
Citation: | Huiyong Peng, Zhangwei Zhu, Jie Xing, Qian Xu, Changfeng Man, Shengjun Wang, Yingzhao Liu, Zhengdong Zhang. Expression profiling and bioinformatics analysis of serum exosomal circular RNAs in lymph node metastasis of papillary thyroid carcinoma[J]. The Journal of Biomedical Research, 2025, 39(2): 155-170. DOI: 10.7555/JBR.37.20230304 |
Most papillary thyroid carcinoma (PTC) patients have a good prognosis. However, lymph node metastasis (LNM), the most common manifestation of disease progression, is frequently associated with a poor prognosis. Nevertheless, few studies have focused on the underlying mechanisms of LNM. In the current study, we aimed to investigate the potential role of exosomal circRNAs that contribute to LNM in PTC. We identified
Thyroid cancer is a common primary endocrine tumor, accounting for approximately 2% of all malignancies in the body[1]. Among its subtypes, papillary thyroid carcinoma (PTC) is the most frequent subtype, characterized by goiter, nodules, a hard and fixed texture, an irregular shape, and an unclear boundary[2–3]. PTC affects millions of people worldwide, with an annual incidence of 4.6–14.4 cases per
Exosomes, as biological signal transmitters and carriers, act as messengers for intercellular communication in the extracellular microenvironment. Exosomes are natural, phospholipid-based bilayer membrane vesicles synthesized and released by cells, with a diameter ranging from 30 to 150 nm[7]. Extensive evidence has revealed that exosomes are usually present in various body fluids and play a pivotal role in tumor biology. Exosomes from secretory cells may transport biological components to recipient cells within the tumor microenvironment (TME), thereby participating in tumor development[8]. Luan et al[9] demonstrated that exosomes derived from melanoma cells could transport miR-106b-5p to melanocytes, participating in the epithelial-mesenchymal transition. Moreover, Thakur et al[10] demonstrated that exosomal DNA could be used as a potential circulating biomarker to identify tumor-associated genetic mutations. As such, exosomes may potentially serve as novel biomarkers for the diagnosis and treatment of tumors.
Circular RNAs (circRNAs), which are among the important molecules in exosomes, have attracted the interest of investigators in recent years. Accumulating evidence has indicated that circRNAs are significantly correlated with the development of multiple cancers[11–12]. Unlike other RNAs, circRNAs have a covalently closed cyclic structure, which makes them resistant to degradation by ribonuclease R within the organism[13]. Moreover, circRNAs exhibit cell-type-specific expression and perform different functions based on their intracellular localization[14]. Although investigators have focused on circRNAs and their roles in the progression of PTC[15], the majority of circRNA studies have been confined to cancer cells. The role of exosomal circRNAs in LNM among PTC patients remains unclear.
In the current study, we aimed to investigate the role of exosomal circRNAs in PTC patients with LNM. We first identified differentially expressed circRNAs derived from serum exosomes. Then, we determined the potential roles of significantly dysregulated circRNAs through bioinformatic analysis and preliminary verification.
Five PTC patients with LNM, aged 28 to 67 years, were enrolled from the Zhenjiang Clinical Medical School of Nanjing Medical University. The cases were diagnosed based on clinical manifestations and B-mode ultrasonography, and the diagnosis was then confirmed by pathologists after surgery. Five sex- and age-matched healthy adult subjects were included as healthy controls. Subjects with tumors, thyroid diseases, autoimmune diseases, or chronic infectious diseases were excluded. The peripheral blood parameters of all cases were within the normal range. An additional 14 PTC patients with LNM and 13 healthy controls were included as validation samples. There were no significant differences in age (P = 0.566) or sex (P = 0.586) between cases and controls. Peripheral blood samples (5 mL) were collected from each subject. After blood coagulation, the serum was obtained through centrifugation at 500 g for 20 min. The isolated serum samples were stored at −80 ℃.
The study was approved by the Institutional Review Board of the First People's Hospital of Zhenjiang (Approval No. K-20200012-Y), and was carried out in accordance with the Helsinki Declaration. Informed consent to access medical records and publish clinical data was obtained from each participant.
Human thyroid normal cell line (Nthy-ori 3-1) and PTC metastatic cell line (BCPAP) were cultured in RPMI-1640 medium (Gibco, Waltham, MA, USA) supplemented with 10% fetal bovine serum (Gibco) at 37 ℃ in a 5% CO2. These cell lines were used for validation of exosomal circRNAs and potential regulatory genes by quantitative reverse transcription-PCR (qRT-PCR) analysis.
