibits proliferation, migration and invasion of glioma cells and tissues by directly targeting IGF2BP2 [91]. Specifically, miR-138 represses expression of IGF2BP2 by targeting its 3ʹ-UTR. This intern inhibits EMT and suppresses proliferation and invasion of low-grade glioma cells [17]. miR-216b also suppresses proliferation, migration and invasion of HCC by down-regulating the expression of IGF2BP2, found to be most often over-expressed in HCC tissues [92]. On the other hand, flap endonuclease-1 (FEN1), a multifunctional structure-specific nuclease critical in maintaining normal cell growth, is up-regulated in HCC [93, 94]. Pu et al. [66] reported that over-expression of IGF2BP2 promotes proliferation of HCC both in vitro and in vivo. Mechanistically, IGF2BP2 directly binds the m6A site on FEN1 mRNA, stabilizing the mRNA. Hepatitis B virus suppresses p53-mediated activation of miR-216b and promotes the expression of IGF2BP2. Furthermore, the expression of miR-let-7a which positively correlates with hypermethylation of MIRLRT7A3, modulates the expression of IGF2BP2 [18]. In lung cancer, the over-expressed miR-485-5p inhibits growth and invasion of cancer cells, arrests the G0/G1 cycle and disrupts the TGF-β-induced EMT by directly targeting IGF2BP2 [95]. Finally, the upregulated expression of IGF2BP2, a target for miR-141, promotes proliferation of PDAC via the PI3K/Akt signaling pathway [20].

IGF2BP2 and lncRNAs in cancers

LncRNAs, previously thought to cause transcriptional noise, are a class of non-protein-coding RNAs longer than 200nt that regulate several physiological and pathological processes [96, 97]. Increasing evidence shows that IGF2BP2, in conjunction with multiple lncRNAs, regulate multiple biological functions. For instance, lncRNA HOX transcript antisense RNA (HOTAIR) regulates the expression of target genes by directly interaction with histone modification complexes [98, 99]. IGF2BP2-mediated over-expresssion of HOTAIR promotes proliferation, migration, invasion, microvessel density value (MDV) and EMT, but represses apoptosis of colon cancer cells [9]. Moreover, lncRNA LINRIS inhibits K139 mediated ubiquitination of IGF2BP2, preventing the degradation of IGF2BP2 via autophagy-lysosome pathway [100]. Consequently, LINRIS knockdown weakens downstream effects of IGF2BP2, particularly MYC-mediated glycolysis in CRC cells and proliferation of cancer cells. On the other hand, lncRNA 91H, a long non-coding antisense transcript located at H19/IGF2 locus, participates in tumor development [101, 102]. lncRNA 91H silencing modulates proliferation, migration, invasion, autophagy and expression of mammalian target of rapamycin (mTOR) in CRC cancer cells by suppressing IGF2 expression, which up-regulates the expression of IGF2BP2 [103]. lncRNA RHPN1-AS1 promotes proliferation and metastasis but inhibits apoptosis of HCC cells [104]. lncRNA 91H performs its modulatory function via miR-596, which binds to IGF2BP2.

Intriguingly, research shows that lncRNAs and IGF2BP2 can regulate each other. For instance, IGF2BP2 promotes proliferation and stemness-like properties of pancreatic cancer cells by binding and stabilizing m6A modified DANCR RNA [70].

IGF2BP2 with other m6A-related genes in cancers

Further molecular insights implicate m6A alterations in the pathogenesis and development of cancers via regulating the expression of multiple tumor-associated genes [105, 106]. Besides, different m6A-ralated genes cross-link with each other to modulate the development of multiple cancers [107, 108]. METTL3 predominantly catalyses m6A methyltransferase system and regulates numerous processes in multiple human cancers [109]. METTL3 promotes self-renewal of CRC cell, proliferation and migration of stem cells in vitro as well as tumorigenesis and metastasis of advances CRC in vivo, mainly by targeting sex determining region Y (SRY)-box 2 (SOX2) [15]. However, METTL3 functions are IGF2BP2-dependent, which recognizes the coding sequence regions of methylated SOX2 transcripts, and prevents degradation of SOX2 mRNA.

Discussion

This review describes the role and specific expression patterns of IGF2BP2 in human metabolic diseases and cancers. Even though IGF2BP2 SNPs are widely associated with the risk of developing diabetes, the relationship between expression patters of the resultant proteins and human metabolic diseases and cancers vary among ethnic populations. For instance, even though Grarup et al. [21] found no association between IGF2BP2 gene variants and T2D in Danish population, Groenewoud et al. [22] reported that expression of IGF2BP2 polymorphisms decreased glucose-stimulated insulin secretion in the first phase of diabetes development in Dutch and Germany’s population. More intriguingly, the association between IGF2BP2 SNPs and metabolic diseases varies even within the same ethnic population. For example, Zhang et al. [30] found that IGF2BP2 rs4402960 is associated with T2D in patients from Anhui, a province in China. However, the relationship between IGF2BP2 rs4402960 expression and T2D was insignificant in participants from Shanghai, China [50]. Hence, the associations between IGF2BP2 variations and T2D should be interpreted with caution. However, IGF2BP2 may induce human metabolic diseases via posttranscriptional regulation of various genes associated with specific cell types and pathways. For example, hepatocyte-specific IGF2BP2 knockdown inhibits oxidation of fatty acid, leading to accumulation of triglyceride in mice liver [13]. On the other hand, over-expression of IGF2BP2 increases the risk of developing numerous cancers. However, certain IGF2BP2 gene variants decrease the risk of NSCLC among females of Chinese Han population [19]. This suggests that the role of IGF2BP2 in cancers development varies among tumors and ethnic groups.

