Skip to main content

The regulation of LRPs by miRNAs in cancer: influencing cancer characteristics and responses to treatment

Abstract

The low-density lipoprotein receptor-related protein (LRP) family is a group of cell surface receptors that participate in a variety of biological processes, including lipid metabolism, Wnt signaling, and bone metabolism. miRNAs are small non-coding RNA molecules that regulate gene expression and play a role in many biological processes, including the occurrence and development of tumors. Accumulating evidence demonstrates that LRP members are modulated by miRNAs across multiple cancer types, influencing key oncogenic processes—including tumor cell proliferation, apoptosis suppression, extracellular matrix remodeling, cell adhesion, and angiogenesis. The LRPs, miRNAs, their upstream lncRNAs, and downstream signaling molecules often form complex signaling pathways to regulate the activity of tumor cells. However, the tissue-specific roles and mechanistic underpinnings of these pathways remain incompletely understood. When examining the emerging concept of the interaction between miRNAs and LRPs, we emphasize the significance of these complex regulatory layers in the initiation and progression of cancer. Collectively, these findings are critical for advancing our understanding of the role of the LRPs family in the occurrence and development of tumors, as well as for the development of new strategies for cancer treatment.

Introduction

The low-density lipoprotein receptor-related protein (LRP) family comprises multiple members, including LRP1, LRP1b, LRP2, LRP4, LRP5/6, and LRP8. These transmembrane receptors are expressed on the cell surface and mediate diverse biological functions. Although LRPs have been extensively studied in oncology, their precise mechanisms of action in tumorigenesis remain largely elusive.

Preclinical studies suggest that LRP1 may attenuate cancer cell aggressiveness by downregulating matrix metalloproteinases (MMPs) and suppressing β-catenin signaling [1,2,3,4]. Additionally, LRP1 has been implicated in modulating cancer progression via the ERK1/2 pathway [4]. In contrast, LRP4 appears to promote tumor growth, migration, and invasion in gastric cancer (GC) and papillary thyroid cancer (PTC), likely through activation of the PI3K/AKT pathway [5, 6]. LRP5, a single-pass transmembrane coreceptor of the canonical Wnt signaling pathway, plays a pivotal role in tumorigenesis. By binding to Wnt ligands, LRP5 activates the Wnt/β-catenin signaling cascade through inhibition of GSK-3β, thereby promoting cell proliferation, differentiation, and epithelial-to-mesenchymal transition (EMT)– key processes driving primary tumor formation in various solid cancer [7]. Accumulating evidence demonstrates that LRP5 enhances tumorigenesis in GC [8] and in sporadic colorectal cancer [9], while also facilitating migration in ovarian cancer [10] and prostate cancer (PC) [11].

LRP6 is significantly upregulated in multiple malignancies, including hepatocellular carcinoma (HCC), retinoblastoma, breast cancer (BC), and prostate cancer (PC) [12, 13]. Similar to LRP5, LRP6 plays a crucial role in aberrant Wnt signaling activation. Furthermore, emerging evidence indicates that LRP6 contributes to cancer progression through alternative pathways, including the CXCL12/CXCR4 axis, KRAS signaling, and mTORC1-mediated regulation of oncogenic processes [14].

Notably, LRP8 overexpression has been identified across various cancer types, such as non-small cell lung cancer (NSCLC) [15], Triple-negative breast cancer (TNBC) [16], osteosarcoma [17], GC [18], ovarian cancer [19] and its elevated expression levels have been notably linked to adverse clinical and pathological characteristics as well as an unfavorable prognosis. LRP8 appears to exert its oncogenic effects through multiple molecular pathways across various tumor types. Mechanistically, LRP8 induces ERK1/2 phosphorylation to promote cell cycle progression [20], while simultaneously potentiating Wnt signaling-mediated β-catenin accumulation [21]. Additionally, LRP8 activates STAT3 phosphorylation and subsequent nuclear signaling transduction, further contributing to its tumorigenic potential [17].

MicroRNAs (miRNAs) are small non-coding RNAs that post-transcriptionally regulate gene expression through complementary binding to the 3’ untranslated region (3’ UTR) of target mRNAs. This interaction typically induces mRNA degradation or translational repression, effectively silencing gene expression [22, 23]. As key epigenetic regulators, miRNAs play critical roles in various cellular processes and are particularly implicated in tumorigenesis, cancer progression, and clinical outcome prediction. Notably, global miRNA downregulation has been recognized as a hallmark feature of human malignancies [24, 25]. Emerging evidence indicates that multiple miRNAs regulate LRP family members during tumor progression. However, the precise regulatory mechanisms and functional relationships between specific miRNAs and individual LRP members remain to be fully elucidated and warrant further investigation.

The roles of miRNA-regulated LRPs in gastrointestinal tract cancers

Colorectal cancer

The expression of LPR1 in colon cancer has been first discovered in 2007, It mainly expresses in stromal fibroblast and at the invasion front. With increasing tumor stages, the expression of LRP1 was decreasing in that study [26]. A reduction in LRP1 levels independently forecasts inferior overall survival (OS) and progression-free survival (PFS) in colon cancer patients [27]. It serves as a predictive indicator for the recurrence of colorectal cancer as well [28]. In contrast to normal colonic mucosa and stroma, LRP1 expression is markedly reduced in adenocarcinoma cells, a phenomenon partly attributed to mutations within the LRP1 gene. Despite the presence of numerous CpG islands in the promoter region of the LRP1 gene, the methylation status of this region remains relatively low. So, the low expression of LRP1 is not caused by methylation [27], but by other epigenetic pre-transcriptional processes such as miRNAs. In HCT116 cells, the expression of the LRP1 protein is reduced by miR-103/107 mimics, while treatment with inhibitors targeting miR-103 and miR-107 leads to the restoration of LRP1 protein levels. Doxorubicin (Dox) at lethal doses upregulates miR-103/107 through the p53 pathway, leading to the suppression of The translation of LRP1 is directly modulated by targeting its 3’ UTR, which in turn results in cell death. Since LRP1 is the target gene of P53, the p53 is necessary for LRP1 expression. The p53-mediated miRNA regulatory pathway functions as a feedback loop that inhibits the translation of LRP1 transcripts, thereby promoting cell death [29]. Meanwhile, p53 can regulate HIF-1 and tumor angiogenesis through the transcriptional regulation of miR-107 in colon cancer [30]. miR-107 facilitates the growth, migration and invasiveness of colon cancer cells and suppresses their apoptosis through the miR-107-PRE3/4 pathway [31,32,33]. Furthermore, miRNA-107 has been identified as a downstream effector of the long non-coding RNA (LncRNA) MIR503HG [31] and circMETTL3 [33] in colon cancer (Fig. 1A). miR-103 shows higher expression levels in Pancreatic Cancers (PC) tissue than adjacent normal tissues [34]. Additionally, various research has highlighted the significance of miR-103/107 as a promoter of cancer progression [35,36,37]. However, some studies indicate that miR-103 may play different roles in tumor. By regulating G1/S transition, miR-103 inhibited intestinal crypt cells proliferation and survival [38]. miR-103 exhibits substantial downregulation in the blood samples of patients with early-stage colon cancer and may serve as a potential biomarker for the recurrence of this condition. miR-103 may play different roles in different types of specimens, which needs more experiments to prove [39].

Growing evidence underscores the pivotal function of LPR6 in colorectal cancer (CRC). Overexpression of LPR6 is observed in both colorectal cancer cell lines and in malignant human tissues [40]. LRP6 facilitates the invasive and metastatic capabilities of CRC by modulating cytoskeletal dynamics [41]. CD44 is overexpression in CRC and have a prognostic value. It is essential for the activation and proper membrane targeting of LRP6, functioning as a modulator within the Wnt/β-catenin signaling pathway [42]. LRP6 serves as a co-receptor for Wnt ligands, playing an indispensable role in the transmission of Wnt signals [43]. miR-92a has risen to prominence as a potential diagnostic indicator for CRC, as noted in reference [44]. This microRNA is known to stimulate cell proliferation [45], as well as the migration and invasiveness of CRC cells [46]. The level of miR-92a in tumor tissues correlates significantly with the likelihood of lymph node metastasis in CRC patients [47]. miR-92a induces stem cell-like characteristics in colorectal cancer cells through the stimulation of the Wnt/β-catenin signaling [48]. miR-92a induces the suppression of DKK3 through the interaction with Myc on the 3’UTR.DKK3 suppressed Wnt signaling by inhibiting the LRP6 levels. So miR-92a indirectly promoted the expression of LRP6 in CRC. LRP6, Wnt, Myc, and miR-92a are interconnected in a positive feedback loop that includes the inhibition of DKK3 in CRC [49]. It is shown that MALAT1 as a diagnostic, prognostic, metastases and therapy biomarker for CRC [50]. MALAT1 enhances the proliferation, invasion, and migration of CRC cells [51], while also diminishing their apoptosis and drug responsiveness [52], and boosting their tumorigenic potential [53] by modulating various signaling pathways and miRNAs [54]. Research has disclosed that MALAT1 is physically linked to the miR-15 family, which suppresses LRP6 expression by binding to the3’UTR of the LRP6 mRNA. This interaction augments the β-catenin signaling, resulting in increased transcription of downstream target genes such as RUNX2 [55]. miR-487b is downregulated in NSCLC tumor cell lines and reported as a Wnt inhibitors [56]. miR-487b acts as an oncosuppressor in CRC primarily by targeting key oncogenes including MYC, SUZ12, and KRAS [57]. miR-487b exerts a suppressive influence on the proliferation and invasive capabilities of CRC cells. It is found to be under expressed in CRC liver metastasis and serves as an independent prognostic indicator for 5-year OS. miR-487b diminishes the activity of the KRAS signaling pathway and curbs the WNT/β-catenin pathway by directly targeting LRP6 in CRC [58](Fig. 1B).

Fig. 1
figure 1

The miRNA/LRPSs axis in Colorectal cancer. (A) Under various stress conditions, LRP1, p53, and miR-103/107 constitute a feedback regulatory loop, modulating the apoptosis of colorectal cancer cells. Oncogenic lncRNAs and circRNAs repress miR-103/107, thereby enhancing LRP1 expression. (B) LRP6, miR-92, and DKK3 form a positive feedback loop in colorectal cancer, synergistically with oncogenic lncRNAs, to promote LRP6 expression. This enhancement facilitates increased invasion, metastasis, and chemoresistance

Pancreatic cancer

LRP1, a substantial multifunctional receptor on the cell surface, is found in pancreatic ductal adenocarcinoma (PDAC). Research indicates that elevated levels of LRP1 correlate with diminished survival rates and increased invasiveness in pancreatic adenocarcinoma [59]. It serves as a receptor for eHSP90α, facilitating its role in promoting metastasis through the activation of the AKT signaling pathway [59, 60]. In pancreatic cancer, PAI-1 enhances the malignant phenotype of cancer cells through the LRP-1/ERK/c-JUN pathway [61]. Other studies have reported the tumor inhibitory effect of LRP1. For instance, the reduction in expression of the MIF has been shown to slow the growth of PDAC cell xenografts and to suppress cell proliferation under both normoxic and hypoxic conditions. This regulatory effect is mediated by the MIF-p53-LRP1-uPAR signaling.

miR-429 belonged to miR-200 gene family it is located in the hypermethylated region of chromosome 1, and is low expressed in a variety of tumor cells [62]. Numerous investigations have uncovered the modulatory function of miRNA-429 in PDAC. miR-429 can be related to poor outcome of PDAC patients [63, 64]. miR-429 significantly suppresses cell viability and invasion of the PaCa-2 and BxCP3 cells by NT-3 [65]. miR-429 also suppresses the growth of PANC1 and SW1990 cell lines in vitro by directly targeting TBK1 [64]. miR-429 is negatively regulated by OIP5-AS1. It suppressed PDAC cell growth, migration and reversed EMT process by targeted to FOXD1 and inhibited ERK pathway [66, 67]. LRP1 were predicted to be target genes of miR-429 in PDAC [63]. As a gene targeted by miR-429, LRP1 could potentially serve as a biomarker for the clinical diagnosis of PDAC (Fig. 2A).

LRP5 is a surface protein on cells that facilitates the internalization of ligands. It plays a role in enhancing the canonical Wnt signaling pathway within cells and is crucial for insulin secretion triggered by glucose [68,69,70]. LRP5 has been proved overexpressed in metastatic pancreatic endocrine neoplasms as compared with non-metastatic pancreatic endocrine neoplasms [71]. LRP5/6 has been recognized as a target substrate for CDK14 in vivo [72]. The expression of CDK14 is notably increased in PDAC tissue samples. Phosphorylation of LRP5/6 by CDK14 is a crucial factor in the activation of the Wnt signaling pathway. In PDAC, miR-26b directly targets CDK14, suppressing its expression, along with the expression of phosphorylated LRP6. This leads to a reduction in the aggressiveness of cancer cells and a decrease in tumor growth both in vitro and in vivo [73]. miR-194 suppresses the expression of CDK14 by directly targeting it, thereby significantly reducing the protein levels of phosphorylated LRP6. This action results in the modulation of PDAC cell proliferation and migration [74]. Concurrently, the H19 modulates miR-194, which in turn has an antagonistic effect on the aforementioned factors. miR-194 has been correlated with the overall survival rates of pancreatic cancer patients [75, 76]. In contrast to the aforementioned research findings, overexpression of miR-194 may promote tumor growth and local invasion in an orthotopic pancreatic cancer mouse mode [77] (Fig. 2B). At present, there is no research to confirm whether LRP5/6 is directly regulated by miR-194 or miR-26b, and the role of miR-26b, miR-194 and LRP6 in pancreatic cancer still require further investigation.

Significant increases in FGD5-AS1 expression is observed in Pancreatic Cancers [78]. Elevated levels of FGD5-AS1 expression are associated with an unfavorable prognosis in patients with Pancreatic Cancers. FGD5-AS1 exhibits tumor promoting activities by activated STAT3/NF-κB signaling pathway [79]. FGD5-AS1 promotes cell proliferation and migration by sequestering miR-520a-3p [80]. FGD5-AS1 functions as a competing endogenous RNA (ceRNA) to enhance the expression of BHLHE40 through the interaction with miR-15a-5p in Pancreatic Cancers cells. BHLHE40, in turn, promotes the proliferation, migration, and apoptosis of these cells [81]. In Pancreatic Cancers, miR-577 serves as a downstream target of FGD5-AS1. It counteracts the proliferative effects of FGD5-AS1 by binding to the 3’UTR of the LRP6 gene, thereby modulating the Wnt/β-catenin signaling pathway [82]. Furthermore, miR-577 can be absorbed by LINC01094, which stimulates the proliferation and metastasis of PDAC both in vitro and in vivo by activating the PI3K/AKT signaling pathway [83]. circRNA_0007334 competitive adsorbs miR-577 to enhance the migration ability of pancreatic ductal adenocarcinoma cells [84]. Previous research has indicated that miR-454 can act as either an oncogene or a tumor suppressor, depending on the cancer type. In the case of HCC, miR-454 has been shown to enhance cell proliferation, invasion, and EMT. Additionally, miR-454 stimulates the growth of tumors engrafted with HepG2 cells in vivo. Thus, in the context of HCC, miR-454 behaves as an oncogene [85]. miR-454 stimulates the proliferation and invasion of PC cells by activating the WNT/β-catenin signaling pathway [86]. Conversely, miR-454 has a potent effect in reducing tumor weight and volume in vivo by disrupting the Wnt signaling pathway in ovarian cancer [87]. It inhibits the proliferation and invasiveness of ovarian cancer cells by targeting the E2F6 gene [88]. miR-454 functions as a suppressor in tumor growth, angiogenesis in PDAC, by inhibiting Wnt/β-catenin signaling through targeting LRP6 [89](Fig. 2C). Moreover, miR-454-overexpressing formed significantly less PDAC lung metastases than control cells.

Fig. 2
figure 2

The miRNA/LRPSs axis in Pancreatic cancer. (A) OIP5-AS1 upregulates LRP1 expression by inhibiting miR-429, thus augmenting invasion, metastasis, and chemoresistance in pancreatic cancer. (B) H19 enhances LRP5/6 expression by suppressing miR-194, leading to increased proliferation and migration in pancreatic cancer

C. Oncogenic lncRNAs and circRNAs diminish the suppressive effects of tumor-suppressor miRNAs on LRP6, thereby promoting LRP6 expression and enhancing cell proliferation, metastasis, and angiogenesis.