The serum samples were centrifuged at
Total RNA was obtained from serum exosomes using the exoRNeasy Midi Kit (Cat. #77144, QIAGEN). A NanoDrop ND-1000 spectrophotometer was used to detect the concentration and quality of RNA (Thermo Fisher Scientific, Waltham, MA, USA). Next-generation sequencing (NGS) was performed by Cloud-Seq Biotech, Inc. (Shanghai, China). After the removal of ribosomal RNA, the sequencing libraries were constructed using the GenSeq® Low Input RNA Library Prep Kit (Cloud-Seq Biotech). The constructed sequencing libraries were quality controlled and quantified by Q30 with the BioAnalyzer 2100 system (Agilent, Santa Clara, CA, USA), followed by 150 bp double-ended sequencing using the Illumina NovaSeq 6000 (Illumina, Inc., San Diego, CA, USA).
Cutadapt software (v1.9.3) was used to obtain high-quality reads. These reads were then compared to the reference genome/transcriptome using STAR software (v2.5.1b), and circRNAs were identified using DCC software (v0.4.4). Subsequently, the identified circRNAs were annotated using the circBase database (http://www.circbase.org/). The data were standardized, and the differentially expressed circRNAs were screened using edgeR software (v3.16.5) and were presented as logarithmic counts per million. CircRNAs with a fold change ≥ 2.0 and a P-value < 0.05 were determined to be significantly dysregulated.
The Gene Ontology (GO; http://www.geneontology.org) enrichment analysis of host genes was performed to annotate and speculate on the functions of their corresponding circRNAs, including biological process (BP), cellular component (CC), and molecular function (MF). Additionally, the potential enrichment signaling pathways of circRNAs were predicted by the Kyoto Encyclopedia of Genes and Genomes (KEGG) (https://www.genome.jp/kegg/) analysis of circRNA-derived host genes.
The ceRNA regulatory network is an important regulatory mode among circRNA, miRNA, and mRNA. The potential sponging relationships between circRNAs and miRNAs were predicted using miRanda (v3.3) and TargetScan software (v8.0). The intersection of the results from four prediction programs, including miRWalk, miRDB, miRTarBase, and TargetScan, was obtained to predict the target genes of miRNAs. Cytoscape software (v3.8.2) was used to construct the circRNA-miRNA-mRNA regulatory networks. The functions of mRNAs in regulatory networks were further determined through the GO and KEGG pathway enrichment analyses. Furthermore, the predicted miRNAs and genes were screened through the existing studies to identify which miRNAs had been reported to show an inverse expression trend to their corresponding circRNAs in PTC and which genes had been documented to be correlated with PTC patients with LNM.
Total RNA was extracted from cell lines using an RNA rapid extraction kit (Yishan, Shanghai, China) according to the manufacturer's instructions. cDNA reverse transcription was carried out using the ReverTraAca® RT-qPCR kit (Toyobo, Osaka, Japan) according to the manufacturer's instructions. TB Green® Premix Ex Taq Ⅱ (Takara, Osaka, Japan) was used to amplify cDNA in the ABI7500 instrument (Applied Biosystems, Foster City, CA, USA). The primers are summarized in Supplementary Table 1 (available online). The transcript levels of circRNAs and mRNAs were normalized to actin beta (ACTB).
The STRING database (https://string-db.org/) was used to predict the potential relationships between circRNAs and protein-coding genes, and PPI networks were generated to describe the interactions among these protein-coding genes using Cytoscape software (PPI score > 0.9). CentiScaPe, a Cytoscape plugin, was used to calculate the degree centrality. The Molecular Complex Detection algorithm (MCODE) was used to determine the hub modules in the constructed PPI networks. Subsequently, the hub genes were identified based on degree centrality and module clustering[16].
The transcript levels of potential regulatory genes were analyzed in the TCGA-THCA database (https://portal.gdc.cancer.gov/projects/TCGA-THCA). The database contained the sequencing data of 24 PTC patients with LNM and paired normal thyroid tissues. Additionally, immunohistochemical data for the potential regulatory genes in PTC and normal thyroid tissues were downloaded from the Human Protein Atlas (HPA) database (https://www.proteinatlas.org/). The staining intensity was classified into four categories: not detected, low, medium, and high.
The sequencing data were analyzed using R software. Quantitative data were presented as mean ± standard deviation, and significant differences between two groups were assessed using Student's t-test or χ2 test (SPSS 16.0.0.247 software). A P-value < 0.05 was considered statistically significant.