Ning Dai [4] had reviewed and summarized the expression of IGF2BP2 impairs insulin secretion. In addition, IGF2BP2 regulates multiple biological processes post-transcriptionally. Additionally, IGF2BP2 regulates multiple physiological processes including embryonic development, neuronal differentiation and metabolism, insulin resistance in diabetics and carcinogenesis [5]. Ning Dai focused on the associations and mechanism underlying expression of IGF2BP2 SNPs and the development of metabolic diseases including T2D, nonalcoholic steatohepatitis, obesity and fatty liver disease. We also summarized current works on the association among IGF2BP2, miRNAs, lncRNAs, mRNAs and other m6A-related genes and development as well as regulation of cancers. Findings of this research may uncover new frontier for the exploration of development and treatment of tumors.

Recent researches have focused on the role of metabolic pathways in various cancer parameters. Specifically, cancer cells exhibit metabolic reprogramming such as dysregulated glucose uptake, excessive lipid synthesis and glutaminolysis [110]. These transformations are essential parameters in the maintenance and development of malignant phenotypes in harsh microenvironments [111,112,113,114,115]. The role of IGF2BP2 in glucose tolerance, insulin sensitivity, fatty acid oxidation and the development of metabolic diseases had been reviewed recently [13, 21,22,23, 28, 53]. The metabolic role of IGF2BP2 in cancers is scarce. However, available evidence demonstrates that IGF2BP2 targets GLUT1, promoting aerobic glycolysis and proliferation of PDAC cells [68, 75,76,77]. IGF2BP2 also promotes metabolism of esophageal cancer and HNSCC [16, 67].

High-throughput sequencing technology recently revealed that m6A modification, circRNAs, miRNAs and lncRNAs are emerging important regulators of several biological process [116]. Overall, we speculate the complex relationship among IGF2BP2, circRNAs, miRNAs and lncRNAs participates in the development of both metabolic diseases and cancers. Nonetheless, further researches are needed to unlock the precise interrelationships among pathways regulated by the above molecules.

Conclusion

The interrelationship among IGF2BP2, miRNAs, lncRNAs and their target genes with regard to cancers and metabolic diseases are reported but inconclusive. Nevertheless, current available evidence suggests the critical role of IGF2BP2 SNPs in the development of the two diseases.

Availability of data and materials

Not applicable.

References

  1. Nielsen J, Christiansen J, Lykke-Andersen J, Johnsen AH, Wewer UM, Nielsen FC. A family of insulin-like growth factor II mRNA-binding proteins represses translation in late development. Mol Cell Biol. 1999;19:1262–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Christiansen J, Kolte AM, Hansen T, Nielsen FC. IGF2 mRNA-binding protein 2: biological function and putative role in type 2 diabetes. Stem Cells Int. 2009;43:187–95.

    CAS  Google Scholar 

  3. Dai N, Rapley J, Angel M, Yanik MF, Blower MD, Avruch J. mTOR phosphorylates IMP2 to promote IGF2 mRNA translation by internal ribosomal entry. Genes Dev. 2011;25:1159–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Dai N. The diverse functions of IMP2/IGF2BP2 in metabolism. Trends Endocrinol Metab. 2020;31:670–9.

    Article  CAS  PubMed  Google Scholar 

  5. Cao J, Mu Q. The roles of insulin-like growth factor 2 mRNA-binding protein 2 in cancer and cancer stem cells. Stem Cells Intern. 2018;2018:4217259.

    Google Scholar 

  6. Maity A, Das B. N6-methyladenosine modification in mRNA: machinery, function and implications for health and diseases. FEBS J. 2016;283:1607–30.

    Article  CAS  PubMed  Google Scholar 

  7. Li X, Li Y, Lu H. miR-1193 suppresses proliferation and invasion of human breast cancer cells through directly targeting IGF2BP2. Oncol Res. 2017;25:579–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Dai N, Ji F, Wright J, Minichiello L, Sadreyev R, Avruch J. IGF2 mRNA binding protein-2 is a tumor promoter that drives cancer proliferation through its client mRNAs IGF2 and HMGA1. 2017; 6.

  9. Wu XL, Lu RY, Wang LK, Wang YY, Dai YJ, Wang CY, et al. Long noncoding RNA HOTAIR silencing inhibits invasion and proliferation of human colon cancer LoVo cells via regulating IGF2BP2. J Cell Biochem. 2018;120:1221–31.

    Article  CAS  Google Scholar 

  10. Ke S, Alemu EA, Mertens C, Gantman EC, Fak JJ, Mele A, et al. A majority of m6A residues are in the last exons, allowing the potential for 3′ UTR regulation. Genes Dev. 2015;29:2037–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Dominissini D, Moshitch-Moshkovitz S, Schwartz S, Salmon-Divon M, Ungar L, Osenberg S, et al. Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq. Nature. 2012;485:201–6.

