Open Access

Interplay between Wnt signaling molecules and exosomal miRNAs in breast cancer (Review)

  • Authors:
    • Hailong Li
    • Xia Li
    • Wei Du
  • View Affiliations

  • Published online on: June 28, 2024     https://doi.org/10.3892/or.2024.8766
  • Article Number: 107
  • Copyright: © Li et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Breast cancer (BC) is the most common malignancy in women worldwide. Wnt signaling is involved in tumorigenesis and cancer progression, and is closely associated with the characteristics of BC. Variation in the expression of exosomal microRNAs (miRNAs) modulates key cancer phenotypes, such as cellular proliferation, epithelial‑mesenchymal transition, metastatic potential, immune evasion and treatment resistance. The present review aimed to discuss the importance of Wnt signaling and exosomal miRNAs in regulating the occurrence and development of BC. In addition, the present review determined the crosstalk between Wnt signaling and exosomal miRNAs, and highlighted potential diagnostic biomarkers and therapeutic targets.

Introduction

Breast cancer (BC) is the most common malignancy among women worldwide, with an heterogeneous nature resulting from various risk factors, such as endogenous and exogenous estrogen exposure, lifestyle choices, dietary habits and exposure to toxic environmental elements, such as heavy metals and chemicals (1,2). In total, ~15% of BC cases can be attributed to genetic susceptibility and genetic factors (3). Notably, research has revealed a substantial discrepancy in the 5-year survival rates between patients with advanced invasive BC (representing 24% of cases with distant metastasis) and those with early-stage BC, with a 99% survival rate (4). In addition, some patients with cancer may benefit from monotherapies, such as hormone therapy, immunotherapy or chemotherapy; however, the effectiveness of these therapies may diminish over time, and some patients may become resistant (5). Thus, the development of novel therapeutic targets is crucial for the treatment of BC.

In addition to regulating cell proliferation and cell fate during embryonic development and tissue homeostasis, Wnt signaling determines cell polarity (6,7). Notably, Wnt is a secreted glycoprotein within this pathway (8). Diverse intracellular signaling pathways may be activated by these interactions, which may intersect or function independently (9). Collectively, an integrin gene, Int-1, and a segmental polarity gene, Wingless, form the term ‘Wnt’ (10,11). Wnt-1, formerly known as the Int-1 oncogene, has been recognized as the integration site for the mouse mammary tumor virus (MMTV) (10). In addition, BC was the first tumor to be associated with Wnt signaling. Results of previous studies demonstrated that Wnt signaling is involved in multiple aspects of cancer, including proliferation (12,13), metastasis (14,15), immune regulation (16,17), therapeutic resistance (18) and phenotype shaping (19). Moreover, a number of inhibitors targeting components of the Wnt signaling pathway exhibit potential in the treatment of cancer (20). Thus, the present review demonstrated the interaction between exosomal microRNAs (miRNAs) and proteins in signaling cascades, demonstrating their role in BC and in mechanisms of the Wnt signaling pathway.

Exosomes are nano-sized vesicle structures ranging from 50–150 nm. These are derived from endosomes and are present in diverse tumor cells (21,22). Numerous proteins and lipids are present in these vesicles, in addition to miRNA (23). A variety of model systems have been used to demonstrate that cancer cells secrete extracellular vesicles, and these subsequently metastasize from primary tumors to the circulatory system (24). Research focused on exosomal miRNAs have demonstrated their role in cellular and molecular biology (25). Notably, miRNAs are endogenous small RNAs that are 20–24 nucleotides in length. In tumor cells, miRNAs play a crucial regulatory role in various signaling pathways within exosomes (26). Cancer-specific exosomal miRNAs exhibit distinct expression patterns and play a significant role in the progression of the disease, highlighting their potential as biomarkers of cancer (27). Notably, BC tissue expresses altered exosomal miRNAs both before and after invasion (28,29). Thus, through studying the functional role of exosomal miRNA in BC, a novel theoretical basis may be developed to understand its etiology.