We collected peripheral blood from PTC patients with LNM and healthy controls to extract exosomes from human serum, and designated the exosomes as PTC (LNM)-Exos and HC-Exos, respectively. Characterization of the exosomes showed that PTC (LNM)-Exos and HC-Exos exhibited a typical cup-shaped morphology (Fig. 1A). Additionally, serum exosomes from both groups expressed the exosomal markers (CD63 and CD81) but were negative for calnexin, a cellular constituent used as a negative protein marker of exosomes (Fig. 1B). Subsequently, we analyzed the size of exosomes using the NanoFCM analysis and found that the size of exosomes was approximately 80–100 nm in diameter (Fig. 1C). These results indicated that the extracted serum components were exosomes, providing a basis for further experiments.
The expression profile of exosome circRNAs was examined using NGS (GEO ID: GSE247120). Hierarchical cluster analysis revealed distinct patterns of circRNAs between the PTC (LNM)-Exos and HC-Exos groups (Fig. 2A). There were
Next, we analyzed the human chromosomal distribution of the
The potential biological functions of differentially expressed circRNAs were determined by GO enrichment analysis. A total of 679 GO terms associated with upregulated circRNAs were identified, and the top 10 GO terms are presented in Fig. 4A. For downregulated circRNAs,
The potential signaling pathways correlated with dysregulated circRNAs were determined by the KEGG pathway analysis of their host genes. According to the KEGG classification, a total of 91 significantly enriched signaling pathways were identified, including 34 pathways correlated with upregulated circRNAs and 57 pathways involved in downregulated circRNAs. Among them, 15 signaling pathways were closely related to both upregulated and downregulated circRNAs. The top 10 enriched signaling pathways for upregulated and downregulated circRNAs are shown in Fig. 5A and 5B, respectively. The WNT, ERBB, and calcium signaling pathways have been reported as key factors in the progression of PTC[17–19]. These findings suggest that dysregulated exosomal circRNAs may play a significant role in PTC with LNM.
Currently, research on the mechanisms of disease-related mRNAs has shifted to circRNAs, particularly their roles in the crosstalk among circRNAs, miRNAs, and mRNAs. As expected, a total of
Next, we examined the transcript levels of these intersecting top five miRNAs in PTC by reported studies and identified seven miRNAs that exhibited an inverse expression pattern, compared with their corresponding circRNAs. We further investigated the role of the 127 predicted target genes in PTC by reported studies and identified 14 candidate genes linked with LNM in PTC patients (Supplementary Table 3, available online). Together, these circRNA/miRNA/mRNA axes, including six circRNAs, seven miRNAs, and 14 target genes, provided important clues for the mechanisms of LNM in PTC.
To further investigate the specific role of circRNAs, we selected the six screened circRNAs for preliminary validation by expanding the sample size and cell lines. The BCPAP cell line was selected for validation because it was established from the tumor tissues of a female patient with metastatic PTC[20]. Firstly, we renamed the selected circRNAs based on their source genes. Our data showed that the expression levels of three out of four circRNAs in serum exosomes and four out of five circRNAs in cell-derived exosomes were consistent with the sequencing data in serum exosomes (Fig. 7A) and in cell line-derived exosomes (Fig. 7B), respectively. However, circLRRC47 was not amplified by appropriate primers, and circZDHHC17-NAV3 was not detected in serum exosomes because of its low expression. Intriguingly, compared with that of the controls, circFCHO2 expression was attenuated in serum exosomes from PTC patients with LNM and in BCPAP cell-derived exosomes, which was contrary to the sequencing results.
Then, a total of 24 paired tissue samples from PTC patients with LNM were screened in the TCGA database. The expression levels of 11 potential regulatory protein-coding genes of circTACC2, circFCHO2, circBIRC6, and circZDHHC17-NAV3 were analyzed in the TCGA database and verified in Nthy-ori 3-1 and BCPAP cell lines. The significant expression levels of SIRT1, PRRX1, and BCL2L11 showed the same trend in both the TCGA database and cell lines (Fig. 7C and 7D). In contrast, the expression trend of the other eight potentially regulated genes was inconsistent between the TCGA database and cell lines.
Furthermore, immunohistochemical staining data derived from the HPA database were analyzed to determine the protein levels of the potentially regulated genes. As shown in Fig. 8, heightened staining intensity of EGFR and SIRT1, as well as attenuated staining intensity of MDM4, SMAD4, and BCL2L11, were observed in PTC tissues, compared with normal thyroid tissues. Notably, strong staining for EGFR was also observed in other PTC samples (data not shown). Based on the ceRNA network and the expression trends of miRNAs in PTC, the circTACC2-miR-7-EGFR and circBIRC6-miR-24-3p-BCL2L11 axes were identified as potentially involved in PTC with LNM through preliminary verification.