    Article  CAS  PubMed  Google Scholar 

  12. Dai N, Zhao L, Wrighting D, Krämer D, Majithia A, Wang Y, et al. IGF2BP2/IMP2-deficient mice resist obesity through enhanced translation of Ucp1 mRNA and other mRNAs encoding mitochondrial proteins. J Cell Sci. 2015;21:609–21.

    CAS  Google Scholar 

  13. Regué L, Minichiello L. Liver-specific deletion of IGF2 mRNA binding protein-2/IMP2 reduces hepatic fatty acid oxidation and increases hepatic triglyceride accumulation. J Biol Chem. 2019;294:11944–51.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Barghash A, Helms V, Kessler SM. Overexpression of IGF2 mRNA-binding protein 2 (IMP2/p62) as a feature of basal-like breast cancer correlates with short survival. Scand J Immunol. 2015;82:142–3.

    Article  CAS  PubMed  Google Scholar 

  15. Li T, Hu PS, Zuo Z, Lin JF, Li X, Wu QN, et al. METTL3 facilitates tumor progression via an m(6)A-IGF2BP2-dependent mechanism in colorectal carcinoma. Mol Cancer. 2019;18:112.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Barghash A, Golob-Schwarzl N, Helms V, Haybaeck J, Kessler SM. Elevated expression of the IGF2 mRNA binding protein 2 (IGF2BP2/IMP2) is linked to short survival and metastasis in esophageal adenocarcinoma. Elife. 2016;7:49743–50.

    Google Scholar 

  17. Yang Y, Liu X, Cheng L, Li L, Wei Z, Wang Z, et al. Tumor suppressor microRNA-138 suppresses low-grade glioma development and metastasis via regulating IGF2BP2. Diabetes Metab Syndr Obes. 2020;13:2247–60.

    CAS  Google Scholar 

  18. Waly AA, El-Ekiaby N, Assal RA, Abdelrahman MM, Hosny KA, El Tayebi HM, et al. Methylation in MIRLET7A3 gene induces the expression of IGF-II and its mRNA binding proteins IGF2BP-2 and 3 in hepatocellular carcinoma. Front Physiol. 2018;9:1918.

    Article  PubMed  Google Scholar 

  19. Chen S, Qiu H, Liu C, Wang Y, Tang W, Kang M. Relationship between IGF2BP2 and IGFBP3 polymorphisms and susceptibility to non-small-cell lung cancer: a case–control study in Eastern Chinese Han population. J Cell Biochem. 2018;10:2965–75.

    CAS  Google Scholar 

  20. Xu X, Yu Y, Zong K, Lv P, Gu Y. Up-regulation of IGF2BP2 by multiple mechanisms in pancreatic cancer promotes cancer proliferation by activating the PI3K/Akt signaling pathway. J Exp Clin Cancer Res. 2019;38:497.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Grarup N, Rose CS, Andersson EA, Andersen G, Nielsen AL, Albrechtsen A, et al. Studies of association of variants near the HHEX, CDKN2A/B, and IGF2BP2 genes with type 2 diabetes and impaired insulin release in 10,705 Danish subjects: validation and extension of genome-wide association studies. Diabetes. 2007;56:3105–11.

    Article  CAS  PubMed  Google Scholar 

  22. Groenewoud MJ, Dekker JM, Fritsche A, Reiling E, Nijpels G, Heine RJ, et al. Variants of CDKAL1 and IGF2BP2 affect first-phase insulin secretion during hyperglycaemic clamps. Diabetologia. 2008;51:1659–63.

    Article  CAS  PubMed  Google Scholar 

  23. van Hoek M, Langendonk JG, de Rooij SR, Sijbrands EJ, Roseboom TJ. Genetic variant in the IGF2BP2 gene may interact with fetal malnutrition to affect glucose metabolism. Diabetes. 2009;58:1440–4.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Sanghera DK, Ortega L, Han S, Singh J, Ralhan SK, Wander GS, et al. Impact of nine common type 2 diabetes risk polymorphisms in Asian Indian Sikhs: PPARG2 (Pro12Ala), IGF2BP2, TCF7L2 and FTO variants confer a significant risk. BMC Med Genet. 2008;9:59.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Han X, Luo Y, Ren Q, Zhang X, Wang F, Sun X, et al. Implication of genetic variants near SLC30A8, HHEX, CDKAL1, CDKN2A/B, IGF2BP2, FTO, TCF2, KCNQ1, and WFS1 in type 2 diabetes in a Chinese population. BMC Med Genet. 2010;11:81.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Gu T, Horová E, Möllsten A, Seman NA, Falhammar H, Prázný M, et al. IGF2BP2 and IGF2 genetic effects in diabetes and diabetic nephropathy. J Diabetes Complicat. 2012;26:393–8.

    Article  Google Scholar 

  27. Ng MC, Park KS, Oh B, Tam CH, Cho YM, Shin HD, et al. Implication of genetic variants near TCF7L2, SLC30A8, HHEX, CDKAL1, CDKN2A/B, IGF2BP2, and FTO in type 2 diabetes and obesity in 6,719 Asians. Diabetes. 2008;57:2226–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Wu Y, Li H, Loos RJ, Yu Z, Ye X, Chen L, et al. Common variants in CDKAL1, CDKN2A/B, IGF2BP2, SLC30A8, and HHEX/IDE genes are associated with type 2 diabetes and impaired fasting glucose in a Chinese Han population. Diabetes. 2008;57:2834–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Zhang SM, Xiao JZ, Ren Q, Han XY, Tang Y, Yang WY, et al. Replication of association study between type 2 diabetes mellitus and IGF2BP2 in Han Chinese population. Chin Med J (Engl). 2013;126:4013–8.