Overview of Wnt signalling

In 1973, genetic research using Drosophila melanogaster revealed the components of the Wnt signaling pathway (30). In 1982, Nusse et al (10) demonstrated that the mouse Wnt1 gene promotes the occurrence and development of mammary carcinomas in MMTV (10). Subsequently, numerous studies have demonstrated that the Wnt signaling pathway exerts a significant impact on human tumors. For example, the deletion of genes associated with rectal cancer leads to excessive activation of Wnt signaling, thereby promoting tumor development (30). Results of a previous study also revealed that Wnt-dependent systemic inflammation drives BC metastasis when p53 is lost (31). Notably, 19 Wnt genes have been identified in the human genome, and these encode secreted lipoglycoproteins (32). These lipoglycoproteins play a fundamental role in controlling cell size, intercellular communication, embryonic development and stem cell self-renewal (33). In addition, Wnt signaling pathways are divided into three types; namely, i) canonical Wnt signaling pathways, ii) Wnt-planar cell polarity (PCP) signaling pathways, and iii) Wnt-Ca2+ signaling pathways (34). Canonical Wnt signaling transduction, also known as Wnt/β-catenin protein signaling, is the most well-established pathway. This pathway is induced by Wnt1, Wnt2, Wnt3a, Wnt8b, Wnt10a and Wnt10b (35). β-catenin proteins are the core components of canonical Wnt signaling, and these bind to the cytoplasmic tail of E-cadherin to achieve intercellular adhesion (36,37). The inhibition of canonical Wnt signaling has been identified as an effective approach in inhibiting the advancement of gastric cancer (38). Moreover, Wnt-PCP signaling is triggered by Wnt4, Wnt5a, Wnt5b, Wnt7b and Wnt11 (39,40). Research into the Ca2+-Wnt signaling pathway is lacking; however, this plays a key role in determining cell fate (41), cancer progression (42,43) and inflammatory response (44) during early embryogenesis. Fzd2 and Wnt5a are the main initiators of the Wnt-Ca2+ signaling pathway (45). Results of a recent study revealed that a non-canonical Wnt signaling pathway contributes to the establishment of a cancer stem cell niche in cancer-associated fibroblasts (CAFs) (46). In summary, Wnt signaling plays an important role in the occurrence and development of tumors.

The role of canonical Wnt in BC development and therapy

The present review focused on canonical Wnt signaling (Fig. 1). Results of a recent study demonstrated that canonical Wnt signaling enhanced melanocyte regeneration; however, this suppressed the invasion, migration and proliferation of melanoma cells (13). In the absence of Wnt signaling, β-catenin binds to cytoplasmic complexes containing casein kinase-1-alpha (CK1Alpha), glycogen synthase kinase-3-beta (GSK3β), axis inhibitor (Axin) and adenomatous polyposis colon proteins (47). This process enhances the phosphorylation of β-catenin and its association with β-transducing protein repeat sequence protein, leading to the ubiquitination of β-catenin and its subsequent breakdown by the proteasome. Results of a recent study revealed that inhibition of Wnt signaling promotes apoptosis of human colorectal cancer cells (48). When Wnt signaling is activated in response to Wnt binding to Frizzled, the Dishevelled (DSH) protein is activated (49). Activated DSH protein enhances the phosphorylation of GSK3β, thereby leading to inhibition of this protein. This results in the accumulation of free unphosphorylated β-catenin in the cytoplasm, which is subsequently transported to the nucleus. Prior to Wnt signaling transduction, lymphoid enhance factor (LEF) and T-cell factor (TCF) bind to specific sequences of the promoter/enhancer region of the target gene and, together with Groucho and HDAC, inhibit gene expression in the nucleus. Wnt signaling leads to elevated levels of β-catenin in the nucleus, which binds to TCF/LEF and promotes alterations in transcriptional mechanisms, resulting in the activation of several target genes. In addition, KDM2A promotes the degradation of TCF/LEF transcription factors, which regulate canonical Wnt signaling (50). The transfer of proteins from the E-cadherin-binding pool to the cytoplasmic pool increases the amount of free β-catenin available to activate target genes. Interactions between β-catenin and histone acetyltransferase CBP (CREb-binding protein), chromatin remodeling SWI/SNF complexes, and the binding of BCL-9 to Pygopus (Pygo) and BRG1 mediate transcriptional activation. Notably, results of a previous study revealed that the Wnt/β-catenin/BCL-9 signaling pathway impacts the proliferation of multiple myeloma cells (51). CHIBBY directly interacts with the C-terminal region of β-catenin; thus, inhibiting β-catenin-mediated transcriptional activation through competitive binding with LEF1.