A total of 600 upregulated circRNA-derived host genes and
PTC may exhibit aggressive behavior, usually preceded by LNM[21]. According to previous studies, the incidence of LNM in PTC ranged from 30% to 80%[22–23]. LNM is a significant risk factor for poor prognosis in PTC patients[6]. However, the mechanisms underlying LNM in PTC remain unclear. Recently, exosomal circRNAs have attracted attention because of their unique function in material communication of TME. Dai et al[24] found that hsa_circ_0082002 and hsa_circ_0003863 in exosomes may contribute to the diagnosis of PTC. Lin et al[25] demonstrated that exosomal hsa_circ_007293 shared functional properties with the miR-653/paired box 6 axis and promoted the progression of PTC. Despite these findings, the molecular mechanisms regulating PTC with LNM by exosomal circRNAs still require further clarification. In the current study, we characterized the expression profiles of circRNAs in serum exosomes and identified
We subsequently investigated the potential functions of the dysregulated circRNAs through bioinformatics analyses of their host genes. In the GO terms, cellular components are fundamental elements of tumor biological behavior, and the enriched biological molecules, such as p53 and GTPase, have been reported to play critical roles in the progression of PTC[27–28]. In addition, 91 signaling pathways correlated with the host genes of dysregulated circRNAs were identified. Among these pathways, the glucagon signaling pathway, the most relevant pathway for upregulated circRNAs, promotes PTC invasion and migration by mediating the epithelial-mesenchymal transition and p38/extracellular-regulated kinase (ERK) pathways[29]. Other relevant pathways correlated with upregulated circRNAs, such as the WNT and ERBB signaling pathways, are known to be involved in LNM in PTC patients[17–18]. The phospholipase D signaling pathway was the most relevant pathway for downregulated circRNAs, which synergistically activates signal transducer and activator of transcription 3 (STAT3) by directly interacting with the RET/PTC gene[30]. Additionally, calmodulin-dependent kinaseⅡ (CaMKⅡ) contributes to the activation of ERK cascade, thereby promoting the progression of PTC[19]. The HIF-1 signaling pathway facilitates the aggressive and progressive metastatic form of PTC by reprogramming the glucose/iodine metabolic program in hypoxic conditions[31]. Furthermore, MAPK is present in various signaling pathways and is broadly known to be involved in LNM of PTC[32]. The PI3K/AKT, another important signaling pathway closely related to multiple KEGG pathways, functions in thyroid tumorigenesis and progression by enhancing protein synthesis and overall reprogramming of cancer cell metabolism[33–34]. Taken together, these enriched signaling pathways suggest that dysregulated exosomal circRNAs may be involved in the LNM of PTC and serve as novel research targets for understanding the mechanisms of LNM in PTC.
As an important regulatory mechanism for circRNAs, the crosstalk among circRNA, miRNA, and mRNA has been widely investigated. There are miRNA binding sites in the circRNA sequence, and miRNA may target the 3′ UTR of mRNA. The exact mechanism is that circRNA acts as a sponge for miRNA, resulting in decreased miRNA expression, thereby releasing the suppression of target genes by miRNAs[35]. CiRS-7 was the first identified circRNA in humans and mice, which promoted the expression levels of miR-7 target genes by sponging miR-7[36]. In PTC, the functional mechanism of circRNA primarily involves miRNA sponging[15]. For example, the upregulated hsa_circ_0058124 identified in PTC patients promoted the invasion and metastasis of tumor cells by targeting miR-218-5p, thereby terminating the inhibitory effect of NUMB[37]. Similarly, circTIAM1 was demonstrated to activate the HNRNPA1 signaling pathway and promote the progression of PTC by sponging miR-646[38]. Based on the ceRNA network and the key role of exosomes as a "bridge" in TME communication, we speculated that these dysregulated exosomal circRNAs might influence LNM in PTC by sponging miRNAs. Hence, we selected the top 10 dysregulated circRNAs for their miRNA and mRNA target prediction. Subsequent GO and KEGG enrichment analyses of 127 predicted target genes revealed a significant enrichment in multiple PTC-related signaling pathways, including p53, MAPK, and PI3K/AKT pathways. The deletion of P53, a classic tumor suppressor gene, is one of the most characteristic genetic changes contributing to poorly differentiated thyroid malignancies[39]. In PTC, the activation of MAPK and PI3K/AKT signaling pathways leads to abnormal proliferation and metastasis of PTC cells[40–41].