    CAS  Google Scholar 

  30. Zhang LF, Pei Q, Yang GP, Zhao YC, Mu YF, Huang Q, et al. The effect of IGF2BP2 gene polymorphisms on pioglitazone response in Chinese type 2 diabetes patients. Pharmacology. 2014;94:115–22.

    Article  CAS  PubMed  Google Scholar 

  31. Rao P, Wang H, Fang H, Gao Q, Zhang J, Song M, et al. Association between IGF2BP2 polymorphisms and type 2 diabetes mellitus: a case–control study and meta-analysis. Int J Environ Res Public Health. 2016;13:574.

    Article  PubMed Central  CAS  Google Scholar 

  32. Omori S, Tanaka Y, Takahashi A, Hirose H, Kashiwagi A, Kaku K, et al. Association of CDKAL1, IGF2BP2, CDKN2A/B, HHEX, SLC30A8, and KCNJ11 with susceptibility to type 2 diabetes in a Japanese population. Diabetes. 2008;57:791–5.

    Article  CAS  PubMed  Google Scholar 

  33. Jia H, Yu L, Jiang Z, Ji Q. Association between IGF2BP2 rs4402960 polymorphism and risk of type 2 diabetes mellitus: a meta-analysis. Arch Med Res. 2011;42:361–7.

    Article  CAS  PubMed  Google Scholar 

  34. Rodriguez S, Eiriksdottir G, Gaunt TR, Harris TB, Launer LJ, Gudnason V, et al. IGF2BP1, IGF2BP2 and IGF2BP3 genotype, haplotype and genetic model studies in metabolic syndrome traits and diabetes. Growth Horm IGF Res. 2010;20:310–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Votsi C, Toufexis C, Michailidou K, Antoniades A, Skordis N, Karaolis M, et al. Type 2 diabetes susceptibility in the Greek-Cypriot population: replication of associations with TCF7L2, FTO, HHEX, SLC30A8 and IGF2BP2 polymorphisms. Genes (Basel). 2017;8:16.

    Article  CAS  Google Scholar 

  36. Herder C, Rathmann W, Strassburger K, Finner H, Grallert H, Huth C, et al. Variants of the PPARG, IGF2BP2, CDKAL1, HHEX, and TCF7L2 genes confer risk of type 2 diabetes independently of BMI in the German KORA studies. Horm Metab Res. 2008;40:722–6.

    Article  CAS  PubMed  Google Scholar 

  37. Nemr R, Echtay A, Dashti EA, Almawi AW, Al-Busaidi AS, Keleshian SH, et al. Strong association of common variants in the IGF2BP2 gene with type 2 diabetes in Lebanese Arabs. Diabetes Res Clin Pract. 2012;96:225–9.

    Article  CAS  PubMed  Google Scholar 

  38. Benrahma H, Charoute H, Lasram K, Boulouiz R, Atig RK, Fakiri M, et al. Association analysis of IGF2BP2, KCNJ11, and CDKAL1 polymorphisms with type 2 diabetes mellitus in a Moroccan population: a case–control study and meta-analysis. Biochem Genet. 2014;52:430–42.

    Article  CAS  PubMed  Google Scholar 

  39. Lasram K, Ben Halim N, Benrahma H, Mediene-Benchekor S, Arfa I, Hsouna S, et al. Contribution of CDKAL1 rs7756992 and IGF2BP2 rs4402960 polymorphisms in type 2 diabetes, diabetic complications, obesity risk and hypertension in the Tunisian population. J Diabetes. 2015;7:102–13.

    Article  CAS  PubMed  Google Scholar 

  40. Chauhan G, Spurgeon CJ, Tabassum R, Bhaskar S, Kulkarni SR, Mahajan A, et al. Impact of common variants of PPARG, KCNJ11, TCF7L2, SLC30A8, HHEX, CDKN2A, IGF2BP2, and CDKAL1 on the risk of type 2 diabetes in 5,164 Indians. Diabetes. 2010;59:2068–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Huang Q, Yin JY, Dai XP, Pei Q, Dong M, Zhou ZG, et al. IGF2BP2 variations influence repaglinide response and risk of type 2 diabetes in Chinese population. Acta Pharmacol Sin. 2010;31:709–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Chon SJ, Kim SY, Cho NR, Min DL, Hwang YJ, Mamura M. Association of variants in PPARγ2, IGF2BP2, and KCNQ1 with a susceptibility to gestational diabetes mellitus in a Korean population. Yonsei Med J. 2013;54:352–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Tarnowski M, Bujak J, Kopytko P, Majcher S, Ustianowski P, Dziedziejko V, et al. Effect of FTO and IGF2BP2 gene polymorphisms on duration of pregnancy and Apgar scores in women with gestational diabetes. J Obstet Gynaecol. 2019;39:151–6.

    Article  CAS  PubMed  Google Scholar 

  44. Liu J, Song G, Zhao G, Meng T. Lack of association between IGF2BP2 rs4402960 polymorphism and gestational diabetes mellitus: a case-control study, meta-analysis and trial sequential analysis. Biosci Rep. 2020;40.