Results of previous studies demonstrated that activation or silencing of the Wnt signaling pathway impact epithelial-mesenchymal transition (EMT)-dependent metastasis, the immune microenvironment and the resistance of BC (5254). Notably, cells undergo EMT during the differentiation of epithelial cells into mesenchymal cells (55). EMT is an important feature of BC, and plays a key role in triple negative BC (56). Thus, research is focused on regulating EMT for the treatment of BC (57). Li et al (58) demonstrated that activation of Wnt signaling enhanced the invasion and metastasis of BC (58). Notably, a number of intrinsic EMT-transcription factors (TFs) are mechanically activated by Wnt/catenin signaling, epidermal growth factor (EGF)/fibroblast growth factor (FGF)-receptor tyrosine kinase signaling and Notch signaling. These pathways also initiate changes in the expression of genes, including E-cadherin and ZO-1. In addition, the aforementioned pathways may activate N-Cadherin, MMPs, integrins and fibronectin (59). EMT-TFs expressed by Wnt regulate BC morphogenesis, including lamellipodia formation, and directly secrete MMPs, resulting in their migration and invasion (60). Results of a previous study revealed that Wnt/β-catenin signaling suppresses antitumor immunity (61). A cancerous breast cell that expresses Wnt signaling may develop strategies to avoid being recognized and destroyed by the immune system (62). To prevent phagocytosis by macrophages, BC cells express CD24 and CD47 through interactions with Siglec-10 and SIRP-α, respectively (63,64). Notably, CD24 directly targets Wnt1, while CD47 indirectly targets SNAI1 and ZEB1 through Wnt signaling in BC (65,66). Therefore, Wnt signaling plays a key role in the immune microenvironment of BC. Metastatic BC is characterized by frequent changes in the TP53 gene (67). The loss of TP53 in BC cells trigger the secretion of Wnt1, Wnt6 and Wnt7a (31). These proteins bind to Fzd7 and Fzd9 on the surface of TAM, stimulating the production of IL-1 by TAMβ. Results of a previous study revealed that mutations in TP53 may lead to drug resistance in BC (68). For example, TP53 mutations are associated with endocrine therapeutic resistance in early luminal BC (69). Wnt-driven systemic inflammation and immunosuppression niches are associated with BC multidrug resistance. Cancer resistance is considered a multifaceted issue, involving tumor heterogeneity, drug efflux/inactivation and activation of survival pathways (70). Results of a previous study demonstrated that inactivation of Wnt signaling leads to BC entering a drug insensitive resting state (71), leading to multidrug resistance. Thus, Wnt signaling is a dynamic and multifaceted process in BC, and Wnt signaling may exhibit potential as a target in the treatment of BC.

The function of non-canonical Wnt in BC development

The non-canonical Wnt pathways include the Wnt-PCP signaling pathway and the Wnt-Ca2+ signaling pathway. Genetically engineered mice exhibit increased distant metastasis and collective cell migration following Vangl-dependent Wnt-PCP signaling (72). In basal BC, overexpression of the Wnt/PCP protein, VANGL2, is associated with a poor prognosis and an increase in tumor size (73). In addition, results of a previous study revealed that exosomes released by fibroblasts activate Wnt-PCP signaling to drive BC cell invasion (14). Secreted fried-associated protein 2 (SFRP2) is overexpressed in the blood vessels of 85% of human breast tumors, and SFRP2 promotes tumor angiogenesis through the Wnt-Ca2+ pathway (74). At present, research is focused on the non-canonical Wnt pathway in BC, and this pathway exhibits potential in further understanding the pathogenesis of BC.

The value of exosomal miRNAs in BC

Exosomes are membrane-bound microvesicles that range from 30–150 nanometers in size, and these are secreted into the extracellular environment by all cells, including prokaryotes and eukaryotes. Exosomes contain a diverse array of miRNAs, mRNA, proteins, lipids and other substances (75). They play a crucial role in facilitating intercellular material exchange and information transmission. miRNAs accelerate mRNA degradation or inhibit mRNA translation through interactions with the 3′-untranslated region of target mRNAs. This regulation of post-transcriptional gene expression in recipient cells has been observed in various models (23). Abnormal expression or mutations in miRNAs are associated with a range of tumors, including BC, where they function as oncogenes or tumor suppressors (76,77). However, miRNAs are inherently unstable. Exosomes, with a phospholipid bilayer membrane structure, provide stability to miRNAs via protection from enzymatic degradation (78). Therefore, exosomal miRNAs are valuable for understanding the pathogenesis of malignant tumors. Extensive research has demonstrated that exosomal miRNAs exhibit potential as biomarkers and therapeutic targets in BC (Fig. 2).