Hepatocellular cancer

While a growing body of research suggests a link between LRP1 and cancer progression, the exact function and specific mechanisms by which it influences various cancer types remain subjects of ongoing discussionFurther research is required to understand the role of LRP1 in HCC. Existing studies have indicated that reduced LRP1 levels are correlated with a poor prognosis for HCC patients following curative surgery. The suppression of LRP1 is found to increase the expression of MMP9, which in turn boosts the migration and invasiveness of HCC cells in vitro and escalates the rate of pulmonary metastasis in liver orthotopic tumors [3]. Previous study have reported that over expression of LRP4 is found in both HCC patients and a range of HCC cell lines. Moreover, LRP4 depletion also significantly reduces cellular proliferation and invasion ability [90]. overexpression of LRP4, is significantly associated with T stage, pathologic stage, vascular invasion, and poor prognosis for patients with HCC [91]. LRP4-MuSK signal is required in Agrin induces activation of YAP, and promoted liver cancer development [92].

Oncogenic function of lncRNA HUMT is revealed in TNBC. It is a metastasis-associated lncRNA and predicts poorer clinical prognosis [93, 94]. High expression of miR-455-5p significantly correlates with better overall survival in HCC tissues and blood exosomes [93]. miR-455-5p has the capability to inhibit tumor growth, colony formation, as well as the migration and invasiveness of cancer cells by disrupting the IGF-1R/AKT/GLUT1 pathway. The reduction expression of miR-455-5p linkes to poorer patient outcomes [95]. lncRNA HOXA-AS3 promotes the proliferation, migration, and invasion of HCC cells, modulates the cell cycle, and suppresses apoptosis via the regulatory axis involving miR-455-5p and PD-L1 [96]. Intriguingly, research has uncovered that HUMT, which is overexpressed in HCC, functions as a miRNA sponge for miRNA-455-5p, leading to an increase in LRP4 levels and thus facilitating the proliferation and metastasis of HCC [97](Fig. 3A).

Likewise, the stimulation of the Wnt/β-catenin pathway plays a crucial role in the development of hepatocellular carcinoma HCC. As the upstream of LRP6 receptor interacts with FZD family which have seven transmembrane receptors to activate the Wnt/β-catenin pathway, LRP6 may play a role in hepatocarcinogenesis through hyperactivation of the Wnt/β-catenin pathway. LRP6 is found to be overexpressed in HCC, and its stable overexpression in HCC cells leads to enhanced cell proliferation, migration, and invasion both in vitro and in vivo [98]. Elevated levels of LRP6 expression are linked to the aggressive characteristics and unfavorable outcomes in HCC. CCN2 binds with LRP6 and enhances the invasiveness, migration, and proliferation abilities in HCC through upregulating phosphorylation level of LRP6 [99]. Research has identified that ABCG1, an ATP-binding cassette transporter that facilitates tumor cell migration and invasion, is modulated by the LRP6-Wnt/β-catenin pathway in HCC [100]. Stimulation of the LRP6-Wnt/β-catenin pathway results in elevated levels and activity of FRMD5, a protein that plays a pivotal role in the growth, mobility, tumor formation, and spread of HCC cells [101].

It is found that miR-1269a is deregulated in HCC [102]. miR-1269a is a signature for differentiating HCC patients from the healthy control [103]. Survival analysis of clinical samples shows that miR-1269a is associated with prognosis in HCC [104]. miR-1269a suppresses the proliferation of HCC cells and induces apoptosis by repressing the expression of its target gene, LRP6 [105]. Decreased expression of miR-202 correlates with tumor dimensions, vascular invasion, and the TNM staging in HCC patients, as well as with diminished overall survival rates. miR-202 is capable of curbing cellular glucose uptake, lactate generation, and proliferation by targeting the gene HK2 [106]. miR-202 significantly inhibits cell proliferation, migration, invasion and EMT, as well as induced apoptosis and cell cycle arrest and prevented tumor formation in vivo by downregulating BCL2 expression [107]. Furthermore, miR-202 hinders the proliferation, tumorigenic potential, and cell cycle advancement in HCC cells by directly targeting LRP6 [108]. Research has shown that miR-432 levels are negatively associated with the expression levels of β-catenin and LRP6. By directly targeting the 3’ UTRs of LRP6, miR-432 reduces the activity of the Wnt/β-catenin pathway, thereby significantly inhibiting the proliferation of HCC cells [109]. A deficiency in miR-610 is observed in HCC cells and tissues, and this deficiency correlates with the survival rates of HCC patients. The downregulation of miR-610 reduces Wnt/β-catenin signaling by directly inhibiting LRP6, which in turn enhances the proliferation and tumorigenic capacity of HCC [110].

A multitude of studies have substantiated the oncogenic function of TMPO-AS1 in various types of cancer. TMPO-AS1 acts as an enhancer of the aggressive characteristics of HCC cells by sequestering miR-320a [111], miR-429 [112] and miR-329-3p [113]. TMPO-AS1 promotes HCC proliferation, metastasis, and EMT by increasing LRP6. Additionally, TMPO-AS serves as a miRNA sponge for miRNA-126-3p, thereby activating the miR-126-3p/LRP6/β-catenin pathway [114]. Being a direct target of miR-126-3p, the protein levels of LRP6 exhibit an inverse relationship with the expression of miR-126-3p, and it stimulates the metastasis and angiogenesis of HCC both in vitro and in vivo [115]. DLGAP1-AS1, functioning as a molecular sponge for miR-26a-5p and miR-26b-5, has been shown to contribute to the growth and metastasis of HCC. The target inhibitory effect of miR-26a/b-5p on LRP6 is reversed by DLGAP1-AS1. DLGAP1-AS1 facilitates the progression of HCC and EMT by positively modulating the activity of the Wnt/β-catenin pathway through LRP6 [116].

circFBLIM1 is upregulated in HCC, which may inhibit growth and invasion, and promote apoptosis in HCC through sponging miR-346 [117]. circFBLIM1 is also over expressed in HCC serum exosomes and HCC cells. It accelerates the advancement and glycolytic activity of HCC by serving as a sponge for miR-338, thereby leading to an upregulation of LRP6 [118](Fig. 3B).

Fig. 3
figure 3

The miRNA/LRPSs axis in Hepatocellular cancer. (A) LncRNAs stimulate the expression of LRP4 by suppressing miR-455-5p, which in turn promotes cell proliferation, metastasis, and cell cycle progression in hepatocellular carcinoma. (B) Multiple miRNAs have been identified to negatively regulate LRP6 in hepatocellular carcinoma. Oncogenic lncRNAs and circRNAs counteract these miRNAs, increasing LRP6 expression and thereby promoting cell proliferation, invasion, metastasis, migration, and angiogenesis

Gastric cancer

LRP4 exhibits elevated expression in gastric cancer (GC) and is associated with an unfavorable prognosis for GC patients. It enhances the migration, invasion, and EMT of GC cells by modulating the PI3K/AKT signaling pathway. LRP4 serves as a direct target for miR-140-5p, and its levels are inversely associated with the expression of. miR-140-5p in GC tissues [6]. The expression of miR-140-5p is notably reduced in GC tissues, and its levels are correlated with lymph node metastasis, TNM stage, and diminished overall survival rates in GC patients [119, 120]. miR-140-5p suppresses the proliferation and invasive capacity of GC cells by reducing the expression of WNT1 and β-catenin, and by directly targeting the 3’UTR of the YES1 gene [119]. miR-140-5p is potentially implicated in the mediation of resistance to 5-FU in GC via the regulatory axis involving SNHG20/miR-140-5p/NDRG3 [121](Fig. 4A).

Polymorphisms in the LRP5 gene are linked to an unfavorable prognosis and diminished response to first-line chemotherapy with the EOF regimen in individuals suffering from advanced GC [122]. LRP5 promotes proliferation, invasion, migration and EMT in vitro in GC cell through Hsp90ab1-LRP5 interaction, thereby activates of the AKT and Wnt/β- catenin signaling pathways [123]. LRP5 is found in higher concentrations in GC tissues and shows a positive correlation with the progression to advanced clinical stages and a poorer prognosis. It boosts the proliferation, invasiveness, and drug resistance of GC cells by activating the Wnt signaling pathway and promoting aerobic glycolysis [8, 124]. The expression of the long non-coding RNA SBF2-AS1 is elevated in GC tissues and is associated with more advanced clinical stages and a lower survival rate. SBF2-AS1 promotes GC progression via targeting miR-545/EMS1 pathway [125]. While another study shows that miR-545-3p could have a suppressive effect on osteogenesis via targeting LRP5 [126]. Intriguingly, in vitro SBF2-AS1 knockdown inhibits the Wnt/LRP5 signaling pathway [124](Fig. 4B).

Fig. 4
figure 4

The miRNA/LRPSs axis in Gastric cancer. (A) SNHG20 promotes LRP4 expression by suppressing miR-140-5p, enhancing proliferation, invasion, and migration in gastric cancer. (B) SBF2-AS1 enhances LRP5 expression by inhibiting miR-545, thereby promoting proliferation, invasion, EMT, and drug resistance in gastric cancer

Abnormal LRP8 expression have also been associated multiple digestive system tumors. An overabundance of LRP8 in Huh7 cells leads to a decrease in apoptosis and is a contributing factor to the resistance of HCC cells to sorafenib treatment [127]. LRP8 is high expression in pancreatic cancer, and contributed to cell cycle and cell proliferation through activating ERK1/2 pathway [20]. The antimigratory role of MPA is achieved through down-regulation of LRP8 in gastric cancer cell [128]. Furthermore, miR-142 suppresses the proliferation, migration, invasion, and EMT of GC cells in vitro, as well as tumor growth in vivo, by directly targeting LRP8 [129].

Esophageal cancer

The expression of LRP6 is increased in esophageal squamous cell carcinoma (ESCC). In addition, knockout of LRP6 inhibits migration, invasion and EMT of EC-109 and EC-9706 cells [130].lncRNA ESCCAL-1 is upregulated in ESCC for loss of methylation in its promoter [131]. It acts as a biomarker of poor prognosis, which exhibits promising diagnostic value [131,132,133]. ESCCAL-1 has been demonstrated to enhance the proliferation, migration, and invasion of ESCC cells while simultaneously inhibiting their apoptosis [134]. It also suppresses the ubiquitin-mediated degradation of Gal-1, thereby promoting the progression of the cell cycle [133]. The ablation of the ESCCAL-1 gene markedly curbs the in vivo growth of ESCC cells [131, 135]. ESCCAL-1 enhances the growth, migration, and invasion of ESCC by suppressing the miR-590/LRP6 pathway. Concurrently, LRP6, being a direct target of miR-590, intensifies the malignancy of cells via the activation of the Wnt/β-catenin signaling pathway in ESCC [136].

The roles of miRNA-regulated LRPs in breast cancer

Although LRP1 plays a role in inhibiting tumor development in many types of tumors, it may act as a different role in breast cancer (BC). The high expression of LRP1 could predict decreased overall survival [137]. LRP1 interacts with eHsp90α to regulate lymph angiogenesis by elevating the level of phosphorylated AKT [138]. eHsp90α-LRP1 complex activates EMT and migration in breast cancer cells through AKT, ERK and NF-κB pathway [139]. Outgrowth of lamellipodia protrusions is one of the characteristics of cancer cell migration and metastasis. LRP1 is capable of interacting with tPA to promote the formation of lamellipodia in breast cancer cells by triggering the NF-κB signaling pathway [140]. Acted as a receptor for secreted Hsp90α, LRP1 can inhibit hypoxia-induced apoptosis of breast cancer cells via ERK1/2 and the Akt pathways [141]. LRP1 facilitates tumor growth and the formation of new blood vessels, known as angiogenesis, in TNBC by modulating the TGF-β signaling pathway and the plasminogen/plasmin system [142]. LRP1 prompts migration and invasion of tumor cells in serum-free conditions by combined activating EGFR and the eHsp90α autocrine signaling [143].

It has been established that LRP6 is excessively expressed in BC [139]. Elevated levels of LRP6 expression are notably correlated with the status of HER-2 and Ki67. Patients with luminal B type BC who exhibit high LRP6 expression levels have significantly poorer survival rates compared to those with low LRP6 expression. LRP6 stimulates the formation of clones, invasion, and wound healing in MCF-7 and MCF-10 A cell lines. The suppression of LRP6 leads to the inhibition of xenograft growth [144]. Acting as a co-receptor for the Wnt/β-catenin pathway, LRP6 can enhance the progression of TNBC, as well as cell migration and invasion, by modulating the Wnt/β-catenin pathway [13].

miR-424 have been reported to be down-regulated in NSCLC, cervical cancer, ovarian cancer, prostate cancer and some digestive system tumor [145]. miR-424 regulates the cell cycle and cell proliferation probable by targeting CDK1, through the Hippo and ERK pathway [146]. The expression of miR-424 prompts invasion ability in extremely aggressive TNBC cell lines by direct targeting CDC42, thus inhibited tumorigenesis and metastasis in xenograft [147]. LRP6 is likely the most significant miR-424 target in the canonical wnt signaling. miR-424 exerted its function by reducing LRP6 mRNA levels and protein expression in BC cells [148]. In BC cell lines, the expression levels of miR-130a-3p are found to be reduced [149]. miR-130a-3p is also the target gene of several lncRNA. The expression of lncRNA HOTAIR was increased in BC. It associated with the metastasis in vitro and vivo and poor prognosis of patients through acting as a spong of endogenous miR-130a-3p [150]. Concurrently, the H19 hastens the proliferation, migration, and invasion of BC cells, and it also intensifies apoptosis through the miR-130a-3P/SATB1 pathway during the progression of BC [151]. Furthermore, miR-130a-3p hinders the proliferation, migration, and invasive capabilities of cells by specifically targeting the RAB5B gene [152]. Elevated levels of miR-130a-3p curb the proliferation, growth in the absence of attachment, and migratory behavior of TNBC cells by reducing the expression of WNT cascade genes, including LRP6 [149, 153]. Bioinformatic analysis suggests that LRP6 is likely a target gene for miR-130a-3p, which has the potential to repress the mRNA expression of LRP6 [153](Fig. 5).

Fig. 5
figure 5

The miRNA/LRPSs axis in breast cancer. miRNAs exert an inhibitory effect on the expression of LRP6, thereby impeding breast cancer progression. Conversely, lncRNAs can counteract the suppressive function of these miRNAs by downregulating their expression, leading to enhanced tumor proliferation, invasion, and migration in breast cancer

LRP8 is highly expressed in breast cancer, especially in ER−/HER2 + BC and TNBC [16, 154, 155]. Increased expression of LRP8 correlates with an unfavorable prognosis for BC patients [155]. LRP8 controlled cell survival, colony formation, cell cycle progression and tumorigenicity in a xenograft model in TNBC through canonical Wnt/β-catenin signaling pathway and MAPK pathways [16, 154]. Bioinformatics prediction and luciferase reporter assay confirms that miR-1262 is an upstream factor for LRP8 [155]. Previous studies find that overexpression of miR-1262 inhibited colon cancer [156], Gastric cardia adenocarcinoma (GCA) [157] and lung cancer [158]. miR-1262 diminishes the proliferative, clonogenic, invasive, and migratory abilities of BC cells, capabilities that are otherwise augmented by LRP8 [155].

The roles of miRNA-regulated LRPs in prostate cancer

Previous research indicated that LRP1B ranks among the top 10 genes most frequently absent in human cancer samples [159]. Meanwhile LRP1B is also one of the most recurrently mutated genes in prostate adenocarcinoma [160]. LRP1B mutations may have improved outcomes to ICI in many cancer types [161]. LRP1B can significantly inhibit growth and migration of colon cancer by interacted with DVL2 [162]. Diminished expression of LRP1B in renal cancer cells facilitates the processes of invasion, migration, and growth without the need for attachment [163]. miR-301b-3p exhibits increased levels in PC. A rise in miR-301b-3p expression correlates with a decrease in LRP1B mRNA levels within prostate cancer cells. miR-301b-3p is found to stimulate tumor progression, including growth, migration, and invasion, by directly suppressing LRP1B [164]. The expression of miR-500 is elevated in prostate cancer and is linked to adverse clinical outcomes for patients with the disease. Knock-down of miR-500 inhibits PC growth [165] In PC tissues and cells, miR-500 is observed to be excessively expressed, while the expression of LRP1B is notably reduced. miR-500 is implicated in promoting cell proliferation and the cell cycle progression by specifically targeting LRP1B within PC cells [166].

miR-455-5p functions as a suppressor of tumorigenesis across a range of cancers, curtailing cellular proliferation, as well as the migration and invasion. In the context of PC, miR-455-5p serves as a tumor suppressor, hindering the proliferation of PC cells and inducing apoptosis by activating and cleaving caspase 3, as well as by targeting the CCR5 gene [167]. miR-455-5p is significantly under-expressed in PC, and its reduced levels are correlated with a less favorable prognosis for patients with the disease. miR-455-5p impedes the migration and invasiveness of prostate cancer cells. The increased expression of miR-455-5p is known to repress the expression of LRP8, which has been confirmed as a target gene for miR-455-5p using the TargetScan Human 7.1 database. High levels of LRP8 expression correlate with a negative outcome for patients with prostate cancer [168].