  45. Mtiraoui N, Turki A, Nemr R, Echtay A, Izzidi I, Al-Zaben GS, et al. Contribution of common variants of ENPP1, IGF2BP2, KCNJ11, MLXIPL, PPARγ, SLC30A8 and TCF7L2 to the risk of type 2 diabetes in Lebanese and Tunisian Arabs. Diabetes Metab. 2012;38:444–9.

    Article  CAS  PubMed  Google Scholar 

  46. Vatankhah Yazdi K, Kalantar SM, Houshmand M, Rahmanian M, Manaviat MR, Jahani MR. SLC30A8, CDKAL1, TCF7L2, KCNQ1 and IGF2BP2 are associated with type 2 diabetes mellitus in Iranian patients. DMSO. 2020;13:897–906.

    Article  Google Scholar 

  47. Duesing K, Fatemifar G, Charpentier G, Marre M, Tichet J, Hercberg S, et al. Evaluation of the association of IGF2BP2 variants with type 2 diabetes in French Caucasians. Diabetes. 2008;57:1992–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Rong R, Hanson RL, Ortiz D, Wiedrich C, Kobes S, Knowler WC, et al. Association analysis of variation in/near FTO, CDKAL1, SLC30A8, HHEX, EXT2, IGF2BP2, LOC387761, and CDKN2B with type 2 diabetes and related quantitative traits in Pima Indians. Diabetes. 2009;58:478–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Kommoju UJ, Maruda J, Kadarkarai S, Irgam K, Kotla JP, Velaga L, et al. No detectable association of IGF2BP2 and SLC30A8 genes with type 2 diabetes in the population of Hyderabad. India Meta Gene. 2013;1:15–23.

    Article  PubMed  Google Scholar 

  50. Wu HH, Liu NJ, Yang Z, Tao XM, Du YP, Wang XC, et al. IGF2BP2 and obesity interaction analysis for type 2 diabetes mellitus in Chinese Han population. Eur J Med Res. 2014;19:40.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Chistiakov DA, Nikitin AG, Smetanina SA, Bel’chikova LN, Suplotova LA, Shestakova MV, et al. The rs11705701 G>A polymorphism of IGF2BP2 is associated with IGF2BP2 mRNA and protein levels in the visceral adipose tissue—a link to type 2 diabetes susceptibility. Rev Diabet Stud. 2012;9:112–22.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Zhao Y, Ma YS, Fang Y, Liu L, Wu SD, Fu D, et al. IGF2BP2 genetic variation and type 2 diabetes: a global meta-analysis. DNA Cell Biol. 2012;31:713–20.

    Article  CAS  PubMed  Google Scholar 

  53. Li X, Allayee H, Xiang AH, Trigo E, Hartiala J, Lawrence JM, et al. Variation in IGF2BP2 interacts with adiposity to alter insulin sensitivity in Mexican Americans. Obesity (Silver Spring). 2009;17:729–36.

    Article  PubMed Central  CAS  Google Scholar 

  54. Han L, Li Y, Tang L, Chen Z, Zhang T, Chen S, et al. IGF2BP2 rs11705701 polymorphisms are associated with prediabetes in a Chinese population: a population-based case–control study. Exp Ther Med. 2016;12:1849–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Huang T, Wang L, Bai M, Zheng J, Yuan D, He Y, et al. Influence of IGF2BP2, HMG20A, and HNF1B genetic polymorphisms on the susceptibility to Type 2 diabetes mellitus in Chinese Han population. Biosci Rep. 2020;40.

  56. Salem SD, Saif-Ali R, Ismail IS, Al-Hamodi Z, Poh R, Muniandy S. IGF2BP2 alternative variants associated with glutamic acid decarboxylase antibodies negative diabetes in Malaysian subjects. Nat Commun. 2012;7:e45573.

    CAS  Google Scholar 

  57. Flyvbjerg A. The role of the complement system in diabetic nephropathy. Nat Rev Nephrol. 2017;13:311–8.

    Article  CAS  PubMed  Google Scholar 

  58. Jarad G, Cunningham J, Shaw AS, Miner JH. Proteinuria precedes podocyte abnormalities inLamb2/− mice, implicating the glomerular basement membrane as an albumin barrier. J Clin Invest. 2006;116:2272–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Schaeffer V, Hansen KM, Morris DR, LeBoeuf RC, Abrass CK. RNA-binding protein IGF2BP2/IMP2 is required for laminin-β2 mRNA translation and is modulated by glucose concentration. Am J Physiol Renal Physiol. 2012;303:F75-82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Bach LA, Hale LJ. Insulin-like growth factors and kidney disease. Am J Kidney Dis. 2015;65:327–36.

    Article  CAS  PubMed  Google Scholar 

  61. Fu J, Lee K, Chuang PY, Liu Z, He JC. Glomerular endothelial cell injury and cross talk in diabetic kidney disease. Am J Physiol Renal Physiol. 2015;308:F287–97.

    Article  CAS  PubMed  Google Scholar 

  62. Jing F, Zhao J, Jing X, Lei G. Long noncoding RNA Airn protects podocytes from diabetic nephropathy lesions via binding to Igf2bp2 and facilitating translation of Igf2 and Lamb2. Cell Biol Int. 2020;44:1860.