Through comprehensive analysis, we screened 14 predicted circRNA/miRNA/mRNA axes potentially correlated with LNM in PTC, including four upregulated and two downregulated circRNAs. Validation in expanded samples and cell lines as well as database analysis revealed that the circTACC2-miR-7-EGFR and circBIRC6-miR-24-3p-BCL2L11 axes might be involved in LNM of PTC. EGFR is associated with the progression of PTC, triggering a PI3K-AKT signal cascade to promote the invasion and metastasis of tumor cells[42]. However, the mechanisms underlying elevated EGFR levels in PTC remain poorly understood. In the current study, we screened a novel upregulated circRNA, circTACC2, which might promote EGFR expression by interacting with miR-7 in PTC patients with LNM. Notably, miR-7 expression has been reported to be decreased in PTC specimens, where it inhibits the progression of PTC[43]. Another downregulated ecircRNA, circBIRC6, may facilitate the progression of PTC by regulating the miR-24-3p/BCL2L11 axis. BCL2L11, a member of the BCL2 family, functions as a tumor suppressor by regulating cell apoptosis[44]. Moreover, as commonly observed to be downregulated in PTC patients, BCL2L11 has been reported to be induced after treatment with Src and MEK1/2 inhibitors, thereby enhancing apoptosis[45]. Together, these ceRNA networks underscore the significant role of exosomal circRNAs in the progression of PTC.
Moreover, we screened 12 hub genes (i.e., PLCD3, PIP5K1A, PIP4K2A, PIP5K1C, HNRNPC, NUP54, NUP93, GEMIN5, FIP1L1, SNRPD3, NUP98, and NUPL1) with high connectivity from PPI networks by bioinformatics analysis. Among them, PLCD3 has been identified as an oncogenic gene in PTC, with its regulatory mechanism mediated through the has_circ_0003747/miR-338-3p/PLCD3 axis[46]. Lin et al[47] also found that PLCD3 expression was correlated with LNM and disease stage in thyroid cancer via the Hippo signaling pathway. Thus, the elevated levels of PLCD3 may play a critical role in identifying LNM of PTC. Additionally, PLCD3 is a BRAFV600E-associated biomarker with both prognostic and diagnostic significance in PTC patients[48]. Our data showed that PLCD3 mRNA expression levels were increased in PTC patients with LNM. However, immunohistochemical staining intensity was moderate in both PTC and normal thyroid tissues from the HPA database. One possible explanation for this discrepancy is that the immunohistochemical staining may be too strong, which requires further verification. Regarding other hub genes, GEMIN5 expression was upregulated in low-metastatic breast cancer cells, and the siRNA-mediated reduction of GEMIN5 expression increased the motility of tumor cells by affecting the expression of spliceosomal proteins[49], suggesting that the depressed GEMIN5 expression may be associated with LNM in PTC. In addition, SNRPD3, as a co-factor for MYCN oncogenesis, participates in maintaining the fidelity and balance of alternative splicing events in neuroblastoma cells[50]. However, the role of attenuated SNRPD3 expression in PTC patients with LNM remains poorly understood.
The findings of the current study provide novel insights into potential candidate genes for future research. However, some limitations should be acknowledged. First, the sample size for verification was small, which requires future verification by expanding the sample size. Second, the detailed mechanisms of certain circRNAs in PTC patients with LNM remain unexplored.
In summary, the current study provides the first expression profile of exosomal circRNAs and elucidates the connection between circRNAs, miRNAs, and mRNAs in PTC patients with LNM. The identified exosomal circRNAs and their potential functions may shed light on new molecular mechanisms underlying the LNM in PTC and serve as potential biomarkers in PTC patients with LNM.
The authors acknowledge and appreciate all the volunteers who were involved in the study and our colleagues for their valuable efforts and comments on this paper.
This work was supported by the National Natural Science Foundation of China (Grant No. 81800698), the Jiangsu Provincial Medical Key Discipline Cultivation Unit (Grant No. JSDW202241), the Research Project of Jiangsu Commission of Health (Grant No. H2023053), and Zhenjiang Science and the Technology Planning Project (Grant Nos. SH2023006 and SH2023008).
CLC number: R736.1, Document code: A
The authors reported no conflict of interests.