The roles of miRNA-regulated LRPs in thyroid carcinoma

CpG island methylation and DNA copy number loss often occurs in LRP1B in thyroid carcinoma (TC). LRP1B functions to suppress tumorigenesis by curbing the growth and invasion of TC cells [169,170,171]. LRP1B expression is down-regulated in TC cells [172]. miR-548a-5p was overexpressed in cancer cell lines, and prompts tumor growth, cell invasion and MMP-2 reduction by targeting LRP1B in vitro and in vivo [173]. miR-196a-5p is up-regulated in TC tissue and showed an oncogenic role in TC cells [174]. It enhances the proliferation, migration, and invasion of TC cells through direct binding to the 3’UTR of the LRP1B gene [172] (Fig. 6A).

It has been documented that LRP4 is excessively expressed in PTC. The SNPs within the LRP4 gene significantly influence an individual’s genetic predisposition to PTC [175]. LRP4 influences the proliferation, migration, invasion, and EMT of cells through the activation of the PI3K/AKT pathway [5]. miR-199a-5p impedes tumor migration, invasion, and EMT in living organisms by reducing the expression of SNAI1 [176]. miR-199a-5p curbs cell migration and invasion through the downregulation of PD-L1 and Claudin-1 [177]. miR-199a-5p diminishes the viability of TC cells by reducing the proportion of cells in the G2/M and S phases [178]. LRP4 may be the target gene of miR-199a-5p predicted by the miRWalk database (version 2.0) [179] miR-429 exhibits reduced expression in TC tissues and cell lines. It inhibits the proliferation, migration, and invasion of cells, and also induces apoptosis in the TC cell line [180]. In thyroid carcinoma, miR-429 is the target gene of multiple lncRNAs. OIP5-AS1 enhances the proliferation, metastasis and inhibited the apoptosis via adsorbing miR-429 [181]. miR-429 is the target of RNF185-AS1. RNF185-AS1 enhances tumor growth both by sequestering miR-429, thereby facilitating the expression of LRP4 [182] (Fig. 6B).

LRP6 exert as highly tumor-promoting function by activating Wnt/β-catenin pathway in PTC [183]. miR-146b-5p is over expression and shows correlation with the clinicopathological status of PTC. miR-146b-5p stimulates cell proliferation, migration, invasion, and cell cycle advancement in vitro by directly binding to the CCDC6 [184]. The Wnt/β-catenin signaling pathway is pivotal in the progression of TC [185]. Different from other miRNA that targeted LRP6, miR-146b-5p played a role in promoting LRP6 expression. miR-146b-5p increases the LRP6 through directly targeted ZNRF3, which leading to the ubiquitination and degradation of LRP6 [183]. miR-1271 is under-expressed in PTC, where it impedes cell migration, invasion, proliferation, and EMT by targeting IRS1 and inhibiting the AKT pathway [186]. LRP6 has been identified as a direct target of miR-1271. In PTC, the upregulation of Circ_0011373 sequesters miR-1271, leading to increased expression of LRP6. This mechanism may potentially influence the cell cycle, migration, invasion, and apoptosis of PTC cells [187] (Fig. 6C).

Fig. 6
figure 6

The miRNA/LRPSs axis in thyroid carcinoma. (A) IQCH-AS1 upregulates the expression of LRP1B by suppressing miR-196a-5p, consequently promoting proliferation, invasion, and migration in thyroid carcinoma. (B) LncRNAs stimulate the expression of LRP4 by inhibiting miR-429, thereby enhancing tumor proliferation, invasion, and migration in thyroid carcinoma (C) In hepatocellular carcinoma, miRNAs have been identified to negatively regulate LRP6. Furthermore, the circRNA Circ_0011373 can inhibit miR-127, leading to increased LRP6 expression and promoting cell proliferation, invasion, migration, and EMT

The roles of miRNA-regulated LRPs in reproductive system cancers

The NCBI database indicates that LRP6 is abundantly expressed in placental tissues, yet it shows low expression in trophoblast cell lines when compared to JEG-3 gestational choriocarcinoma cells. The overexpression of LRP6 is found to enhance the proliferation and migration of trophoblast cells by upregulating the expression of MMP-2 and MMP-9, while also reducing the levels of tissue inhibitors of TIMP-1 and TIMP- 2. Conversely, miR-346 is more highly expressed in trophoblast cell lines than in JEG-3 gestational choriocarcinoma cells. Elevated levels of miR-346 are observed to suppress the proliferation of these cells and decrease their migration and invasion rates by directly targeting LRP6 in both JEG-3 and gestational choriocarcinoma cells [188].

An excess of LRP8 notably boosts the proliferation, migration, and invasive capabilities of ovarian cancer cells. There is an inverse relationship exists between the expression levels of miR-362-3p and LRP8 in ovarian cancer. miR-362-3p hinders the proliferation, migration, and invasion of ovarian cancer cells by directly targeting LRP8, which results in the downregulation of MMP-2, MMP-9, as well as integrins α5 and β1 [189]. Multiple investigations have uncovered miR-362-3p’s regulatory influence in ovarian cancer, showing that it is under-expressed in both epithelial ovarian cancer tissues and cell lines. It suppresses cell proliferation and migration through the downregulation of MyD88 expression [190]. miR-362-3p also impedes the growth and advancement of ovarian cancer by directly interacting with its target gene SERBP1 in vivo [191]. Intriguingly, MALAT1 could prompt proliferation, invasion, and SU chemoresistance, but inhibits apoptosis through interact with miR-362-3p in RCC [192]. Reducing the levels of MALAT1 can curb the proliferation and diminish the invasive and migratory capacities in HCC by targeting miR-362-3p [193]. MALAT1 could also bind miR-195 to up-regulate LRP6 expression in CRC [55]. Based on the above conclusions, miR-362-3p may perform as a regulator of LRP6 in ovarian tumors, but no relevant research has been found so far.

The roles of miRNA-regulated LRPs in melanoma

LRP1 level is highly elevated in melanoma tissues. LRP1 is indispensable in YAP-induced melanoma tumorigenesis in vitro and in vivo [194]. LRP1 plays a crucial role in PAI-1-induced FAK phosphorylation and the invasive behavior of macrophages in melanoma [195]. Moreover, LRP1 enhances melanoma cell proliferation and massive lung metastasis by activating ERK and MMP-9. It is also critical for drug resistance [196]. APOE2/LRP1 axis may play important roles in tumor growth, metastasis, and protein synthesis in melanoma [197].

miR-103 and miR-107 are miRNAs related to each other, with a difference of just one base in their 3’ regions [198]. They have different functions in different tissues. Research has shown that the miR-103/107 cluster promotes the mobility of CRC cells by directly targeting metastasis suppressors, including death-associated protein kinase DAPK and KLF4 [36], triggers a prolonged duration of Wnt/β-catenin signaling by targeting Axin2 [37]. miR-103/107 modulates cell proliferation via the PI3K/AKT signaling pathway, in part by directing its action towards the PTEN [199]. Additional research has assessed that miR-103/107 diminishes the functionality of P-gp thereby increasing the sensitivity of GC cells to the chemotherapeutic agent DOX by targeting Cav-1 [200]. In melanoma, miR-107 is significantly downregulated, miR-107 can reduce cell proliferation, migration and invasion by targeting POU3F2 in melanoma [201]. Moreover, LINC00662 facilitates the advancement of melanoma by engaging the miR-107/POU3F2 regulatory axis and stimulating the β-catenin pathway [202]. miR-103/107 reduces cell proliferation and induced cell apoptosis by targeting LRP1 [203]. The function of LRP1 in different melanoma cells may also be different. A study found miR-199a-3p, miR-199a-5p, and miR-1908 which predicted metastasis-free survival in melanoma promote metastasis, invasion, and angiogenesis of melanoma by targeting APOE3 and suppressing LRP1 signaling [204] (Fig. 7).

Fig. 7
figure 7

The miRNA/LRPSs axis in melanoma. LINC00662 promotes the expression of LRP1 by suppressing miR-107, which in turn enhances proliferation, invasion, metastasis, and drug resistance in melanoma

The roles of miRNA-regulated LRPs in glioblastoma

The expression of LRP1 is increased under hypoxia, it facilitates glioblastoma (GBM) motility and invasion in an AKT dependent manner [205]. LRP1 is expressed on mast cells (MCs) and is critical for migration of MCs induced by PAI-1 [206]. LRP1 is strongly expressed in the angiogenic part of the tumor, and glioblastoma cells. It acts as a regulator of CXCR3, which prompts tumor cell invasion [207]. LRP1 facilitates drug delivery system such as Au-DOX@PO-ANG due to its capacity to penetrate the blood-brain barrier and access the central nervous system [208]. The expression of LRP-1 is significantly higher in GBM, LRP-1 prompts cell survival, proliferative migration, and decreases apoptosis [209]. In GBM cell line, LRP1 and miR-124-3p could be identified as hypoxia biomarkers [210]. Whether LRP1 is regulated by miR-124 have not been reported. Recent studies have found that miR-124, miR-128 may inhibit the expression of LRP1 by targeting ELF4 [211].

miRNA-205 is markedly under-expressed in glioma, functioning as a suppressor of tumor growth by specifically targeting VEGF-A [212, 213]. The serum concentration of miR-205 is independently linked to overall survival rates and is identified as an individual diagnostic marker [214]. miR-205 inhibits cell migration, invasion and prevented EMT through inhibiting of the Akt/mTOR signaling pathway in GBM [215]. miR-205 is crucial in counteracting the self-renewal of glioma stem cells (GSCs) and their resistance to irradiation [212]. The 3’UTR of the LRP1 gene can be targeted by miR-205, which suppresses cell migration and invasion through the reduction of LRP1 expression [216](Fig. 8A).

LRP6 is highly expressed in gliomas, the expression of LRP6 links with overall survival in all glioblastoma [217, 218]. Elevated levels of LRP6 expression are recognized for initiating Wnt pathway activation, promoting cell proliferation, and contributing to the development of tumors in glioblastoma cells [219]. The expression of miR-513c is found to be reduced in tissues and cell lines of GBM. miR-513c suppresses the proliferation of GBM cells by directly targeting the 3’UTR of the LRP6 gene [220]. Additionally, miR-513c-5p facilitates the suppression of neuroblastoma cell proliferation, colony formation, and invasive capabilities by modulating the silencing of DLX6-AS1, it also causes cell cycle arrest and apoptosis through the PLK4 pathway in vitro [221]. LOC728196 acts as a molecular sponge to miR-513c, which in turn facilitates the growth, migration, and invasiveness of glioma cells by modulating the expression of TCF7 [222].

miR-137 exhibits reduced expression levels in glioblastoma, and its diminished levels correlate with an unfavorable prognostic profile for individuals afflicted with GBM [223]. miR-137 deduces GBM cell proliferation, invasion and angiogenesis by targeting a series of genes, including EZH2, Cox-2, CXCL12, SP1 and CSE1L [224,225,226,227,228]. miR-137 curbs the proliferation of glioblastoma and leads to G1 phase cell cycle arrest in glioblastoma multiforme cells [229]. miR-137 inhibits proliferation, migration and invasion of glioblastoma through Akt/mTOR signaling by targeting PTP4A3 [230]. miR-137 enhances cell growth and reduces cell apoptosis by targeting the 3’-UTR of the EGFR [231]. The downregulation of HOTAIRM1 expression impeded the proliferation and invasive capabilities of glioblastoma cells by serving as a sponge to miR-137 [227]. lncRNA HAS2-AS1 promotes proliferation of GBM cell and tumorigenesis of nude mouse by down-regulating of miR-137 [232]. miR-137 sensitized GBM cells to the TRAIL-mediated apoptosis by targeting XIAP [233]. LRP6 is a target gene of miR-137, which can suppress invasion, EMT and enhance the chemosensitivity of GBM cells to TMZ by reducing LRP6 expression, subsequently affecting β-catenin and its downstream signaling pathways [218].

LINC01094 exhibits elevated expression levels in tissues and cell lines of GBM [234, 235]. LINC01094 facilitates the proliferation, migration, and invasion of cells, and suppresses apoptosis by acting as a sponge for miR-126-5p [235] and miR-577 [234] in GBM. miR-577 is found to be under-expressed in GBM tumor samples and cell lines, where it inhibits GBM growth by directly targeting LRP6 and β-catenin [236]. miR-126 have been reported to regulate the expression of LRP6 in many tumors and non-tumor diseases [115, 237,238,239,240,241]. Whether LINC01094 can regulate LRP6 through miR-126 and miR-577 in GBM needs further study.

ADAMTS9AS1 is upregulated in GBM tissues and cell lines. ADAMTS9-AS1 influences the proliferation, apoptosis, migration, and stem-like properties of glioma cells [187]. ADAMTS9‑AS1 may have a complex regulatory effect on LRP family members. In GBM, ADAMTS9-AS1 is capable of interacting with miR-128 and miR-150, leading to the upregulation of the RAS/MAPK signaling pathway, as well as the LRP6 and Wnt pathways [242]. In breast cancer, ADAMTS9-AS1 can activate the JAK/ STAT signaling by binding miR-301b-3p [243]. miR-301b-3p could potentially enhance the proliferation, migration, and invasiveness of PC cells by targeting LRP1B [164]. For ADAMTS9-AS1 is under-expressed in PC and may serve as a prognostic indicator for the overall survival of PC patients. It is believed that ADAMTS9-AS1 could exert its tumor-suppressive role by modulating LRP1B in prostate cancer (Fig. 8B).

Fig. 8
figure 8

The miRNA/LRPSs axis in glioblastoma. reduce apoptosis in glioblastoma. (A) miR-429 inhibits the expression of LRP1, which can induce proliferation, migration, and survival of glioblastoma cells, while reducing apoptosis. (B) Tumor suppressor miRNAs in glioblastoma exert their inhibitory function on cancer progression by inhibiting LRP6. Oncogenic lncRNAs, on the other hand, augment LRP6 expression by suppressing these miRNAs, thereby inducing cell proliferation, tumorigenesis, EMT, invasion, and chemotherapy resistance, and reducing apoptosis in glioblastoma

The roles of miRNA-regulated LRPs in lung cancer

A study indicates that LRP5 is under-expressed in six out of seventeen cases of lung squamous cell carcinoma [244]. The LRP5 rs3736228 and rs64843 SNPs have been notably linked to an elevated risk for NSCLC and squamous cell carcinoma (SCC), respectively [245]. There was a robust inverse relationship between miR-375 expression levels and LRP5 in both Adenocarcinoma (AC) and Small Cell Lung Cancer (SCLC). Previous research has demonstrated that miR-375-3p inhibits osteogenesis by targeting the LRP5 gene in MC3T3-E1 cells [246]. In H82 cells, the luciferase activity of the reporter for the putative target sites within the 3’UTR of the LRP5 mRNA was not significantly reduced by miR-375, and the ectopic expression of miR-375 had minimal impact on the suppression of the LRP5 protein. It suggests that miR-375 may regulate LRP5 through indirect pathways [247]. Therefore, more experimental results are needed to confirm the regulatory mechanism of miR-375 on LRP5 in lung cancer.

The earliest research found that overexpression of LRP8 is observed in 11 of the 13 lung cancer samples. This result suggests that LRP8 may play an oncogenic role in lung cancer [248]. Previous studies have indicated that LRP8 correlates with adverse clinicopathological features and the prognosis of patients with NSCLC. LRP8 has been shown to enhance the proliferation of NSCLC cells both in vitro and in vivo, and it plays a role in the progression and metastasis of NSCLC by modulating the Wnt/β-catenin signaling pathway [15]. miR-30b-5p is significantly correlated with the overall survival rate of lung cancer, which can be utilized to develop a risk scoring model serving as a prognostic signature for lung cancer [249]. miR-30b-5p exhibits low expression in A549/DDP cells and enhances their sensitivity to DDP. It targets LRP8 in lung cancer, and the increased sensitivity of A549 cells to DDP induced by miR-30b-5p can be negated by the overexpression of LRP8 [250].