    Article  CAS  PubMed  Google Scholar 

  63. Laggai S, Kessler SM, Boettcher S, Lebrun V, Gemperlein K, Lederer E, et al. The IGF2 mRNA binding protein p62/IGF2BP2-2 induces fatty acid elongation as a critical feature of steatosis. J Lipid Res. 2014;55:1087–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Czepukojc B, Abuhaliema A, Barghash A, Tierling S, Naß N, Simon Y, et al. IGF2 mRNA binding protein 2 transgenic mice are more prone to develop a ductular reaction and to progress toward cirrhosis. Front Med (Lausanne). 2019;6:179.

    Article  PubMed Central  Google Scholar 

  65. He X, Li W, Liang X, Zhu X, Zhang L, Huang Y, et al. IGF2BP2 overexpression indicates poor survival in patients with acute myelocytic leukemia. Cell Physiol Biochem. 2018;51:1945–56.

    Article  CAS  PubMed  Google Scholar 

  66. Pu J, Wang J, Qin Z, Wang A, Zhang Y, Wu X, et al. IGF2BP2 promotes liver cancer growth through an m6A-FEN1-dependent mechanism. Front Oncol. 2020;10:578816.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Deng X, Jiang Q, Liu Z, Chen W. Clinical significance of an m6A reader gene, IGF2BP2, in head and neck squamous cell carcinoma. Front Mol Biosci. 2020;7:68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Huang S, Wu Z, Cheng Y, Wei W, Hao L. Insulin-like growth factor 2 mRNA binding protein 2 promotes aerobic glycolysis and cell proliferation in pancreatic ductal adenocarcinoma via stabilizing GLUT1 mRNA. Acta Biochim Biophys Sin (Shanghai). 2019;51:743–52.

    Article  CAS  Google Scholar 

  69. Dahlem C, Barghash A, Puchas P, Haybaeck J, Kessler SM. The insulin-like growth factor 2 mRNA binding protein IMP2/IGF2BP2 is overexpressed and correlates with poor survival in pancreatic cancer. IJMS. 2019;20:3204.

    Article  CAS  PubMed Central  Google Scholar 

  70. Hu X, Peng WX, Zhou H, Jiang J, Zhou X, Huang D, et al. IGF2BP2 regulates DANCR by serving as an N6-methyladenosine reader. Mol Cancer. 2020;27:1782–94.

    CAS  Google Scholar 

  71. Kessler SM, Lederer E, Laggai S, Golob-Schwarzl N, Hosseini K, Petzold J, et al. IMP2/IGF2BP2 expression, but not IMP1 and IMP3, predicts poor outcome in patients and high tumor growth rate in xenograft models of gallbladder cancer. Oncotarget. 2017;8:89736–45.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Liu G, Zhu T, Cui Y, Liu J, Liu J, Zhao Q, et al. Correlation between IGF2BP2 gene polymorphism and the risk of breast cancer in Chinese Han women. Biomed Pharmacother. 2015;69:297–300.

    Article  CAS  PubMed  Google Scholar 

  73. Liu W, Li Y, Wang B, Dai L, Qian W, Zhang JY. Autoimmune response to IGF2 mRNA-binding protein 2 (IMP2/p62) in breast cancer. Scand J Immunol. 2015;81:502–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. McMullen ER, Gonzalez ME, Skala SL, Tran M, Thomas D, Djomehri SI, et al. CCN6 regulates IGF2BP2 and HMGA2 signaling in metaplastic carcinomas of the breast. Brest Cancer Res Treat. 2018;172:577–86.

    Article  CAS  Google Scholar 

  75. Wang J, Xu W, Wang B, Lin G, Wei Y, Abudurexiti M, et al. GLUT1 is an AR target contributing to tumor growth and glycolysis in castration-resistant and enzalutamide-resistant prostate cancers. Cancer Lett. 2020;485:45–55.

    Article  CAS  PubMed  Google Scholar 

  76. Renaudin F, Orliaguet L. Gout and pseudo-gout-related crystals promote GLUT1-mediated glycolysis that governs NLRP3 and interleukin-1β activation on macrophages. Ann Rheum Dis. 2020;79:1506.

    Article  CAS  PubMed  Google Scholar 

  77. Ã…backa H, Hansen JS, Huang P, VenskutonytÄ— R, Hyrenius-Wittsten A, Poli G, et al. Targeting GLUT1 in acute myeloid leukemia to overcome cytarabine resistance. Ann Rheum Dis. 2020.

  78. Qiu H, Wang Y, Kang M, Ding H, Liu C, Tang W, et al. The relationship between IGF2BP2 and PPARG polymorphisms and susceptibility to esophageal squamous-cell carcinomas in the eastern Chinese Han population. Onco Targets Ther. 2017;10:5525–32.

    Article  PubMed  PubMed Central  Google Scholar 

  79. Tang W, Chen S, Liu J, Liu C, Wang Y, Kang M. Investigation of IGF1, IGF2BP2, and IGFBP3 variants with lymph node status and esophagogastric junction adenocarcinoma risk. J Cell Biochem. 2019;120:5510–8.

    Article  CAS  PubMed  Google Scholar 

  80. Liu X, Chen Z, Zhao X, Huang M, Wang C, Peng W, et al. Effects of IGF2BP2, KCNQ1 and GCKR polymorphisms on clinical outcome in metastatic gastric cancer treated with EOF regimen. Pharmacogenomics. 2015;16:959–70.