[1] |
Siegel RL, Miller KD, Fuchs HE, et al. Cancer statistics, 2022[J]. CA Cancer J Clin, 2022, 72(1): 7–33. doi: 10.3322/caac.21708
|
[2] |
Lin R, Yang S, Jia Y, et al. Epigenetic regulation of papillary thyroid carcinoma by long non-coding RNAs[J]. Semin Cancer Biol, 2022, 83: 253–260. doi: 10.1016/j.semcancer.2021.03.027
|
[3] |
Lee TY, Lee S, Bae JS, et al. Distinct clinical manifestations of thyroid cancer after hematopoietic stem cell transplantation[J]. Ann Surg Oncol, 2019, 26(11): 3586–3592. doi: 10.1245/s10434-019-07442-4
|
[4] |
La Vecchia C, Malvezzi M, Bosetti C, et al. Thyroid cancer mortality and incidence: a global overview[J]. Int J Cancer, 2015, 136(9): 2187–2195. doi: 10.1002/ijc.29251
|
[5] |
Albero A, Lopéz JE, Torres A, et al. Effectiveness of chemotherapy in advanced differentiated thyroid cancer: a systematic review[J]. Endocr Relat Cancer, 2016, 23(2): R71–R84. doi: 10.1530/ERC-15-0194
|
[6] |
Yu J, Deng Y, Liu T, et al. Lymph node metastasis prediction of papillary thyroid carcinoma based on transfer learning radiomics[J]. Nat Commun, 2020, 11(1): 4807. doi: 10.1038/s41467-020-18497-3
|
[7] |
Jeppesen DK, Fenix AM, Franklin JL, et al. Reassessment of exosome composition[J]. Cell, 2019, 177(2): 428–445. e18.
|
[8] |
Marar C, Starich B, Wirtz D. Extracellular vesicles in immunomodulation and tumor progression[J]. Nat Immunol, 2021, 22(5): 560–570. doi: 10.1038/s41590-021-00899-0
|
[9] |
Luan W, Ding Y, Xi H, et al. Exosomal miR-106b-5p derived from melanoma cell promotes primary melanocytes epithelial-mesenchymal transition through targeting EphA4[J]. J Exp Clin Cancer Res, 2021, 40(1): 107. doi: 10.1186/s13046-021-01906-w
|
[10] |
Thakur BK, Zhang H, Becker A, et al. Double-stranded DNA in exosomes: a novel biomarker in cancer detection[J]. Cell Res, 2014, 24(6): 766–769. doi: 10.1038/cr.2014.44
|
[11] |
Wang Y, Liu J, Ma J, et al. Exosomal circRNAs: biogenesis, effect and application in human diseases[J]. Mol Cancer, 2019, 18(1): 116. doi: 10.1186/s12943-019-1041-z
|
[12] |
Dai J, Su Y, Zhong S, et al. Exosomes: key players in cancer and potential therapeutic strategy[J]. Signal Transduct Target Ther, 2020, 5(1): 145. doi: 10.1038/s41392-020-00261-0
|
[13] |
Li X, Yang L, Chen L. The biogenesis, functions, and challenges of circular RNAs[J]. Mol Cell, 2018, 71(3): 428–442. doi: 10.1016/j.molcel.2018.06.034
|
[14] |
Liu C, Chen L. Circular RNAs: characterization, cellular roles, and applications[J]. Cell, 2022, 185(12): 2016–2034. doi: 10.1016/j.cell.2022.04.021
|
[15] |
Xu X, Jing J. Advances on circRNAs contribute to carcinogenesis and progression in papillary thyroid carcinoma[J]. Front Endocrinol (Lausanne), 2021, 11: 555243. doi: 10.3389/fendo.2020.555243
|
[16] |
Xu B, Huang X, Yan Y, et al. Analysis of expression profiles and bioinformatics suggests that plasma exosomal circular RNAs may be involved in ischemic stroke in the Chinese Han population[J]. Metab Brain Dis, 2022, 37(3): 665–676. doi: 10.1007/s11011-021-00894-2
|
[17] |
Xu M, Lin B, Zheng D, et al. LEM domain containing 1 promotes thyroid cancer cell proliferation and migration by activating the Wnt/β-catenin signaling pathway and epithelial-mesenchymal transition[J]. Oncol Lett, 2021, 21(6): 442. doi: 10.3892/ol.2021.12703
|
[18] |
Sa R, Liang R, Qiu X, et al. IGF2BP2-dependent activation of ERBB2 signaling contributes to acquired resistance to tyrosine kinase inhibitor in differentiation therapy of radioiodine-refractory papillary thyroid cancer[J]. Cancer Lett, 2022, 527: 10–23. doi: 10.1016/j.canlet.2021.12.005
|
[19] |