The roles of miRNA-regulated LRPs in other type of cancers

The expression LRP1 is strong in dermatofibrosarcoma protuberans (DFSP), but weak in dermatofibroma (DF), but is not seen in normal fibroblasts [251]. Previous research find miR-205 as the most differentially expressed miRNA on cutaneous squamous cell carcinoma (cSCC) and malignant skin cancer [252]. miR-205 prompts cell cycle arrest at the G2M phase in melanoma cell line [253] and suppressed the migration, invasion and proliferation of cancer cell [254]. LRP1 is a target of miR-205. In DFSP, the downregulation of miR-205 results in aberrant cell proliferation, which losing the ability to inhibit the expression of LRP1 and triggers the ERK pathway [251]. miR-196a directly targets LRP4 in neuroendocrine tumor cell lines CNDT2.5 and NCI-H727, and it modulates downstream genes associated with the WNT signaling pathway, including LRP5 and LRP6 [255].

Conclusion and remarks

In recent years, a multitude of scientific experiments have played a significant role in exploring new molecular mechanisms of tumors, developing new tumor treatment strategies, and researching new anti-tumor drugs. Studying the molecular pathways of tumors is of great significance for a deeper understanding of the biological characteristics and development mechanisms of tumors, as well as for guiding clinical treatment. The LRPs family, a group of transmembrane proteins, is crucial for cellular signal transduction and participates in a multitude of biological processes. A growing body of research indicates that the LRPs family is instrumental in tumor biology, with functions ranging from the facilitation or suppression of tumor cell proliferation, migration, and survival to the modulation of the tumor microenvironment. LRPs are involved in the regulation of many key oncogenes, and they also act as target genes for a range of microRNAs. Here, we have elaborated the role of miRNA in regulating LRP and its downstream genes in the pathogenesis of various human malignancies (Table 1). Progress in miRNA/LRP research in human oncology holds the promise of elucidating the intricate molecular controls of cancer, potentially pointing the way for novel avenues in cancer management and therapeutics.

Table 1 The regulatory roles of MiRNA in various human malignancies

Data availability

No datasets were generated or analysed during the current study.

Abbreviations

3’:

UTR 3’ untranslated region

5-FU:

5-fluorouracil

A549/DDP:

A cisplatin drug resistant cell line

ABCG1:

ATP-binding cassette transporter G1

AC:

Adenocarcinoma

ADAMTS9AS1:

ADAMTS9 Antisense RNA 1

AKT:

Protein kinase B

APOE:

Lipid transporter apolipoprotein E

Axin2:

Axis Inhibition Protein 2

BC:

Breast cancer

BCL2:

B-cell lymphoma/leukemia type 2

BHLHE40:

Basic helix-loop-helix domain containing, class b, 2

Cav-1:

Caveolin-1

CCDC6:

Coiled-coil domain containing 6

CDC42:

Cell division control 42

CDK1:

Cyclindependent kinase 1

CDK14:

Cyclin-dependent kinase 14

ceRNA:

Endogenous RNA

circFBLIM1:

CircRNA filamin binding LIM protein 1

Cox-2:

Cyclooxygenase-2

CRC:

Colorectal cancer

cSCC:

Cutaneous squamous cell carcinoma

CSE1L:

CSE1 chromosome segregation 1-like

CXCL12:

C-X-C motif ligand 12

CXCL12:

CXC chemokine ligand 12

CXCR3:

CXC chemokine receptor 3

CXCR4:

CXC chemokine receptor 4

DAPK:

Death-associated protein kinase

DF:

Dermatofibroma

DFSP:

Dermatofibrosarcoma protuberans

DKK3:

Dickkopf-3

DLGAP1-AS1:

Discs large associated protein 1 antisense RNA 1

DLX6-AS1:

Distal-less homeobox 6 antisense RNA 1

Dox:

Doxorubicin

DVL2:

Dishevelled segment polarity protein

E2F6:

E2F transcription factor 6

EGFR:

Epidermal growth factor receptor

eHsp90:

Hsp90 protein

eHSP90α:

Extracellular heat shock protein-90α

ELF4:

E74-like factor 4

EMS1:

Eleven-Nineteen Lysine-Rich Carcinoma-Related Gene 1

EMT:

Epithelial-to-mesenchymal

EOF:

Epirubicin, Oxaliplatin, and 5-Fluorouracil

ERK1/2:

Extracellular signal-regulated kinase1/2

ESCC:

Esophageal squamous cell carcinoma

ESCCAL-1:

Esophageal squamous cell carcinoma-associated lncRNA-1

EZH2:

Enhancer of zeste homolog 2

FAK:

Focal adhesion kinase

FGD5-AS1:

FYVE, RhoGEF, and PH domain containing 5 antisense RNA 1

FRMD5:

FERM domain containing 5

FZD:

Frizzled

Gal-1:

Galectin-1

GBM:

Glioblastoma

GC:

Gastric cancer

GLUT1:

Glucose transporter type 1

GSC:

Glioma stem cells

HAS2-AS1:

Hyaluronan Snthase 2 Antisense RNA 1

HCC:

Hepatocellular carcinoma

HER-2:

Human Epidermal Growth Factor Receptor 2

HIF-1:

Hypoxia-inducible factor-1

HOTAIR:

LncRNA HOX transcript antisense intergenic RNA

HOXA-AS3:

HOXA cluster antisense RNA 3“

HUMT:

Human Mitochondrial Translation

ICI:

Immune checkpoint inhibitors

IGF-1R:

Insulin-like growth factor 1 receptor

IQCH-AS1:

IQCH antisense RNA 1

IRS1:

Insulin receptor substrate 1

JAK:

Janus kinase

KLF4:

Krüppel-like factor 4

KRAS:

Kirsten ras oncogene

LINC01094:

Long intergenic non-protein coding RNA 1094

LncRNA:

Long non-coding RNA

LOC728196:

MIR34B and MIR34C host gene

LRPs:

Low-density lipoprotein receptor-related protein

MALAT1:

Metastasis associated with lung adenocarcinoma transcript 1

MAPK:

Mitogen-activated protein kinase

MCs:

Mast cells

MIF:

Macrophage migration inhibitory factor

miRNAs:

MicroRNAs

MMP-2:

Matrix metalloproteinase 2

MMP-9:

Matrix metalloproteinase 9

MPA:

Mycophenolic acid

mRNA:

Messenger RNA

mTORC1:

Mechanistic target of rapamycin complex 1

MyD88:

Myeloid differentiation factor88

NCBI:

National Center for Biotechnology Information

NDRG3:

N-myc downstream-regulated gene 3

NF-κB:

Nuclear factor-kappa B

NSCLC:

Non-Small Cell Lung Cancer

NT-3:

Neurotrophin-3

OIP5-AS1:

Opa Interacting Protein 5 Antisense RNA 1

OS:

Overall survival

PAI-1:

Plasminogen activator inhibitor-1

pak1:

P-21-activated kinase 1

PC:

Prostate cancer

PDAC:

Pancreatic ductal adenocarcinoma

PD-L1:

Programmed Death-Ligand 1p

PFS:

Progression-free survival

P-gp:

P-glycoprotein

PI3K:

Phosphoinositide 3-kinase

PLK4:

Polo-like kinase 4

POU3F2:

POU Class 3 Homeobox 2“

PPP2R1B:

Protein phosphatase 2

prdm14:

PR-domain containing 14

PRE3/4:

Proteasome subunit pre3/4

PTC:

Papillary thyroid cancer

PTEN:

Phosphatase and tensin homologue deleted on chromosome ten

PTP4A3:

Protein tyrosine phosphatases (PTP) 4A3

RAB5B:

Ras-related protein Rab-5B

RAS:

Renin-Angiotensin System

RCC:

Renal cell carcinoma

RNF185-AS1:

RNF185 Antisense RNA 1

RUNX2:

Runt-related transcription factor 2

SATB1:

Special AT-rich sequence-binding protein-1

SBF2-AS1:

SET-binding factor 2 antisense RNA1

SCC:

Squamous cell carcinoma

SCLC:

Small Cell Lung Cancer

SERBP1:

Serum amyloid a binding protein 1

SNAI1:

Snail family zinc finger 1

SNHG20:

Small nucleolar RNA host gene 20

SNPs:

Single nucleotide polymorphisms

SP1:

Specificity Protein 1

STAT:

Signal transducer and activator of transcription

SU:

Sunitinib

SUZ12:

Suppressor of Zeste 12

TC:

Thyroid carcinoma

TCF7:

Transcript tion factor 7

TIMP-1:

Tissue inhibitor of metalloproteinase-1

TIMP-2:

Tissue inhibitor of metalloproteinase-2

TMPO-AS1:

Thymopoietin associated lncRNA 1

TMZ:

Temozolomide

TNBC:

Triple-negative breast cancer

TNM:

Tumor-node-metastasis

tPA:

Tissue plasminogen activator

VEGF-A:

Vascular endothelial growth factor A

WNT:

Wingless-Type MMTV Integration Site Family

XIAP:

X-linked inhibitors of apoptosis protein

YAP:

Yes-associated protein

YES1:

Yamaguchi sarcoma viral oncogene homolog 1

ZNRF3:

Zinc RING finger 3

References

  1. Van Gool B, Dedieu S, Emonard H, Roebroek AJ. The matricellular receptor LRP1 forms an interface for signaling and endocytosis in modulation of the extracellular tumor environment. Front Pharmacol. 2015;6:271.

    PubMed  PubMed Central  Google Scholar 

  2. Gonias SL, Gaultier A, Jo M. Regulation of the urokinase receptor (uPAR) by LDL receptor-related protein-1 (LRP1). Curr Pharm Des. 2011;17(19):1962–9.

    Article  PubMed  CAS  Google Scholar 

  3. Huang XY, Shi GM, Devbhandari RP, Ke AW, Wang Y, Wang XY, et al. Low level of low-density lipoprotein receptor-related protein 1 predicts an unfavorable prognosis of hepatocellular carcinoma after curative resection. PLoS ONE. 2012;7(3):e32775.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Lindner I, Hemdan NY, Buchold M, Huse K, Bigl M, Oerlecke I, et al. Alpha2-macroglobulin inhibits the malignant properties of Astrocytoma cells by impeding beta-catenin signaling. Cancer Res. 2010;70(1):277–87.

    Article  PubMed  CAS  Google Scholar 

  5. Zhou X, Xia E, Bhandari A, Zheng C, Xiang J, Guan Y, et al. LRP4 promotes proliferation, migration, and invasion in papillary thyroid cancer. Biochem Biophys Res Commun. 2018;503(1):257–63.

    Article  PubMed  CAS  Google Scholar 

  6. Mao Z, Wang Z, Zhang S, Pu Y, Wang J, Zhang T, et al. LRP4 promotes migration and invasion of gastric cancer under the regulation of microRNA-140-5p. Cancer Biomark. 2020;29(2):245–53.

    Article  PubMed  CAS  Google Scholar 

  7. Joiner DM, Ke J, Zhong Z, Xu HE, Williams BO. LRP5 and LRP6 in development and disease. Trends Endocrinol Metab. 2013;24(1):31–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Nie X, Wang H, Wei X, Li L, Xue T, Fan L, et al. LRP5 promotes gastric Cancer via activating canonical Wnt/beta-Catenin and Glycolysis pathways. Am J Pathol. 2022;192(3):503–17.

    Article  PubMed  CAS  Google Scholar 

  9. Prabhu KS. The Selenoprotein P-LRP5/6-WNT3A complex promotes tumorigenesis in sporadic colorectal cancer. J Clin Invest. 2023;133(13).

  10. Hong J, Xie Z, Yang Z, Yang F, Liao H, Rao S, et al. Inactivation of Wnt-LRP5 signaling suppresses the proliferation and migration of ovarian cancer cells. Transl Cancer Res. 2021;10(5):2277–85.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Gan S, Qu F, Zhang X, Pan X, Xu D, Cui X, et al. LRP5 competes for SPOP binding to enhance tumorigenesis mediated by Daxx and PD-L1 in prostate cancer. Exp Cell Res. 2024;434(1):113857.

    Article  PubMed  CAS  Google Scholar 

  12. Lu W, Li Y. Salinomycin suppresses LRP6 expression and inhibits both Wnt/beta-catenin and mTORC1 signaling in breast and prostate cancer cells. J Cell Biochem. 2014;115(10):1799–807.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Ma J, Lu W, Chen D, Xu B, Li Y. Role of Wnt Co-Receptor LRP6 in triple negative breast Cancer cell migration and invasion. J Cell Biochem. 2017;118(9):2968–76.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Li L, Zhao L, Yang J, Zhou L. Multifaceted effects of LRP6 in cancer: exploring tumor development, immune modulation and targeted therapies. Med Oncol. 2024;41(7):180.

    Article  PubMed  CAS  Google Scholar 

  15. Fang Z, Zhong M, Zhou L, Le Y, Wang H, Fang Z. Low-density lipoprotein receptor-related protein 8 facilitates the proliferation and invasion of non-small cell lung cancer cells by regulating the Wnt/beta-catenin signaling pathway. Bioengineered. 2022;13(3):6807–18.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Lin CC, Lo MC, Moody R, Jiang H, Harouaka R, Stevers N et al. Corrigendum to Targeting LRP8 inhibits breast cancer stem cells in triple-negative breast cancer [Canc. Lett. 438 (2018, Dec 1) 165–173]. Cancer Lett. 2020;480:51.

  17. Zheng S, Wei Y, Jiang Y, Hao Y. LRP8 activates STAT3 to induce PD-L1 expression in osteosarcoma. Tumori. 2021;107(3):238–46.

    Article  PubMed  CAS  Google Scholar 

  18. Liu B, Bukhari I, Li F, Ren F, Xia X, Hu B et al. Enhanced LRP8 expression induced by Helicobacter pylori drives gastric cancer progression by facilitating beta-Catenin nuclear translocation. J Adv Res. 2024.

  19. Xu Y, Zhou Y, Yi X, Nie X. LRP8 promotes tumorigenesis in ovarian cancer through inhibiting p53 signaling. Cell Biol Int. 2024;48(5):626–37.

    Article  PubMed  CAS  Google Scholar 

  20. Du S, Wang H, Cai J, Ren R, Zhang W, Wei W, et al. Apolipoprotein E2 modulates cell cycle function to promote proliferation in pancreatic cancer cells via regulation of the c-Myc-p21(Waf1) signalling pathway. Biochem Cell Biol. 2020;98(2):191–202.

    Article  PubMed  CAS  Google Scholar 

  21. Zhang J, Zhang X, Zhang L, Zhou F, van Dinther M, Ten Dijke P. LRP8 mediates Wnt/beta-catenin signaling and controls osteoblast differentiation. J Bone Min Res. 2012;27(10):2065–74.

    Article  CAS  Google Scholar 

  22. Lin S, Gregory RI. MicroRNA biogenesis pathways in cancer. Nat Rev Cancer. 2015;15(6):321–33.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. van Kouwenhove M, Kedde M, Agami R. MicroRNA regulation by RNA-binding proteins and its implications for cancer. Nat Rev Cancer. 2011;11(9):644–56.

    Article  PubMed  Google Scholar 

  24. Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, et al. MicroRNA expression profiles classify human cancers. Nature. 2005;435(7043):834–8.

    Article  PubMed  CAS  Google Scholar 

  25. Volinia S, Calin GA, Liu CG, Ambs S, Cimmino A, Petrocca F, et al. A MicroRNA expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci U S A. 2006;103(7):2257–61.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Obermeyer K, Krueger S, Peters B, Falkenberg B, Roessner A, Rocken C. The expression of low density lipoprotein receptor-related protein in colorectal carcinoma. Oncol Rep. 2007;17(2):361–7.

    PubMed  CAS  Google Scholar 

  27. Boulagnon-Rombi C, Schneider C, Leandri C, Jeanne A, Grybek V, Bressenot AM, et al. LRP1 expression in colon cancer predicts clinical outcome. Oncotarget. 2018;9(10):8849–69.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Dasgupta N, Kumar Thakur B, Chakraborty A, Das S. Butyrate-Induced in vitro colonocyte differentiation network model identifies ITGB1, SYK, CDKN2A, CHAF1A, and LRP1 as the prognostic markers for colorectal Cancer recurrence. Nutr Cancer. 2019;71(2):257–71.