    Article  CAS  PubMed  Google Scholar 

  81. Ye S, Song W, Xu X, Zhao X, Yang L. IGF2BP2 promotes colorectal cancer cell proliferation and survival through interfering with RAF-1 degradation by miR-195. FEBS Lett. 2016;590:1641–50.

    Article  CAS  PubMed  Google Scholar 

  82. Mansoori B, Mohammadi A. HMGA2 and Bach-1 cooperate to promote breast cancer cell malignancy. J Cell Physiol. 2019;234:17714–26.

    Article  CAS  PubMed  Google Scholar 

  83. Kazemi T, Mokhtarzadeh A, Gjerstorff MF, Baradaran B, Dai FQ, Li CR, et al. miR-150-5p inhibits non-small-cell lung cancer metastasis and recurrence by targeting HMGA2 and β-catenin signaling. Expert Opin Ther Targets. 2019;16:675–85.

    Google Scholar 

  84. Mansoori B, Duijf PHG, Mohammadi A, Najafi S, Roshani E, Shanehbandi D, et al. Overexpression of HMGA2 in breast cancer promotes cell proliferation, migration, invasion and stemness. Expert Opin Ther Targets. 2020: 1–11.

  85. Li Z, Zhang Y, Ramanujan K, Ma Y, Kirsch DG, Glass DJ. Oncogenic NRAS, required for pathogenesis of embryonic rhabdomyosarcoma, relies upon the HMGA2-IGF2BP2 pathway. Cancer Res. 2013;73:3041–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Oliveira-Mateos C, Sánchez-Castillo A, Soler M. The transcribed pseudogene RPSAP52 enhances the oncofetal HMGA2-IGF2BP2-RAS axis through LIN28B-dependent and independent let-7 inhibition. Int J Mol Sci. 2019;10:3979.

    Google Scholar 

  87. Petri BJ, Klinge CM. Regulation of breast cancer metastasis signaling by miRNAs. Cancer Metastasis Rev. 2020;39:837–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Wang JY, Zhang Q, Wang DD, Yan W, Sha HH, Zhao JH, et al. MiR-29a: a potential therapeutic target and promising biomarker in tumors. Biosci Rep. 2018;38:BSR20171265.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Wang JY, Chen LJ. The role of microRNAs in the invasion and metastasis of cervical cancer. Biosci Rep. 2019;39:BSR20181367.

    Article  PubMed  PubMed Central  Google Scholar 

  90. Wang J, Zhang Q, Wang D, Yang S, Zhou S, Xu H, et al. Microenvironment-induced TIMP2 loss by cancer-secreted exosomal miR-4443 promotes liver metastasis of breast cancer. J Cell Physiol. 2020;235:5722–35.

    Article  CAS  PubMed  Google Scholar 

  91. Ding L, Wang L, Guo F. microRNA-188 acts as a tumour suppressor in glioma by directly targeting the IGF2BP2 gene. Mol Med Rep. 2017;16:7124–30.

    Article  CAS  PubMed  Google Scholar 

  92. Liu FY, Zhou SJ, Deng YL, Zhang ZY, Zhang EL, Wu ZB, et al. MiR-216b is involved in pathogenesis and progression of hepatocellular carcinoma through HBx-miR-216b-IGF2BP2 signaling pathway. Cell Death Dis. 2015;6:e1670.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Zaher MS, Rashid F, Song B, Joudeh LI, Sobhy MA, Tehseen M, et al. Missed cleavage opportunities by FEN1 lead to Okazaki fragment maturation via the long-flap pathway. Nucleic Acids Res. 2018;46:2956–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Liu S, Li Q, Chen K, Zhang Q, Li G, Zhuo L, et al. The emerging molecular mechanism of m(6)A modulators in tumorigenesis and cancer progression. Biomed Pharmacother. 2020;127:110098.

    Article  CAS  PubMed  Google Scholar 

  95. Huang RS, Zheng YL, Li C, Ding C, Xu C, Zhao J. MicroRNA-485-5p suppresses growth and metastasis in non-small cell lung cancer cells by targeting IGF2BP2. Life Sci. 2018;199:104–11.

    Article  CAS  PubMed  Google Scholar 

  96. Wang JY, Lu AQ, Chen LJ. LncRNAs in ovarian cancer. Clin Chim Acta. 2018;490:17.

    Article  PubMed  CAS  Google Scholar 

  97. Wang JY, Yang Y, Ma Y, Wang F, Xue A, Zhu J, et al. Potential regulatory role of lncRNA-miRNA-mRNA axis in osteosarcoma. Biomed Pharmacother. 2020;121:109627.

    Article  CAS  PubMed  Google Scholar 

  98. Qu X, Alsager S, Zhuo Y, Shan B. HOX transcript antisense RNA (HOTAIR) in cancer. Cancer Lett. 2019;454:90–7.

    Article  CAS  PubMed  Google Scholar 

  99. Cantile M, Di Bonito M, Cerrone M, Collina F. Long non-coding RNA HOTAIR in breast cancer therapy. Cancers. 2020;12:1197.

    Article  CAS  PubMed Central  Google Scholar 

  100. Wang Y, Lu JH, Wu QN, Jin Y, Wang DS, Chen YX, et al. LncRNA LINRIS stabilizes IGF2BP2 and promotes the aerobic glycolysis in colorectal cancer. Breast Cancer Res Treat. 2019;18:174.