Rusciano MR, Salzano M, Monaco S, et al. The Ca2+-calmodulin-dependent kinase II is activated in papillary thyroid carcinoma (PTC) and mediates cell proliferation stimulated by RET/PTC[J]. Endocr Relat Cancer, 2010, 17(1): 113–123. doi: 10.1677/ERC-09-0214
|
[20] |
Fabien N, Fusco A, Santoro M, et al. Description of a human papillary thyroid carcinoma cell line. Morphologic study and expression of tumoral markers[J]. Cancer, 1994, 73(8): 2206–2212. doi: 10.1002/1097-0142(19940415)73:8<2206::AID-CNCR2820730828>3.0.CO;2-M
|
[21] |
Zhao Q, Ming J, Liu C, et al. Multifocality and total tumor diameter predict central neck lymph node metastases in papillary thyroid microcarcinoma[J]. Ann Surg Oncol, 2013, 20(3): 746–752. doi: 10.1245/s10434-012-2654-2
|
[22] |
Caliskan O, Unlu MT, Yanar C, et al. Predictive factors affecting the development of lateral lymph node metastasis in papillary thyroid cancer[J]. Sisli Etfal Hastan Tip Bul, 2023, 57(3): 312–319. https://pubmed.ncbi.nlm.nih.gov/37900340/
|
[23] |
Haugen BR, Alexander EK, Bible KC, et al. 2015 American thyroid association management guidelines for adult patients with thyroid nodules and differentiated thyroid cancer: the American thyroid association guidelines task force on thyroid nodules and differentiated thyroid cancer[J]. Thyroid, 2016, 26(1): 1–133. doi: 10.1089/thy.2015.0020
|
[24] |
Dai L, Hu W, Jiang H, et al. The diagnostic potential of two exosome-derived circRNAs for papillary thyroid cancer[J]. Int J Clin Oncol, 2023, 28(11): 1461–1474. doi: 10.1007/s10147-023-02400-3
|
[25] |
Lin Q, Qi Q, Hou S, et al. Exosomal circular RNA hsa_circ_007293 promotes proliferation, migration, invasion, and epithelial-mesenchymal transition of papillary thyroid carcinoma cells through regulation of the microRNA-653-5p/paired box 6 axis[J]. Bioengineered, 2021, 12(2): 10136–10149. doi: 10.1080/21655979.2021.2000745
|
[26] |
Paskeh MDA, Entezari M, Mirzaei S, et al. Emerging role of exosomes in cancer progression and tumor microenvironment remodeling[J]. J Hematol Oncol, 2022, 15(1): 83. doi: 10.1186/s13045-022-01305-4
|
[27] |
De Falco V, Castellone MD, De Vita G, et al. RET/papillary thyroid carcinoma oncogenic signaling through the Rap1 small GTPase[J]. Cancer Res, 2007, 67(1): 381–390. doi: 10.1158/0008-5472.CAN-06-0981
|
[28] |
Lu Z, Wen D, Wei W, et al. Silencing of PPM1D inhibits cell proliferation and invasion through the p38 MAPK and p53 signaling pathway in papillary thyroid carcinoma[J]. Oncol Rep, 2020, 43(3): 783–794. doi: 10.3892/or.2020.7458
|
[29] |
Jiang H, Chen X, Sun H, et al. Tumor promoting effects of glucagon receptor: a promising biomarker of papillary thyroid carcinoma via regulating EMT and P38/ERK pathways[J]. Hum Cell, 2020, 33(1): 175–184. doi: 10.1007/s13577-019-00284-y
|
[30] |
Kim YR, Byun HS, Won M, et al. Modulatory role of phospholipase D in the activation of signal transducer and activator of transcription (STAT)-3 by thyroid oncogenic kinase RET/PTC[J]. BMC Cancer, 2008, 8: 144. doi: 10.1186/1471-2407-8-144
|
[31] |
Song H, Qiu Z, Wang Y, et al. HIF-1α/YAP signaling rewrites glucose/iodine metabolism program to promote papillary thyroid cancer progression[J]. Int J Biol Sci, 2023, 19(1): 225–241. doi: 10.7150/ijbs.75459
|
[32] |
Zhan S, Wang T, Li J, et al. Asporin interacts with HER2 to promote thyroid cancer metastasis via the MAPK/EMT signaling pathway[J]. Front Oncol, 2022, 12: 762180. doi: 10.3389/fonc.2022.762180
|
[33] |
Cao Z, Zhang Z, Tang X, et al. Comprehensive analysis of tissue proteomics in patients with papillary thyroid microcarcinoma uncovers the underlying mechanism of lymph node metastasis and its significant sex disparities[J]. Front Oncol, 2022, 12: 887977. doi: 10.3389/fonc.2022.887977