    Article  PubMed  CAS  Google Scholar 

  29. Leslie PL, Franklin DA, Liu Y, Zhang Y. p53 regulates the expression of LRP1 and apoptosis through a stress Intensity-Dependent MicroRNA feedback loop. Cell Rep. 2018;24(6):1484–95.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Yamakuchi M, Lotterman CD, Bao C, Hruban RH, Karim B, Mendell JT, et al. P53-induced microRNA-107 inhibits HIF-1 and tumor angiogenesis. Proc Natl Acad Sci U S A. 2010;107(14):6334–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Han H, Li H, Zhou J. Long non-coding RNA MIR503HG inhibits the proliferation, migration and invasion of colon cancer cells via miR-107/Par4 axis. Exp Cell Res. 2020;395(2):112205.

    Article  PubMed  CAS  Google Scholar 

  32. Liu F, Liu S, Ai F, Zhang D, Xiao Z, Nie X, et al. miR-107 promotes proliferation and inhibits apoptosis of Colon cancer cells by targeting prostate apoptosis Response-4 (Par4). Oncol Res. 2017;25(6):967–74.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Zhang F, Su T, Xiao M. RUNX3-regulated circrna METTL3 inhibits colorectal cancer proliferation and metastasis via miR-107/PER3 axis. Cell Death Dis. 2022;13(6):550.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Piepoli A, Tavano F, Copetti M, Mazza T, Palumbo O, Panza A, et al. Mirna expression profiles identify drivers in colorectal and pancreatic cancers. PLoS ONE. 2012;7(3):e33663.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Zhang Z, Wu S, Muhammad S, Ren Q, Sun C. miR-103/107 promote ER stress-mediated apoptosis via targeting the Wnt3a/beta-catenin/ATF6 pathway in preadipocytes. J Lipid Res. 2018;59(5):843–53.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Chen HY, Lin YM, Chung HC, Lang YD, Lin CJ, Huang J, et al. miR-103/107 promote metastasis of colorectal cancer by targeting the metastasis suppressors DAPK and KLF4. Cancer Res. 2012;72(14):3631–41.

    Article  PubMed  CAS  Google Scholar 

  37. Chen HY, Lang YD, Lin HN, Liu YR, Liao CC, Nana AW, et al. miR-103/107 prolong Wnt/beta-catenin signaling and colorectal cancer stemness by targeting Axin2. Sci Rep. 2019;9(1):9687.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Liao Y, Lonnerdal B. Global MicroRNA characterization reveals that miR-103 is involved in IGF-1 stimulated mouse intestinal cell proliferation. PLoS ONE. 2010;5(9):e12976.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Shivapurkar N, Weiner LM, Marshall JL, Madhavan S, Deslattes Mays A, Juhl H, et al. Recurrence of early stage colon cancer predicted by expression pattern of Circulating MicroRNAs. PLoS ONE. 2014;9(1):e84686.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Rismani E, Fazeli MS, Mahmoodzadeh H, Movassagh A, Azami S, Karimipoor M, et al. Pattern of LRP6 gene expression in tumoral tissues of colorectal cancer. Cancer Biomark. 2017;19(2):151–9.

    Article  PubMed  CAS  Google Scholar 

  41. Yao Q, An Y, Hou W, Cao YN, Yao MF, Ma NN, et al. LRP6 promotes invasion and metastasis of colorectal cancer through cytoskeleton dynamics. Oncotarget. 2017;8(65):109632–45.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Ghatak S, Hascall VC, Karamanos N, Markwald RR, Misra S. Chemotherapy induces feedback up-regulation of CD44v6 in colorectal cancer initiating cells through beta-catenin/MDR1 signaling to sustain chemoresistance. Front Oncol. 2022;12:906260.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Ren Q, Chen J, Liu Y. LRP5 and LRP6 in Wnt signaling: similarity and divergence. Front Cell Dev Biol. 2021;9:670960.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Yang X, Zeng Z, Hou Y, Yuan T, Gao C, Jia W, et al. MicroRNA-92a as a potential biomarker in diagnosis of colorectal cancer: a systematic review and meta-analysis. PLoS ONE. 2014;9(2):e88745.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Lv H, Zhang Z, Wang Y, Li C, Gong W, Wang X. MicroRNA-92a promotes colorectal Cancer cell growth and migration by inhibiting KLF4. Oncol Res. 2016;23(6):283–90.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Wei QD, Zheng WB, Sun K, Xue Q, Yang CZ, Li GX. MiR-92a promotes the invasion and migration of colorectal cancer by targeting RECK. Int J Clin Exp Pathol. 2019;12(5):1565–77.

    PubMed  PubMed Central  CAS  Google Scholar 

  47. Ke TW, Wei PL, Yeh KT, Chen WT, Cheng YW. MiR-92a promotes cell metastasis of colorectal Cancer through PTEN-Mediated PI3K/AKT pathway. Ann Surg Oncol. 2015;22(8):2649–55.

    Article  PubMed  Google Scholar 

  48. Zhang GJ, Li LF, Yang GD, Xia SS, Wang R, Leng ZW, et al. MiR-92a promotes stem cell-like properties by activating Wnt/beta-catenin signaling in colorectal cancer. Oncotarget. 2017;8(60):101760–70.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Sehgal P, Lanauze C, Wang X, Hayer KE, Torres-Diz M, Leu NA, et al. MYC hyperactivates Wnt signaling in APC/CTNNB1-Mutated colorectal Cancer cells through miR-92a-Dependent repression of DKK3. Mol Cancer Res. 2021;19(12):2003–14.

    Article  PubMed  CAS  Google Scholar 

  50. Cervena K, Vodenkova S, Vymetalkova V. MALAT1 in colorectal cancer: its implication as a diagnostic, prognostic, and predictive biomarker. Gene. 2022;843:146791.

    Article  PubMed  CAS  Google Scholar 

  51. Xu Y, Zhang X, Hu X, Zhou W, Zhang P, Zhang J, et al. The effects of LncRNA MALAT1 on proliferation, invasion and migration in colorectal cancer through regulating SOX9. Mol Med. 2018;24(1):52.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Tang D, Yang Z, Long F, Luo L, Yang B, Zhu R, et al. Inhibition of MALAT1 reduces tumor growth and metastasis and promotes drug sensitivity in colorectal cancer. Cell Signal. 2019;57:21–8.

    Article  PubMed  CAS  Google Scholar 

  53. Chaleshi V, Irani S, Alebouyeh M, Mirfakhraie R, Aghdaei HA. Association of lncRNA-p53 regulatory network (lincRNA-p21, lincRNA-ROR and MALAT1) and p53 with the clinicopathological features of colorectal primary lesions and tumors. Oncol Lett. 2020;19(6):3937–49.

    PubMed  PubMed Central  CAS  Google Scholar 

  54. Xu WW, Jin J, Wu XY, Ren QL, Farzaneh M. MALAT1-related signaling pathways in colorectal cancer. Cancer Cell Int. 2022;22(1):126.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Ji Q, Cai G, Liu X, Zhang Y, Wang Y, Zhou L, et al. MALAT1 regulates the transcriptional and translational levels of proto-oncogene RUNX2 in colorectal cancer metastasis. Cell Death Dis. 2019;10(6):378.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Bartis D, Csongei V, Weich A, Kiss E, Barko S, Kovacs T, et al. Down-regulation of canonical and up-regulation of non-canonical Wnt signalling in the carcinogenic process of squamous cell lung carcinoma. PLoS ONE. 2013;8(3):e57393.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Chen X, Lin ZF, Xi WJ, Wang W, Zhang D, Yang F, et al. DNA methylation-regulated and tumor-suppressive roles of miR-487b in colorectal cancer via targeting MYC, SUZ12, and KRAS. Cancer Med. 2019;8(4):1694–709.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Hata T, Mokutani Y, Takahashi H, Inoue A, Munakata K, Nagata K, et al. Identification of microRNA-487b as a negative regulator of liver metastasis by regulation of KRAS in colorectal cancer. Int J Oncol. 2017;50(2):487–96.

    Article  PubMed  CAS  Google Scholar 

  59. Gheysarzadeh A, Ansari A, Emami MH, Razavi AE, Mofid MR. Over-expression of low-density lipoprotein receptor-related Protein-1 is associated with poor prognosis and invasion in pancreatic ductal adenocarcinoma. Pancreatology. 2019;19(3):429–35.

    Article  PubMed  CAS  Google Scholar 

  60. Xue N, Du T, Lai F, Jin J, Ji M, Chen X. Secreted HSP90alpha-LRP1 signaling promotes tumor metastasis and chemoresistance in pancreatic Cancer. Int J Mol Sci. 2022;23(10).

  61. Wang HC, Lin YL, Hsu CC, Chao YJ, Hou YC, Chiu TJ, et al. Pancreatic stellate cells activated by mutant KRAS-mediated PAI-1 upregulation foster pancreatic cancer progression via IL-8. Theranostics. 2019;9(24):7168–83.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Lynch SM, O’Neill KM, McKenna MM, Walsh CP, McKenna DJ. Regulation of miR-200c and miR-141 by methylation in prostate Cancer. Prostate. 2016;76(13):1146–59.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  63. Huang WT, Lin TS, Wu JY, Hong JM, Chen YL, Qiu FN. Evaluation of miR-429 as a novel serum biomarker for pancreatic ductal adenocarcinoma and analysis its tumor suppressor function and target genes. Eur Rev Med Pharmacol Sci. 2022;26(13):4638–53.

    PubMed  Google Scholar 

  64. Song B, Zheng K, Ma H, Liu A, Jing W, Shao C, et al. miR-429 determines poor outcome and inhibits pancreatic ductal adenocarcinoma growth by targeting TBK1. Cell Physiol Biochem. 2015;35(5):1846–56.

    Article  PubMed  CAS  Google Scholar 

  65. Liu D, Song L, Dai Z, Guan H, Kang H, Zhang Y, et al. MiR-429 suppresses neurotrophin-3 to alleviate perineural invasion of pancreatic cancer. Biochem Biophys Res Commun. 2018;505(4):1077–83.

    Article  PubMed  CAS  Google Scholar 

  66. Wu L, Liu Y, Guo C, Shao Y. LncRNA OIP5-AS1 promotes the malignancy of pancreatic ductal adenocarcinoma via regulating miR-429/FOXD1/ERK pathway. Cancer Cell Int. 2020;20:296.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Chen D, Zhao H. The inhibiting effects of microRNA-429 on the progression of pancreatic ductal adenocarcinoma cells by inhibiting epithelial mesenchymal transition. Am J Transl Res. 2021;13(4):3286–93.

    PubMed  PubMed Central  CAS  Google Scholar 

  68. Fujino T, Asaba H, Kang MJ, Ikeda Y, Sone H, Takada S, et al. Low-density lipoprotein receptor-related protein 5 (LRP5) is essential for normal cholesterol metabolism and glucose-induced insulin secretion. Proc Natl Acad Sci U S A. 2003;100(1):229–34.

    Article  PubMed  CAS  Google Scholar 

  69. Mu W, Wang Z, Zoller M. Ping-Pong-Tumor and host in pancreatic Cancer progression. Front Oncol. 2019;9:1359.

    Article  PubMed  PubMed Central  Google Scholar 

  70. Katoh M, Katoh M. Molecular genetics and targeted therapy of WNT-related human diseases (Review). Int J Mol Med. 2017;40(3):587–606.

    PubMed  PubMed Central  CAS  Google Scholar 

  71. Hansel DE, Rahman A, House M, Ashfaq R, Berg K, Yeo CJ, et al. Met proto-oncogene and insulin-like growth factor binding protein 3 overexpression correlates with metastatic ability in well-differentiated pancreatic endocrine neoplasms. Clin Cancer Res. 2004;10(18 Pt 1):6152–8.

    Article  PubMed  CAS  Google Scholar 

  72. Davidson G, Shen J, Huang YL, Su Y, Karaulanov E, Bartscherer K, et al. Cell cycle control of Wnt receptor activation. Dev Cell. 2009;17(6):788–99.

    Article  PubMed  CAS  Google Scholar 

  73. Sun Y, Wang P, Zhang Q, Wu H. CDK14/beta-catenin/TCF4/miR-26b positive feedback regulation modulating pancreatic cancer cell phenotypes in vitro and tumor growth in mice model in vivo. J Gene Med. 2022;24(2):e3343.

    Article  PubMed  CAS  Google Scholar 

  74. Sun Y, Zhu Q, Yang W, Shan Y, Yu Z, Zhang Q, et al. LncRNA H19/miR-194/PFTK1 axis modulates the cell proliferation and migration of pancreatic cancer. J Cell Biochem. 2019;120(3):3874–86.

    Article  PubMed  CAS  Google Scholar 

  75. Lee KH, Lee JK, Choi DW, Do IG, Sohn I, Jang KT, et al. Postoperative prognosis prediction of pancreatic cancer with seven MicroRNAs. Pancreas. 2015;44(5):764–8.

    Article  PubMed  CAS  Google Scholar 

  76. Schultz NA, Andersen KK, Roslind A, Willenbrock H, Wojdemann M, Johansen JS. Prognostic MicroRNAs in cancer tissue from patients operated for pancreatic cancer–five MicroRNAs in a prognostic index. World J Surg. 2012;36(11):2699–707.

    Article  PubMed  Google Scholar 

  77. Zhang J, Zhao CY, Zhang SH, Yu DH, Chen Y, Liu QH, et al. Upregulation of miR-194 contributes to tumor growth and progression in pancreatic ductal adenocarcinoma. Oncol Rep. 2014;31(3):1157–64.

    Article  PubMed  CAS  Google Scholar 

  78. Zhu L, Shi X, Yu X, Wang Z, Zhang M, He Y, et al. Expression and crucial role of long non-coding RNA FGD5-AS1 in human cancers. Am J Transl Res. 2021;13(10):10964–76.

    PubMed  PubMed Central  CAS  Google Scholar 

  79. He Z, Wang J, Zhu C, Xu J, Chen P, Jiang X, et al. Exosome-derived FGD5-AS1 promotes tumor-associated macrophage M2 polarization-mediated pancreatic cancer cell proliferation and metastasis. Cancer Lett. 2022;548:215751.

    Article  PubMed  CAS  Google Scholar 

  80. Lin J, Liao S, Liu Z, Li E, Wu X, Zeng W. LncRNA FGD5-AS1 accelerates cell proliferation in pancreatic cancer by regulating miR-520a-3p/KIAA1522 axis. Cancer Biol Ther. 2021;22(3):257–66.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  81. Qi W, Liu Q, Fu W, Shi J, Shi M, Duan S, et al. BHLHE40, a potential immune therapy target, regulated by FGD5-AS1/miR-15a-5p in pancreatic cancer. Sci Rep. 2023;13(1):16400.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  82. Zhang WT, Zhang JJ, Shao Q, Wang YK, Jia JP, Qian B et al. FGD5–AS1 is an oncogenic LncRNA in pancreatic cancer and regulates the Wnt/beta–catenin signaling pathway via miR–577. Oncol Rep. 2022;47(1).

  83. Luo C, Lin K, Hu C, Zhu X, Zhu J, Zhu Z. LINC01094 promotes pancreatic cancer progression by sponging miR-577 to regulate LIN28B expression and the PI3K/AKT pathway. Mol Ther Nucleic Acids. 2021;26:523–35.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  84. Yang J, Cong X, Ren M, Sun H, Liu T, Chen G, et al. Circular RNA Hsa_circRNA_0007334 is predicted to promote MMP7 and COL1A1 expression by functioning as a MiRNA sponge in pancreatic ductal adenocarcinoma. J Oncol. 2019;2019:7630894.

    Article  PubMed  PubMed Central  Google Scholar 

  85. Yu L, Gong X, Sun L, Yao H, Lu B, Zhu L. miR-454 functions as an oncogene by inhibiting CHD5 in hepatocellular carcinoma. Oncotarget. 2015;6(36):39225–34.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Fu Q, Gao Y, Yang F, Mao T, Sun Z, Wang H, et al. Suppression of microRNA-454 impedes the proliferation and invasion of prostate cancer cells by promoting N-myc downstream-regulated gene 2 and inhibiting WNT/beta-catenin signaling. Biomed Pharmacother. 2018;97:120–7.