    CAS  Google Scholar 

  101. Gao T, Liu X, He B, Nie Z, Zhu C, Zhang P, et al. Exosomal lncRNA 91H is associated with poor development in colorectal cancer by modifying HNRNPK expression. Cancer Cell Int. 2018;18:11.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  102. Yi T, Wang T, Shi Y, Peng X, Tang S, Zhong L, et al. Long noncoding RNA 91H overexpression contributes to the growth and metastasis of HCC by epigenetically positively regulating IGF2 expression. Liver Int. 2020;40:456–67.

    Article  CAS  PubMed  Google Scholar 

  103. Gao T, Liu X, He B, Pan Y, Wang S. Long non-coding RNA 91H regulates IGF2 expression by interacting with IGF2BP2 and promotes tumorigenesis in colorectal cancer. Artif Cells Nanomed Biotechnol. 2020;48:664–71.

    Article  CAS  PubMed  Google Scholar 

  104. Fen H, Hongmin Z, Wei W, Chao Y, Yang Y, Bei L, et al. RHPN1-AS1 drives the progression of hepatocellular carcinoma via regulating miR-596/IGF2BP2 axis. Curr Pharm Des. 2020;25:4630–40.

    Article  PubMed  CAS  Google Scholar 

  105. He L, Li H, Wu A, Peng Y, Shu G, Yin G. Functions of N6-methyladenosine and its role in cancer. Mol Cancer. 2019;18:176.

    Article  PubMed  PubMed Central  Google Scholar 

  106. Sun T, Wu R, Ming L. The role of m6A RNA methylation in cancer. Biomed Pharmacother. 2019;112:108613.

    Article  CAS  PubMed  Google Scholar 

  107. Wang P, Doxtader KA, Nam Y. Structural basis for cooperative function of Mettl3 and Mettl14 methyltransferases. Mol Cell. 2016;63:306–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Jin D, Guo J. m(6)A demethylase ALKBH5 inhibits tumor growth and metastasis by reducing YTHDFs-mediated YAP expression and inhibiting miR-107/LATS2-mediated YAP activity in NSCLC. Mol Cancer. 2020;19:40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Zheng W, Dong X, Zhao Y, Wang S, Jiang H, Zhang M, et al. Multiple functions and mechanisms underlying the role of METTL3 in human cancers. Front Oncol. 2019;9:1403.

    Article  PubMed  PubMed Central  Google Scholar 

  110. Pavlova NN, Thompson CB. The emerging hallmarks of cancer metabolism. Cell Metab. 2016;23:27–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Ma Y, Temkin SM, Hawkridge AM, Guo C, Wang W, Wang XY, et al. Fatty acid oxidation: an emerging facet of metabolic transformation in cancer. Cancer Lett. 2018;435:92–100.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science. 2009;324:1029–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Liu Z, Sun T, Zhang Z, Bi J, Kong C. An 18-gene signature based on glucose metabolism and DNA methylation improves prognostic prediction for urinary bladder cancer. Genomics. 2020.

  114. Ghanavat M, Shahrouzian M, Zayeri ZD, Banihashemi S, Kazemi SM, Saki N. Digging deeper through glucose metabolism and its regulators in cancer and metastasis. Life Sci. 2020;2020:118603.

    Google Scholar 

  115. Zhang M, Liu Q, Zhang M, Cao C, Liu X, Zhang M, et al. Enhanced antitumor effects of follicle-stimulating hormone receptor-mediated hexokinase-2 depletion on ovarian cancer mediated by a shift in glucose metabolism. J Nanobiotechnol. 2020;18:161.

    Article  CAS  Google Scholar 

  116. Assmann TS, Milagro FI, Martínez JA. Crosstalk between microRNAs, the putative target genes and the lncRNA network in metabolic diseases. Mol Med Rep. 2019;20:3543–54.

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This research was funded by the Natural Science Foundation of Jiangsu Province, Grant number BK20161110, Medical Research Grant of Jiangsu Commission of Health, Grant number M2020010, the Priority Academic Program Development of Jiangsu Higher Education Institutions, Grant number JX10231802, Postgraduate Research and Practice Innovation Program of Jiangsu Province, Grant number SJCX17_0387, the Science Foundation of Jiangsu Health vocational college, Grant number JKC201948, the Science and Technology Development Fund of Nanjing Medical University, Grant number NMUB2019235, the Nanjing health science and Technology Development Fund, Grant number YKK18201, Science and technology development fund of Nanjing Medical University, Grant number 2017NJMUZD091, the Research and development fund of Kangda College of Nanjing Medical University, Grant number KD2017KYJJYB017, Jiangsu province maternal and child health association research project, Grant number ZKY201737 and the Jiangsu Provincial Maternal and Child Health Fund, Grant number F201843.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization of the research: JW and LC. Writing of the original draft: JW. Writing review and editing of the manuscript: JW, LC and PQ. Supervision of research work: LC and PQ. Soliciting for funding: JW and PQ. All authors have read and agreed on the submission of the final manuscript.

Corresponding authors

Correspondence to Lijuan Chen or Ping Qiang.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, J., Chen, L. & Qiang, P. The role of IGF2BP2, an m6A reader gene, in human metabolic diseases and cancers. Cancer Cell Int 21, 99 (2021). https://doi.org/10.1186/s12935-021-01799-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12935-021-01799-x

Keywords