|
[34] |
Mirshahidi S, Yuan IJ, Simental A, et al. Targeting tumor microenvironment akt signaling represents a potential therapeutic strategy for aggressive thyroid cancer[J]. Int J Mol Sci, 2023, 24(6): 5471. doi: 10.3390/ijms24065471
|
[35] |
Chen L. The expanding regulatory mechanisms and cellular functions of circular RNAs[J]. Nat Rev Mol Cell Biol, 2020, 21(8): 475–490. doi: 10.1038/s41580-020-0243-y
|
[36] |
Hansen TB, Jensen TI, Clausen BH, et al. Natural RNA circles function as efficient microRNA sponges[J]. Nature, 2013, 495(7441): 384–388. doi: 10.1038/nature11993
|
[37] |
Yao Y, Chen X, Yang H, et al. Hsa_circ_0058124 promotes papillary thyroid cancer tumorigenesis and invasiveness through the NOTCH3/GATAD2A axis[J]. J Exp Clin Cancer Res, 2019, 38(1): 318. doi: 10.1186/s13046-019-1321-x
|
[38] |
Zhang D, Tao L, Xu N, et al. CircRNA circTIAM1 promotes papillary thyroid cancer progression through the miR-646/HNRNPA1 signaling pathway[J]. Cell Death Discov, 2022, 8(1): 21. doi: 10.1038/s41420-021-00798-1
|
[39] |
Manzella L, Stella S, Pennisi MS, et al. New insights in thyroid cancer and p53 family proteins[J]. Int J Mol Sci, 2017, 18(6): 1325. doi: 10.3390/ijms18061325
|
[40] |
Scheffel RS, Dora JM, Maia AL. BRAF mutations in thyroid cancer[J]. Curr Opin Oncol, 2022, 34(1): 9–18. doi: 10.1097/CCO.0000000000000797
|
[41] |
Leandro-García LJ, Landa I. Mechanistic insights of thyroid cancer progression[J]. Endocrinology, 2023, 164(9): bqad118. doi: 10.1210/endocr/bqad118
|
[42] |
Zhang Z, Liu Z, Ren W, et al. The miR-200 family regulates the epithelial-mesenchymal transition induced by EGF/EGFR in anaplastic thyroid cancer cells[J]. Int J Mol Med, 2012, 30(4): 856–862. doi: 10.3892/ijmm.2012.1059
|
[43] |
Hua K, Jin J, Zhang H, et al. MicroRNA-7 inhibits proliferation, migration and invasion of thyroid papillary cancer cells via targeting CKS2[J]. Int J Oncol, 2016, 49(4): 1531–1540. doi: 10.3892/ijo.2016.3660
|
[44] |
Zhang H, Duan J, Qu Y, et al. Onco-miR-24 regulates cell growth and apoptosis by targeting BCL2L11 in gastric cancer[J]. Protein Cell, 2016, 7(2): 141–151. doi: 10.1007/s13238-015-0234-5
|
[45] |
Rose MM, Espinoza VL, Hoff KJ, et al. BCL2L11 induction mediates sensitivity to src and MEK1/2 inhibition in thyroid cancer[J]. Cancers (Basel), 2023, 15(2): 378. doi: 10.3390/cancers15020378
|
[46] |
Dou X, Xia F, Li X. Circ_0003747 promotes thyroid cancer progression by sponging miR-338-3p to upregulate PLCD3 expression[J]. Epigenetics, 2023, 18(1): 2210339. doi: 10.1080/15592294.2023.2210339
|
[47] |
Lin L, Wen J, Lin B, et al. Phospholipase C Delta 3 inhibits apoptosis and promotes proliferation, migration, and invasion of thyroid cancer cells via Hippo pathway[J]. Acta Biochim Biophys Sin (Shanghai), 2021, 53(4): 481–491. doi: 10.1093/abbs/gmab016
|
[48] |
Han Y, Yu X, Yin Y, et al. Identification of potential BRAF inhibitor joint therapy targets in PTC based on WGCAN and DCGA[J]. J Cancer, 2021, 12(6): 1779–1791. doi: 10.7150/jca.51551
|
[49] |
Lee JH, Horak CE, Khanna C, et al. Alterations in Gemin5 expression contribute to alternative mRNA splicing patterns and tumor cell motility[J]. Cancer Res, 2008, 68(3): 639–644. doi: 10.1158/0008-5472.CAN-07-2632
|
[50] |
Salib A, Jayatilleke N, Seneviratne JA, et al. MYCN and SNRPD3 cooperate to maintain a balance of alternative splicing events that drives neuroblastoma progression[J]. Oncogene, 2024, 43(5): 363–377. doi: 10.1038/s41388-023-02897-y
|