    Article  PubMed  CAS  Google Scholar 

  87. Wang L, He M, Fu L, Jin Y. Exosomal release of microRNA-454 by breast cancer cells sustains biological properties of cancer stem cells via the PRRT2/Wnt axis in ovarian cancer. Life Sci. 2020;257:118024.

    Article  PubMed  CAS  Google Scholar 

  88. An Y, Zhang J, Cheng X, Li B, Tian Y, Zhang X, et al. miR-454 suppresses the proliferation and invasion of ovarian cancer by targeting E2F6. Cancer Cell Int. 2020;20:237.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  89. Fan Y, Shi C, Li T, Kuang T. microRNA-454 shows anti-angiogenic and anti-metastatic activity in pancreatic ductal adenocarcinoma by targeting LRP6. Am J Cancer Res. 2017;7(1):139–47.

    PubMed  PubMed Central  CAS  Google Scholar 

  90. Chakraborty S, Lakshmanan M, Swa HL, Chen J, Zhang X, Ong YS, et al. An oncogenic role of Agrin in regulating focal adhesion integrity in hepatocellular carcinoma. Nat Commun. 2015;6:6184.

    Article  PubMed  CAS  Google Scholar 

  91. Liu X, Zeng J, Li H, Li F, Jiang B, Zhao M, et al. A risk model based on Sorafenib-Response target genes predicts the prognosis of patients with HCC. J Oncol. 2022;2022:7257738.

    PubMed  PubMed Central  Google Scholar 

  92. Xiong WC, Mei L. Agrin to YAP in Cancer and neuromuscular junctions. Trends Cancer. 2017;3(4):247–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  93. Zheng S, Yang L, Zou Y, Liang JY, Liu P, Gao G, et al. Long non-coding RNA HUMT hypomethylation promotes lymphangiogenesis and metastasis via activating FOXK1 transcription in triple-negative breast cancer. J Hematol Oncol. 2020;13(1):17.

    Article  PubMed  PubMed Central  Google Scholar 

  94. Tuluhong D, Dunzhu W, Wang J, Chen T, Li H, Li Q, et al. Prognostic value of differentially expressed LncRNAs in Triple-Negative breast cancer: A systematic review and Meta-Analysis. Crit Rev Eukaryot Gene Expr. 2020;30(5):447–56.

    Article  PubMed  Google Scholar 

  95. Hu Y, Yang Z, Bao D, Ni JS, Lou J. miR-455-5p suppresses hepatocellular carcinoma cell growth and invasion via IGF-1R/AKT/GLUT1 pathway by targeting IGF-1R. Pathol Res Pract. 2019;215(12):152674.

    Article  PubMed  CAS  Google Scholar 

  96. Zeng C, Ye S, Chen Y, Zhang Q, Luo Y, Gai L, et al. HOXA-AS3 promotes proliferation and migration of hepatocellular carcinoma cells via the miR-455-5p/PD-L1 Axis. J Immunol Res. 2021;2021:9289719.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Zou X, Sun P, Xie H, Fan L, Ding K, Wang J, et al. Knockdown of long noncoding RNA HUMT inhibits the proliferation and metastasis by regulating miR-455-5p/LRP4 axis in hepatocellular carcinoma. Bioengineered. 2022;13(4):8051–63.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  98. Tung EK, Wong BY, Yau TO, Ng IO. Upregulation of the Wnt co-receptor LRP6 promotes hepatocarcinogenesis and enhances cell invasion. PLoS ONE. 2012;7(5):e36565.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  99. Jia Q, Bu Y, Wang Z, Chen B, Zhang Q, Yu S, et al. Maintenance of stemness is associated with the interation of LRP6 and heparin-binding protein CCN2 autocrined by hepatocellular carcinoma. J Exp Clin Cancer Res. 2017;36(1):117.

    Article  PubMed  PubMed Central  Google Scholar 

  100. Liao X, Zhang Y, Xu B, Ali A, Liu X, Jia Q. Inositol hexaphosphate sensitizes hepatocellular carcinoma to oxaliplatin relating Inhibition of CCN2-LRP6-beta-catenin-ABCG1 signaling pathway. J Cancer. 2021;12(20):6071–80.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  101. Mao X, Tey SK, Ko FCF, Kwong EML, Gao Y, Ng IO, et al. C-terminal truncated HBx protein activates caveolin-1/LRP6/beta-catenin/FRMD5 axis in promoting hepatocarcinogenesis. Cancer Lett. 2019;444:60–9.

    Article  PubMed  CAS  Google Scholar 

  102. Wojcicka A, Swierniak M, Kornasiewicz O, Gierlikowski W, Maciag M, Kolanowska M, et al. Next generation sequencing reveals MicroRNA isoforms in liver cirrhosis and hepatocellular carcinoma. Int J Biochem Cell Biol. 2014;53:208–17.

    Article  PubMed  CAS  Google Scholar 

  103. Dat VHX, Nhung BTH, Chau NNB, Cuong PH, Hieu VD, Linh NTM, et al. Identification of potential MicroRNA groups for the diagnosis of hepatocellular carcinoma (HCC) using microarray datasets and bioinformatics tools. Heliyon. 2022;8(2):e08987.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  104. Xie Y, Wang Y, Xue W, Zou H, Li K, Liu K, et al. Profiling and integrated analysis of differentially expressed MicroRNAs as novel biomarkers of hepatocellular carcinoma. Front Oncol. 2021;11:770918.

    Article  PubMed  CAS  Google Scholar 

  105. Min P, Li W, Zeng D, Ma Y, Xu D, Zheng W, et al. A single nucleotide variant in microRNA-1269a promotes the occurrence and process of hepatocellular carcinoma by targeting to oncogenes SPATS2L and LRP6. Bull Cancer. 2017;104(4):311–20.

    Article  PubMed  Google Scholar 

  106. Wang J, Chen J, Sun F, Wang Z, Xu W, Yu Y, et al. miR-202 functions as a tumor suppressor in hepatocellular carcinoma by targeting HK2. Oncol Lett. 2020;19(3):2265–71.

    PubMed  PubMed Central  CAS  Google Scholar 

  107. Zhuang D, Liang L, Zhang H, Feng X. miR-202 suppresses hepatocellular carcinoma progression via downregulating BCL2 expression. Oncol Res. 2020;28(4):399–408.

    Article  PubMed  PubMed Central  Google Scholar 

  108. Zhang Y, Zheng D, Xiong Y, Xue C, Chen G, Yan B, et al. miR-202 suppresses cell proliferation in human hepatocellular carcinoma by downregulating LRP6 post-transcriptionally. FEBS Lett. 2014;588(10):1913–20.

    Article  PubMed  CAS  Google Scholar 

  109. Jiang N, Chen WJ, Zhang JW, Xu C, Zeng XC, Zhang T, et al. Downregulation of miR-432 activates Wnt/beta-catenin signaling and promotes human hepatocellular carcinoma proliferation. Oncotarget. 2015;6(10):7866–79.

    Article  PubMed  PubMed Central  Google Scholar 

  110. Zhang G, Ai D, Yang X, Ji S, Wang Z, Feng S. MicroRNA-610 inhibits tumor growth of melanoma by targeting LRP6. Oncotarget. 2017;8(57):97361–70.

    Article  PubMed  PubMed Central  Google Scholar 

  111. Wang Z, Huang D, Huang J, Nie K, Li X, Yang X. LncRNA TMPO-AS1 exerts oncogenic roles in HCC through regulating miR-320a/SERBP1 Axis. Onco Targets Ther. 2020;13:6539–51.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  112. Liu X, Shen Z. LncRNA TMPO-AS1 aggravates the development of hepatocellular carcinoma via miR-429/GOT1 Axis. Am J Med Sci. 2020;360(6):711–20.

    Article  PubMed  Google Scholar 

  113. Guo X, Wang Y, LncRNA. TMPO-AS1 promotes hepatocellular carcinoma cell proliferation, migration and invasion through sponging miR-329-3p to stimulate FOXK1-mediated AKT/mTOR signaling pathway. Cancer Med. 2020;9(14):5235–46.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  114. Huang W, Chen Q, Dai J, Zhang Y, Yi Y, Wei X. Long noncoding TMPO antisense RNA 1 promotes hepatocellular carcinoma proliferation and epithelial-mesenchymal transition by targeting the microRNA-126-3p/LRP6/beta-catenin axis. Ann Transl Med. 2021;9(22):1679.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  115. Du C, Lv Z, Cao L, Ding C, Gyabaah OA, Xie H, et al. MiR-126-3p suppresses tumor metastasis and angiogenesis of hepatocellular carcinoma by targeting LRP6 and PIK3R2. J Transl Med. 2014;12:259.

    Article  PubMed  PubMed Central  Google Scholar 

  116. Lin Y, Jian Z, Jin H, Wei X, Zou X, Guan R, et al. Long non-coding RNA DLGAP1-AS1 facilitates tumorigenesis and epithelial-mesenchymal transition in hepatocellular carcinoma via the feedback loop of miR-26a/b-5p/IL-6/JAK2/STAT3 and Wnt/beta-catenin pathway. Cell Death Dis. 2020;11(1):34.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  117. Bai N, Peng E, Qiu X, Lyu N, Zhang Z, Tao Y, et al. circFBLIM1 act as a CeRNA to promote hepatocellular cancer progression by sponging miR-346. J Exp Clin Cancer Res. 2018;37(1):172.

    Article  PubMed  PubMed Central  Google Scholar 

  118. Lai Z, Wei T, Li Q, Wang X, Zhang Y, Zhang S. Exosomal circFBLIM1 promotes hepatocellular carcinoma progression and Glycolysis by regulating the miR-338/LRP6 Axis. Cancer Biother Radiopharm. 2023;38(10):674–83.

    PubMed  CAS  Google Scholar 

  119. Cha Y, He Y, Ouyang K, Xiong H, Li J, Yuan X. MicroRNA-140-5p suppresses cell proliferation and invasion in gastric cancer by targeting WNT1 in the WNT/beta-catenin signaling pathway. Oncol Lett. 2018;16(5):6369–76.

    PubMed  PubMed Central  CAS  Google Scholar 

  120. Fang Z, Yin S, Sun R, Zhang S, Fu M, Wu Y, et al. miR-140-5p suppresses the proliferation, migration and invasion of gastric cancer by regulating YES1. Mol Cancer. 2017;16(1):139.

    Article  PubMed  PubMed Central  Google Scholar 

  121. Yu J, Shen J, Qiao X, Cao L, Yang Z, Ye H, et al. SNHG20/miR-140-5p/NDRG3 axis contributes to 5-fluorouracil resistance in gastric cancer. Oncol Lett. 2019;18(2):1337–43.

    PubMed  PubMed Central  CAS  Google Scholar 

  122. Liu X, Huang MZ, Chen ZY, Zhao XY, Wang CC, Peng W, et al. LRP5 polymorphism-A potential predictor of the clinical outcome in advanced gastric cancer patients treated with EOF regimen. Chin J Cancer Res. 2014;26(4):478–85.

    PubMed  PubMed Central  CAS  Google Scholar 

  123. Wang H, Deng G, Ai M, Xu Z, Mou T, Yu J, et al. Hsp90ab1 stabilizes LRP5 to promote epithelial-mesenchymal transition via activating of AKT and Wnt/beta-catenin signaling pathways in gastric cancer progression. Oncogene. 2019;38(9):1489–507.

    Article  PubMed  CAS  Google Scholar 

  124. Liu Z, Li Q, Wang Y, Ge Y. Knockdown of LncRNA SBF2-AS1 inhibited gastric Cancer tumorigenesis via the Wnt/LRP5 signaling pathway. Acta Med Okayama. 2022;76(6):625–33.

    PubMed  CAS  Google Scholar 

  125. He M, Feng L, Qi L, Rao M, Zhu Y. Long noncoding RNASBF2-AS1 promotes gastric Cancer progression via regulating miR-545/EMS1 Axis. Biomed Res Int. 2020;2020:6590303.

    Article  PubMed  PubMed Central  Google Scholar 

  126. Li L, Qiu X, Sun Y, Zhang N, Wang L. SP1-stimulated miR-545-3p inhibits osteogenesis via targeting LRP5-activated Wnt/beta-catenin signaling. Biochem Biophys Res Commun. 2019;517(1):103–10.

    Article  PubMed  CAS  Google Scholar 

  127. Cai J, Chen J, Wu T, Cheng Z, Tian Y, Pu C, et al. Genome-scale CRISPR activation screening identifies a role of LRP8 in Sorafenib resistance in hepatocellular carcinoma. Biochem Biophys Res Commun. 2020;526(4):1170–6.

    Article  PubMed  CAS  Google Scholar 

  128. Dun B, Sharma A, Teng Y, Liu H, Purohit S, Xu H, et al. Mycophenolic acid inhibits migration and invasion of gastric cancer cells via multiple molecular pathways. PLoS ONE. 2013;8(11):e81702.

    Article  PubMed  PubMed Central  Google Scholar 

  129. Lu J, Ma Y, Zhao Z. MiR-142 suppresses progression of gastric carcinoma via directly targeting LRP8. Clin Res Hepatol Gastroenterol. 2021;45(4):101520.

    Article  PubMed  CAS  Google Scholar 

  130. Guan H, Liu J, Lv P, Zhou L, Zhang J, Cao W. MicroRNA–590 inhibits migration, invasion and epithelial–to–mesenchymal transition of esophageal squamous cell carcinoma by targeting low–density lipoprotein receptor–related protein 6. Oncol Rep. 2020;44(4):1385–92.

    PubMed  PubMed Central  CAS  Google Scholar 

  131. Cao W, Lee H, Wu W, Zaman A, McCorkle S, Yan M, et al. Multi-faceted epigenetic dysregulation of gene expression promotes esophageal squamous cell carcinoma. Nat Commun. 2020;11(1):3675.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  132. Cao W, Wu W, Shi F, Chen X, Wu L, Yang K, et al. Integrated analysis of long noncoding RNA and coding RNA expression in esophageal squamous cell carcinoma. Int J Genomics. 2013;2013:480534.

    Article  PubMed  PubMed Central  Google Scholar 

  133. Cui Y, Yan M, Wu W, Lv P, Wang J, Huo Y, et al. ESCCAL-1 promotes cell-cycle progression by interacting with and stabilizing galectin-1 in esophageal squamous cell carcinoma. NPJ Precis Oncol. 2022;6(1):12.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  134. Liu J, Mayekar MK, Wu W, Yan M, Guan H, Wang J, et al. Long non-coding RNA ESCCAL-1 promotes esophageal squamous cell carcinoma by down regulating the negative regulator of APOBEC3G. Cancer Lett. 2020;493:217–27.

    Article  PubMed  CAS  Google Scholar 

  135. Cui Y, Wu W, Lv P, Zhang J, Bai B, Cao W. Down-regulation of long non-coding RNA ESCCAL_1 inhibits tumor growth of esophageal squamous cell carcinoma in a xenograft mouse model. Oncotarget. 2018;9(1):783–90.

    Article  PubMed  Google Scholar 

  136. Guan H, Lv P, Han P, Zhou L, Liu J, Wu W, et al. Long non-coding RNA ESCCAL-1/miR-590/LRP6 signaling pathway participates in the progression of esophageal squamous cell carcinoma. Cancer Med. 2023;12(1):445–58.

    Article  PubMed  CAS  Google Scholar 

  137. Langer EM, Kendsersky ND, Daniel CJ, Kuziel GM, Pelz C, Murphy KM, et al. ZEB1-repressed MicroRNAs inhibit autocrine signaling that promotes vascular mimicry of breast cancer cells. Oncogene. 2018;37(8):1005–19.

    Article  PubMed  CAS  Google Scholar 

  138. Hou Q, Chen S, An Q, Li B, Fu Y, Luo Y. Extracellular Hsp90alpha promotes tumor lymphangiogenesis and lymph node metastasis in breast Cancer. Int J Mol Sci. 2021;22(14).

  139. Tian Y, Wang C, Chen S, Liu J, Fu Y, Luo Y. Extracellular Hsp90alpha and clusterin synergistically promote breast cancer epithelial-to-mesenchymal transition and metastasis via LRP1. J Cell Sci. 2019;132(15).

  140. Qiu Y, Wang H, Guo Q, Liu Y, He Y, Zhang G, et al. CD44s-activated tPA/LRP1-NFkappaB pathway drives lamellipodia outgrowth in luminal-type breast cancer cells. Front Cell Dev Biol. 2023;11:1224827.

    Article  PubMed  PubMed Central  Google Scholar 

  141. Dong H, Zou M, Bhatia A, Jayaprakash P, Hofman F, Ying Q, et al. Breast Cancer MDA-MB-231 cells use secreted heat shock Protein-90alpha (Hsp90alpha) to survive a hostile hypoxic environment. Sci Rep. 2016;6:20605.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  142. Campion O, Thevenard Devy J, Billottet C, Schneider C, Etique N, Dupuy JW et al. LRP-1 matricellular receptor involvement in triple negative breast Cancer tumor angiogenesis. Biomedicines. 2021;9(10).

  143. Chang C, Tang X, Mosallaei D, Chen M, Woodley DT, Schonthal AH, et al. LRP-1 receptor combines EGFR signalling and eHsp90alpha autocrine to support constitutive breast cancer cell motility in absence of blood supply. Sci Rep. 2022;12(1):12006.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  144. Zhang Y, Shu C, Maimaiti Y, Wang S, Lu C, Zhou J. LRP6 as a biomarker of poor prognosis of breast cancer. Gland Surg. 2021;10(8):2414–27.

    Article  PubMed  PubMed Central  Google Scholar 

  145. Ghafouri-Fard S, Askari A, Hussen BM, Taheri M, Akbari Dilmaghani N. Role of miR-424 in the carcinogenesis. Clin Transl Oncol. 2024;26(1):16–38.

    Article  PubMed  CAS  Google Scholar 

  146. Xie D, Song H, Wu T, Li D, Hua K, Xu H, et al. MicroRNA–424 serves an anti–oncogenic role by targeting cyclin–dependent kinase 1 in breast cancer cells. Oncol Rep. 2018;40(6):3416–26.

    PubMed  PubMed Central  CAS  Google Scholar 

  147. Nandy SB, Orozco A, Lopez-Valdez R, Roberts R, Subramani R, Arumugam A, et al. Glucose insult elicits hyperactivation of cancer stem cells through miR-424-cdc42-prdm14 signalling axis. Br J Cancer. 2017;117(11):1665–75.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  148. Nekritz EA, Rodriguez-Barrueco R, Yan KK, Davis ML, Werner RL, Devis-Jauregui L, et al. miR-424/503 modulates Wnt/beta-catenin signaling in the mammary epithelium by targeting LRP6. EMBO Rep. 2021;22(12):e53201.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  149. Poodineh J, Sirati-Sabet M, Rajabibazl M, Ghasemian M, Mohammadi-Yeganeh S. Downregulation of NRARP exerts anti-tumor activities in the breast tumor cells depending on Wnt/beta-catenin-mediated signals: the role of miR-130a-3p. Chem Biol Drug Des. 2022;100(3):334–45.

    Article  PubMed  CAS  Google Scholar 

  150. He W, Li D, Zhang X. LncRNA HOTAIR promotes the proliferation and invasion/metastasis of breast cancer cells by targeting the miR-130a-3p/Suv39H1 axis. Biochem Biophys Rep. 2022;30:101279.

    PubMed  PubMed Central  CAS  Google Scholar 

  151. Zhong G, Lin Y, Wang X, Wang K, Liu J, Wei W. H19 knockdown suppresses proliferation and induces apoptosis by regulating miR-130a-3p/SATB1 in breast Cancer cells. Onco Targets Ther. 2020;13:12501–13.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  152. Kong X, Zhang J, Li J, Shao J, Fang L. MiR-130a-3p inhibits migration and invasion by regulating RAB5B in human breast cancer stem cell-like cells. Biochem Biophys Res Commun. 2018;501(2):486–93.

    Article  PubMed  CAS  Google Scholar 

  153. Poodineh J, Sirati-Sabet M, Rajabibazl M, Mohammadi-Yeganeh S. MiR-130a-3p blocks Wnt signaling cascade in the triple-negative breast cancer by targeting the key players at multiple points. Heliyon. 2020;6(11):e05434.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  154. Maire V, Mahmood F, Rigaill G, Ye M, Brisson A, Nemati F, et al. LRP8 is overexpressed in estrogen-negative breast cancers and a potential target for these tumors. Cancer Med. 2019;8(1):325–36.

    Article  PubMed  CAS  Google Scholar 

  155. Li L, Qu WH, Ma HP, Wang LL, Zhang YB, Ma Y. LRP8, modulated by miR-1262, promotes tumour progression and forecasts the prognosis of patients in breast cancer. Arch Physiol Biochem. 2022;128(3):657–65.

    Article  PubMed  CAS  Google Scholar 

  156. Zhang W, Huang Z, Xiao Z, Wang H, Liao Q, Deng Z, et al. NF-kappaB downstream miR-1262 disturbs colon cancer cell malignant behaviors by targeting FGFR1. Acta Biochim Biophys Sin (Shanghai). 2023;55(11):1819–32.

    Article  PubMed  CAS  Google Scholar 

  157. Zheng Y, Xie M, Zhang N, Liu J, Song Y, Zhou L, et al. miR-1262 suppresses gastric cardia adenocarcinoma via targeting oncogene ULK1. J Cancer. 2021;12(4):1231–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  158. Xie K, Chen M, Zhu M, Wang C, Qin N, Liang C, et al. A polymorphism in miR-1262 regulatory region confers the risk of lung cancer in Chinese population. Int J Cancer. 2017;141(5):958–66.

    Article  PubMed  CAS  Google Scholar 

  159. Beroukhim R, Mermel CH, Porter D, Wei G, Raychaudhuri S, Donovan J, et al. The landscape of somatic copy-number alteration across human cancers. Nature. 2010;463(7283):899–905.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  160. Zhao X, Lei Y, Li G, Cheng Y, Yang H, Xie L, et al. Integrative analysis of cancer driver genes in prostate adenocarcinoma. Mol Med Rep. 2019;19(4):2707–15.

    PubMed  PubMed Central  CAS  Google Scholar 

  161. Brown LC, Tucker MD, Sedhom R, Schwartz EB, Zhu J, Kao C et al. LRP1B mutations are associated with favorable outcomes to immune checkpoint inhibitors across multiple cancer types. J Immunother Cancer. 2021;9(3).

  162. Wang Z, Sun P, Gao C, Chen J, Li J, Chen Z, et al. Down-regulation of LRP1B in colon cancer promoted the growth and migration of cancer cells. Exp Cell Res. 2017;357(1):1–8.

    Article  PubMed  CAS  Google Scholar 

  163. Ni S, Hu J, Duan Y, Shi S, Li R, Wu H, et al. Down expression of LRP1B promotes cell migration via RhoA/Cdc42 pathway and actin cytoskeleton remodeling in renal cell cancer. Cancer Sci. 2013;104(7):817–25.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  164. Zheng H, Bai L. Hypoxia induced microRNA-301b-3p overexpression promotes proliferation, migration and invasion of prostate cancer cells by targeting LRP1B. Exp Mol Pathol. 2019;111:104301.

    Article  PubMed  CAS  Google Scholar 

  165. Cai B, Chen W, Pan Y, Chen H, Zhang Y, Weng Z, et al. Inhibition of microRNA-500 has anti-cancer effect through its conditional downstream target of TFPI in human prostate cancer. Prostate. 2017;77(10):1057–65.

    Article  PubMed  CAS  Google Scholar 

  166. Zhang Z, Cui R, Li H, Li J. miR-500 promotes cell proliferation by directly targetting LRP1B in prostate cancer. Biosci Rep. 2019;39(4).

  167. Xing Q, Xie H, Zhu B, Sun Z, Huang Y. MiR-455-5p suppresses the progression of prostate Cancer by targeting CCR5. Biomed Res Int. 2019;2019:6394784.

    Article  PubMed  PubMed Central  Google Scholar 

  168. Arai T, Kojima S, Yamada Y, Sugawara S, Kato M, Yamazaki K, et al. Pirin: a potential novel therapeutic target for castration-resistant prostate cancer regulated by miR-455-5p. Mol Oncol. 2019;13(2):322–37.

    Article  PubMed  CAS  Google Scholar 

  169. Song SS, Huang S, Park S. Association of polygenetic risk scores related to cell differentiation and inflammation with thyroid Cancer risk and genetic interaction with dietary intake. Cancers (Basel). 2021;13(7).

  170. Bim LV, Carneiro TNR, Buzatto VC, Colozza-Gama GA, Koyama FC, Thomaz DMD et al. Molecular signature expands the landscape of driver negative thyroid cancers. Cancers (Basel). 2021;13(20).

  171. Gomez-Rueda H, Palacios-Corona R, Gutierrez-Hermosillo H, Trevino V. A robust biomarker of differential correlations improves the diagnosis of cytologically indeterminate thyroid cancers. Int J Mol Med. 2016;37(5):1355–62.

    Article  PubMed  CAS  Google Scholar 

  172. Hu Y, Zhang C, Chang Q, Du J, Lu H, Guo X, et al. MicroRNA-196a-5p targeting LRP1B modulates phenotype of thyroid carcinoma cells. Endokrynol Pol. 2023;74(2):144–52.

    Article  PubMed  CAS  Google Scholar 

  173. Prazeres H, Torres J, Rodrigues F, Pinto M, Pastoriza MC, Gomes D, et al. Chromosomal, epigenetic and microRNA-mediated inactivation of LRP1B, a modulator of the extracellular environment of thyroid cancer cells. Oncogene. 2011;30(11):1302–17.

    Article  PubMed  CAS  Google Scholar 

  174. Fei Y, Li Y, Chen F. LncRNA-IQCH-AS1 sensitizes thyroid cancer cells to doxorubicin via modulating the miR-196a-5p/PPP2R1B signalling pathway. J Chemother. 2023;35(3):250–8.

    Article  PubMed  CAS  Google Scholar 

  175. Zhang F, Fan G, Wang X. Correlation between BTG3, CASP9 and LRP4 single-nucleotide polymorphisms and susceptibility to papillary thyroid carcinoma. Biomark Med. 2022;16(7):537–47.

    Article  PubMed  CAS  Google Scholar 

  176. Ma S, Jia W, Ni S. miR-199a-5p inhibits the progression of papillary thyroid carcinoma by targeting SNAI1. Biochem Biophys Res Commun. 2018;497(1):181–6.

    Article  PubMed  CAS  Google Scholar 

  177. Lin J, Qiu Y, Zheng X, Dai Y, Xu T. The miR-199a-5p/PD-L1 axis regulates cell proliferation, migration and invasion in follicular thyroid carcinoma. BMC Cancer. 2022;22(1):756.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  178. Sun D, Han S, Liu C, Zhou R, Sun W, Zhang Z, et al. Microrna-199a-5p functions as a tumor suppressor via suppressing connective tissue growth factor (CTGF) in follicular thyroid carcinoma. Med Sci Monit. 2016;22:1210–7.

    Article  PubMed  PubMed Central  Google Scholar 

  179. Wei S, Yun X, Ruan X, Wei X, Zheng X, Gao M. Identification of potential pathogenic candidates or diagnostic biomarkers in papillary thyroid carcinoma using expression and methylation profiles. Oncol Lett. 2019;18(6):6670–8.

    PubMed  PubMed Central  CAS  Google Scholar 

  180. Wu G, Zheng H, Xu J, Guo Y, Zheng G, Ma C, et al. miR-429 suppresses cell growth and induces apoptosis of human thyroid cancer cell by targeting ZEB1. Artif Cells Nanomed Biotechnol. 2019;47(1):548–54.

    Article  PubMed  CAS  Google Scholar 

  181. Yu CS, Wang YB, Li Q, Yang EL, Dong BB. Long non-coding RNA OIP5-AS1 serves as a competing endogenous RNA to modulate X-linked inhibitor of apoptosis protein expression via adsorbing miR-429 in papillary thyroid cancer. J Biol Regul Homeost Agents. 2021;35(3):909–20.

    PubMed  CAS  Google Scholar 

  182. Liu D, Zhang M, Song Y, Yang N. RNF185 antisense RNA 1 (RNF185-AS1) promotes proliferation, migration, and invasion in papillary thyroid carcinoma. Anticancer Drugs. 2022;33(6):595–606.

    Article  PubMed  CAS  Google Scholar 

  183. Deng X, Wu B, Xiao K, Kang J, Xie J, Zhang X, et al. MiR-146b-5p promotes metastasis and induces epithelial-mesenchymal transition in thyroid cancer by targeting ZNRF3. Cell Physiol Biochem. 2015;35(1):71–82.

    Article  PubMed  CAS  Google Scholar 

  184. Jia M, Shi Y, Li Z, Lu X, Wang J. MicroRNA-146b-5p as an OncomiR promotes papillary thyroid carcinoma development by targeting CCDC6. Cancer Lett. 2019;443:145–56.

    Article  PubMed  CAS  Google Scholar 

  185. Sastre-Perona A, Santisteban P. Role of the Wnt pathway in thyroid cancer. Front Endocrinol (Lausanne). 2012;3:31.

    Article  PubMed  CAS  Google Scholar 

  186. Chen Y, Hao SA, Jiang Y, Gao B, Tian WG, Zhang S, et al. MicroRNA-1271 inhibits the progression of papillary thyroid carcinoma by targeting IRS1 and inactivating AKT pathway. Eur Rev Med Pharmacol Sci. 2019;23(18):7989–99.

    PubMed  CAS  Google Scholar 

  187. Huang G, Mao L, Hu X. Circ_0011373 promotes papillary thyroid carcinoma progression by regulating miR-1271/LRP6 axis. Horm (Athens). 2023;22(3):375–87.

    Article  Google Scholar 

  188. Zhang L, Li H, Li M, Zhang W, Yang Z, Zhang S. LRP6 is involved in the proliferation, migration and invasion of trophoblast cells via miR–346. Int J Mol Med. 2020;46(1):211–23.

    PubMed  PubMed Central  CAS  Google Scholar 

  189. Li C, Yang Y, Wang H, Song Y, Huang H. miR-362-3p suppresses ovarian cancer by inhibiting LRP8. Transl Oncol. 2022;15(1):101284.

    Article  PubMed  CAS  Google Scholar 

  190. Yuan J, Li T, Yi K, Hou M. The suppressive role of miR-362-3p in epithelial ovarian cancer. Heliyon. 2020;6(7):e04258.

    Article  PubMed  PubMed Central  Google Scholar 

  191. Cao S, Li N, Liao X. miR-362-3p acts as a tumor suppressor by targeting SERBP1 in ovarian cancer. J Ovarian Res. 2021;14(1):23.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  192. Wang Z, Chang X, Zhu G, Gao X, Chang L. Depletion of LncRNA MALAT1 inhibited Sunitinib resistance through regulating miR-362-3p-mediated G3BP1 in renal cell carcinoma. Cell Cycle. 2020;19(16):2054–62.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  193. Xie B, Wang Z, Li T, Xue J, Zhang C. LncRNA MALAT1 inhibits the proliferation and invasiveness of laryngeal squamous cell carcinoma Hep-2 cells by modulating miR-362-3p. Am J Transl Res. 2022;14(6):3729–40.

    PubMed  PubMed Central  CAS  Google Scholar 

  194. Xiong H, Yu Q, Gong Y, Chen W, Tong Y, Wang Y, et al. Yes-Associated protein (YAP) promotes tumorigenesis in melanoma cells through stimulation of Low-Density lipoprotein Receptor-Related protein 1 (LRP1). Sci Rep. 2017;7(1):15528.

    Article  PubMed  PubMed Central  Google Scholar 

  195. Thapa B, Koo BH, Kim YH, Kwon HJ, Kim DS. Plasminogen activator inhibitor-1 regulates infiltration of macrophages into melanoma via phosphorylation of FAK-Tyr(9)(2)(5). Biochem Biophys Res Commun. 2014;450(4):1696–701.

    Article  PubMed  CAS  Google Scholar 

  196. Salama Y, Lin SY, Dhahri D, Hattori K, Heissig B. The fibrinolytic factor tPA drives LRP1-mediated melanoma growth and metastasis. FASEB J. 2019;33(3):3465–80.

    Article  PubMed  CAS  Google Scholar 

  197. Adaku N, Ostendorf BN, Mei W, Tavazoie SF. Apolipoprotein E2 stimulates protein synthesis and promotes melanoma progression and metastasis. Cancer Res. 2023;83(18):3013–25.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  198. van Rooij E, Purcell AL, Levin AA. Developing MicroRNA therapeutics. Circ Res. 2012;110(3):496–507.

    Article  PubMed  Google Scholar 

  199. Yu QF, Liu P, Li ZY, Zhang CF, Chen SQ, Li ZH, et al. MiR-103/107 induces tumorigenicity in bladder cancer cell by suppressing PTEN. Eur Rev Med Pharmacol Sci. 2018;22(24):8616–23.

    PubMed  Google Scholar 

  200. Zhang Y, Qu X, Li C, Fan Y, Che X, Wang X, et al. miR-103/107 modulates multidrug resistance in human gastric carcinoma by downregulating Cav-1. Tumour Biol. 2015;36(4):2277–85.

    Article  PubMed  CAS  Google Scholar 

  201. Zhao G, Wei Z, Guo Y. MicroRNA-107 is a novel tumor suppressor targeting POU3F2 in melanoma. Biol Res. 2020;53(1):11.

    Article  PubMed  PubMed Central  Google Scholar 

  202. Luo M, Lei R, Zhao Q, Shen Y, He Z, Xu J. LINC00662 promotes melanoma progression by competitively binding miR-107 and activating the beta-catenin signaling pathway. Int J Med Sci. 2024;21(2):265–76.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  203. Salama Y, Takahashi S, Tsuda Y, Okada Y, Hattori K, Heissig B. YO2 induces melanoma cell apoptosis through p53-Mediated LRP1 downregulation. Cancers (Basel). 2022;15(1).

  204. Pencheva N, Tran H, Buss C, Huh D, Drobnjak M, Busam K, et al. Convergent multi-miRNA targeting of ApoE drives LRP1/LRP8-dependent melanoma metastasis and angiogenesis. Cell. 2012;151(5):1068–82.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  205. Gopal U, Bohonowych JE, Lema-Tome C, Liu A, Garrett-Mayer E, Wang B, et al. A novel extracellular Hsp90 mediated co-receptor function for LRP1 regulates EphA2 dependent glioblastoma cell invasion. PLoS ONE. 2011;6(3):e17649.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  206. Roy A, Coum A, Marinescu VD, Polajeva J, Smits A, Nelander S, et al. Glioma-derived plasminogen activator inhibitor-1 (PAI-1) regulates the recruitment of LRP1 positive mast cells. Oncotarget. 2015;6(27):23647–61.

    Article  PubMed  PubMed Central  Google Scholar 

  207. Boye K, Pujol N, Chen IDA, Daubon YP, Lee T. The role of CXCR3/LRP1 cross-talk in the invasion of primary brain tumors. Nat Commun. 2017;8(1):1571.

    Article  PubMed  PubMed Central  Google Scholar 

  208. He C, Zhang Z, Ding Y, Xue K, Wang X, Yang R, et al. LRP1-mediated pH-sensitive polymersomes facilitate combination therapy of glioblastoma in vitro and in vivo. J Nanobiotechnol. 2021;19(1):29.

    Article  CAS  Google Scholar 

  209. N RS, Behera MM, Naik SK, Das SK, Gopan S, Ghosh A, et al. Elevated expression of cholesterol transporter LRP-1 is crucially implicated in the pathobiology of glioblastoma. Front Neurol. 2022;13:1003730.

    Article  Google Scholar 

  210. Shayan S, Bahramali G, Arashkia A, Azadmanesh K. In Silico identification of hypoxic signature followed by reverse transcription-quantitative PCR validation in Cancer cell lines. Iran Biomed J. 2023;27(1):23–33.

    Article  PubMed  Google Scholar 

  211. Kosti A, Chiou J, Guardia GDA, Lei X, Balinda H, Landry T, et al. ELF4 is a critical component of a miRNA-transcription factor network and is a Bridge regulator of glioblastoma receptor signaling and lipid dynamics. Neuro Oncol. 2023;25(3):459–70.

    Article  PubMed  CAS  Google Scholar 

  212. Yue X, Wang P, Xu J, Zhu Y, Sun G, Pang Q, et al. MicroRNA-205 functions as a tumor suppressor in human glioblastoma cells by targeting VEGF-A. Oncol Rep. 2012;27(4):1200–6.

    Article  PubMed  CAS  Google Scholar 

  213. Ghasemi A, Fallah S. Epigenetic modification of MicroRNA-205 and its association with glioblastoma multiform. Clin Lab. 2017;63(7):1079–88.

    PubMed  CAS  Google Scholar 

  214. Yue X, Lan F, Hu M, Pan Q, Wang Q, Wang J. Downregulation of serum microRNA-205 as a potential diagnostic and prognostic biomarker for human glioma. J Neurosurg. 2016;124(1):122–8.

    Article  PubMed  CAS  Google Scholar 

  215. Chen W, Kong KK, Xu XK, Chen C, Li H, Wang FY, et al. Downregulation of miR–205 is associated with glioblastoma cell migration, invasion, and the epithelial-mesenchymal transition, by targeting ZEB1 via the Akt/mTOR signaling pathway. Int J Oncol. 2018;52(2):485–95.

    PubMed  CAS  Google Scholar 

  216. Song H, Bu G. MicroRNA-205 inhibits tumor cell migration through down-regulating the expression of the LDL receptor-related protein 1. Biochem Biophys Res Commun. 2009;388(2):400–5.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  217. Xiao D, Huang J, Pan Y, Li H, Fu C, Mao C, et al. Chromatin remodeling factor LSH is upregulated by the LRP6-GSK3beta-E2F1 Axis linking reversely with survival in gliomas. Theranostics. 2017;7(1):132–43.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  218. Li DM, Chen QD, Wei GN, Wei J, Yin JX, He JH, et al. Hypoxia-Induced miR-137 Inhibition increased glioblastoma multiforme growth and chemoresistance through LRP6. Front Oncol. 2020;10:611699.

    Article  PubMed  Google Scholar 

  219. Yang SH, Kang B, Choi Y, Rho HW, Son HY, Huh YM. Genetic changes and growth promotion of glioblastoma by magnetic nanoparticles and a magnetic field. Nanomed (Lond). 2021;16(10):787–800.

    Article  CAS  Google Scholar 

  220. Xu J, Sun T, Hu X. microRNA-513c suppresses the proliferation of human glioblastoma cells by repressing low-density lipoprotein receptor-related protein 6. Mol Med Rep. 2015;12(3):4403–9.

    Article  PubMed  CAS  Google Scholar 

  221. Jia P, Wei E, Liu H, Wu T, Wang H. Silencing of long non-coding RNA DLX6-AS1 weakens neuroblastoma progression by the miR-513c-5p/PLK4 axis. IUBMB Life. 2020;72(12):2627–36.

    Article  PubMed  CAS  Google Scholar 

  222. Wang O, Huang Y, Wu H, Zheng B, Lin J, Jin P. LncRNA LOC728196/miR-513c axis facilitates glioma carcinogenesis by targeting TCF7. Gene. 2018;679:119–25.

    Article  PubMed  CAS  Google Scholar 

  223. Li HY, Li YM, Li Y, Shi XW, Chen H. Circulating microRNA-137 is a potential biomarker for human glioblastoma. Eur Rev Med Pharmacol Sci. 2016;20(17):3599–604.

    PubMed  Google Scholar 

  224. Sun J, Zheng G, Gu Z, Guo Z. MiR-137 inhibits proliferation and angiogenesis of human glioblastoma cells by targeting EZH2. J Neurooncol. 2015;122(3):481–9.

    Article  PubMed  CAS  Google Scholar 

  225. Chen L, Wang X, Wang H, Li Y, Yan W, Han L, et al. miR-137 is frequently down-regulated in glioblastoma and is a negative regulator of Cox-2. Eur J Cancer. 2012;48(16):3104–11.

    Article  PubMed  CAS  Google Scholar 

  226. Li D, Shan W, Fang Y, Wang P, Li J. miR-137 acts as a tumor suppressor via inhibiting CXCL12 in human glioblastoma. Oncotarget. 2017;8(60):101262–70.

    Article  PubMed  PubMed Central  Google Scholar 

  227. Hao Y, Li X, Chen H, Huo H, Liu Z, Chai E. Over-expression of long noncoding RNA HOTAIRM1 promotes cell proliferation and invasion in human glioblastoma by up-regulating SP1 via sponging miR-137. NeuroReport. 2020;31(2):109–17.

    Article  PubMed  CAS  Google Scholar 

  228. Li KK, Yang L, Pang JC, Chan AK, Zhou L, Mao Y, et al. MIR-137 suppresses growth and invasion, is downregulated in oligodendroglial tumors and targets CSE1L. Brain Pathol. 2013;23(4):426–39.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  229. Silber J, Lim DA, Petritsch C, Persson AI, Maunakea AK, Yu M, et al. miR-124 and miR-137 inhibit proliferation of glioblastoma multiforme cells and induce differentiation of brain tumor stem cells. BMC Med. 2008;6:14.

    Article  PubMed  PubMed Central  Google Scholar 

  230. Wang L, Liu J, Zhong Z, Gong X, Liu W, Shi L, et al. PTP4A3 is a target for Inhibition of cell proliferatin, migration and invasion through Akt/mTOR signaling pathway in glioblastoma under the regulation of miR-137. Brain Res. 2016;1646:441–50.

    Article  PubMed  CAS  Google Scholar 

  231. Zhang Z, Song X, Tian H, Miao Y, Feng X, Li Y, et al. MicroRNA-137 inhibits growth of glioblastoma through EGFR suppression. Am J Transl Res. 2017;9(3):1492–9.

    PubMed  PubMed Central  CAS  Google Scholar 

  232. Lu Y, Guo G, Hong R, Chen X, Sun Y, Liu F, et al. LncRNA HAS2-AS1 promotes glioblastoma proliferation by sponging miR-137. Front Oncol. 2021;11:634893.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  233. Geng F, Yang F, Liu F, Zhao J, Zhang R, Hu S, et al. A miR-137-XIAP axis contributes to the sensitivity of TRAIL-induced cell death in glioblastoma. Front Oncol. 2022;12:870034.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  234. Dong X, Fu X, Yu M, Li Z. Long intergenic Non-Protein coding RNA 1094 promotes initiation and progression of glioblastoma by promoting microRNA-577-Regulated stabilization of Brain-Derived neurotrophic factor. Cancer Manag Res. 2020;12:5619–31.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  235. Li XX, Yu Q. Linc01094 accelerates the growth and Metastatic-Related traits of glioblastoma by sponging miR-126-5p. Onco Targets Ther. 2020;13:9917–28.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  236. Zhang W, Shen C, Li C, Yang G, Liu H, Chen X, et al. miR-577 inhibits glioblastoma tumor growth via the Wnt signaling pathway. Mol Carcinog. 2016;55(5):575–85.

    Article  PubMed  CAS  Google Scholar 

  237. Zeng P, Yang J, Liu L, Yang X, Yao Z, Ma C, et al. ERK1/2 Inhibition reduces vascular calcification by activating miR-126-3p-DKK1/LRP6 pathway. Theranostics. 2021;11(3):1129–46.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  238. Sheikh MSA, Almaeen A, Alduraywish A, Alomair BM, Salma U, Fei L, et al. Overexpression of miR-126 protects Hypoxic-Reoxygenation-Exposed HUVEC cellular injury through regulating LRP6 expression. Oxid Med Cell Longev. 2022;2022:3647744.

    Article  PubMed  PubMed Central  Google Scholar 

  239. Jansen F, Stumpf T, Proebsting S, Franklin BS, Wenzel D, Pfeifer P, et al. Intercellular transfer of miR-126-3p by endothelial microparticles reduces vascular smooth muscle cell proliferation and limits Neointima formation by inhibiting LRP6. J Mol Cell Cardiol. 2017;104:43–52.

    Article  PubMed  CAS  Google Scholar 

  240. Ye X, Hemida MG, Qiu Y, Hanson PJ, Zhang HM, Yang D. MiR-126 promotes coxsackievirus replication by mediating cross-talk of ERK1/2 and Wnt/beta-catenin signal pathways. Cell Mol Life Sci. 2013;70(23):4631–44.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  241. Yang QY, Yu Q, Zeng WY, Zeng M, Zhang XL, Zhang YL, et al. Killing two birds with one stone: miR-126 involvement in both cancer and atherosclerosis. Eur Rev Med Pharmacol Sci. 2022;26(17):6145–68.

    PubMed  Google Scholar 

  242. Javanmard AR, Jahanbakhshi A, Nemati H, Mowla SJ, Soltani BM. ADAMTS9-AS1 long Non–coding RNA sponges miR–128 and miR-150 to regulate Ras/MAPK signaling pathway in glioma. Cell Mol Neurobiol. 2023;43(5):2309–22.

    Article  PubMed  CAS  Google Scholar 

  243. Chen J, Cheng L, Zou W, Wang R, Wang X, Chen Z. ADAMTS9-AS1 constrains breast Cancer cell invasion and proliferation via sequestering miR-301b-3p. Front Cell Dev Biol. 2021;9:719993.

    Article  PubMed  PubMed Central  Google Scholar 

  244. Lee EH, Chari R, Lam A, Ng RT, Yee J, English J, et al. Disruption of the non-canonical WNT pathway in lung squamous cell carcinoma. Clin Med Oncol. 2008;2008(2):169–79.

    PubMed  Google Scholar 

  245. Wang Y, Zhang Y, Fang M, Bao W, Deng D. Two novel susceptibility loci for non-small cell lung cancer map to low-density lipoprotein receptor-related protein 5. Oncol Lett. 2016;12(4):2307–18.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  246. Sun T, Li CT, Xiong L, Ning Z, Leung F, Peng S, et al. miR-375-3p negatively regulates osteogenesis by targeting and decreasing the expression levels of LRP5 and beta-catenin. PLoS ONE. 2017;12(2):e0171281.

    Article  PubMed  PubMed Central  Google Scholar 

  247. Jin Y, Liu Y, Zhang J, Huang W, Jiang H, Hou Y, et al. The expression of miR-375 is associated with carcinogenesis in three subtypes of lung Cancer. PLoS ONE. 2015;10(12):e0144187.

    Article  PubMed  PubMed Central  Google Scholar 

  248. Garnis C, Campbell J, Davies JJ, Macaulay C, Lam S, Lam WL. Involvement of multiple developmental genes on chromosome 1p in lung tumorigenesis. Hum Mol Genet. 2005;14(4):475–82.

    Article  PubMed  CAS  Google Scholar 

  249. Cheng Y, Yang S, Shen B, Zhang Y, Zhang X, Liu T, et al. Molecular characterization of lung cancer: A two-miRNA prognostic signature based on cancer stem-like cells related genes. J Cell Biochem. 2020;121(4):2889–900.

    Article  PubMed  CAS  Google Scholar 

  250. Qiu H, Shen X, Chen B, Chen T, Feng G, Chen S, et al. miR-30b-5p inhibits cancer progression and enhances cisplatin sensitivity in lung cancer through targeting LRP8. Apoptosis. 2021;26(5–6):261–76.

    Article  PubMed  CAS  Google Scholar 

  251. Chauhan N, Dhasmana A, Jaggi M, Chauhan SC, Yallapu MM. miR-205: A potential biomedicine for Cancer therapy. Cells. 2020;9(9).

  252. Wang D, Zhang Z, O’Loughlin E, Wang L, Fan X, Lai EC, et al. MicroRNA-205 controls neonatal expansion of skin stem cells by modulating the PI(3)K pathway. Nat Cell Biol. 2013;15(10):1153–63.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  253. Dar AA, Majid S, de Semir D, Nosrati M, Bezrookove V, Kashani-Sabet M. miRNA-205 suppresses melanoma cell proliferation and induces senescence via regulation of E2F1 protein. J Biol Chem. 2011;286(19):16606–14.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  254. Liu S, Tetzlaff MT, Liu A, Liegl-Atzwanger B, Guo J, Xu X. Loss of microRNA-205 expression is associated with melanoma progression. Lab Invest. 2012;92(7):1084–96.

    Article  PubMed  CAS  Google Scholar 

  255. Li SC, Shi H, Khan M, Caplin M, Meyer T, Oberg K, et al. Roles of miR-196a on gene regulation of neuroendocrine tumor cells. Mol Cell Endocrinol. 2015;412:131–9.

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

No funding was received for this work.

Author information

Authors and Affiliations

Authors

Contributions

ZXL conceptualized the manuscript; LYQ, FW and ZXL drafted the manuscript; YXW and FW prepared the figures; LYQ, YXW and ZXL contributed in the discussion and edited the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Zixuan Li.

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-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, 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 you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. 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-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Qu, L., Wang, F., Wang, Y. et al. The regulation of LRPs by miRNAs in cancer: influencing cancer characteristics and responses to treatment. Cancer Cell Int 25, 182 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12935-025-03804-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12935-025-03804-z

Keywords