Exosomal miRNAs as potential biomarkers

A biomarker quantifies a normal biological or pathological process, or an impact of a therapeutic intervention (79). Exosomal miRNAs exhibit potential as biomarkers in the prediction, diagnosis and prognosis of BC (80). The present study demonstrated that serum exosomal miR-21 may be used as a biomarker for the early detection and diagnosis of BC (81). Moreover, the plasma exosomal miR-21-5p has also been identified as a potential biomarker for the diagnosis of BC (82). Li et al (83) demonstrated that exosomes miR-3662, miR-146a and miR-1290 exhibit potential in predicting disease, and these may be used as biomarkers for diagnosis and treatment. Through monitoring exosomal miRNAs, BC occurrence, recurrence, prognosis and responses to common therapies can be predicted.

Exosomal miRNAs and apoptosis

Apoptosis plays a key role in promoting tumor occurrence. Exosomal miRNAs play a significant role in the apoptosis of BC cells, exhibiting abnormal expression patterns in patients (84). Notably, BC cells release exosomes containing miR-1910-3p, which inhibits apoptosis and facilitates tumor development through the transfer of miR-1910-3p to target cells (85). In addition, Wei et al (86) revealed that miR-128 is specifically sorted into exosomes and enhances the proliferation of MCF-7 cells through targeting the Bax gene. Bax plays a key role in the inhibition of apoptosis. A previous study identified exosomal miR-134-5p as a potential therapeutic target for BC, as it promotes apoptosis through inhibiting the PI3K/AKT pathway (86).

Exosomal miRNAs and angiogenesis

In hypoxic environments, the secretion of exosomal miRNAs by cancer cells is upregulated to modulate tumor angiogenesis, a critical and dynamic process in the progression of tumorigenesis (87). Exosomal miR-210 enhances angiogenesis through the modulation of vascular remodeling-associated genes, Ephrin A3 and PTP1B, consequently influencing the development of BC and the dissemination of hypoxic BC cells to adjacent tissues (88). Results of previous studies demonstrated that exosomal miRNAs play a significant role in promoting vascular survival through the regulation of FGF, vascular endothelial growth factor (VEGF), EGF and angiopoietin-1, ultimately contributing to the progression and metastasis of BC (89,90). In patients with BC, miR-221, miR-27b and miR-132 enhance angiogenesis through modulating the angiogenic properties of VEGF (91,92). However, several exosomal miRNAs have been identified as inhibitors of tumor angiogenesis. Notably, exosomal miR-16 inhibits angiogenesis in BC cells through the direct regulation of VEGF expression (93). Overall, exosomal miRNAs play a role in modulating the progression of BC via angiogenesis.

Exosomal miRNAs and EMT

EMT facilitates the acquisition of metastatic capabilities by epithelial cells, serving as a crucial prerequisite for metastasis. Results of a previous study demonstrated that exosomes released by CAFs transfer specific miRNAs, including exosomal miR-21, miR-378e and miR-143 to BC cells, promoting EMT characteristics (94). Yan et al (95) further elucidated that CAF-derived exosomal miR-18b induces EMT, invasion and metastasis in BC through targeting TCEAL7 to activate the NF-κB signaling pathway. Wang et al (96) demonstrated that exosomal miR-181d-5p derived from CAFs promotes the proliferation, invasion, migration and EMT of BC cells through regulating CDX2 and HOXA5 genes (96). Thus, targeting CAF-derived exosomal miR-18b and miR-181d-5p may exhibit potential in the treatment of BC. In addition, miR-103-107 may inhibit miRNA biosynthesis through targeting the Dicer gene in BC, resulting in enhanced EMT and metastatic characteristics in epithelial tumor cells (97).

Moreover, exosomal miRNAs exhibit anticancer properties in BC through the inhibition of EMT. For example, miR-34a, a transcriptional target of p53, suppresses the aggressiveness of BC cells through targeting EMT and the zinc finger transcriptional inhibitor, Snail (98). In addition, exosomal miR-16-5p attenuates EMT via downregulation of EPHA1 and NF-κB signaling pathways, ultimately impeding the proliferation, invasion and migration of BC cells (99). Moreover, miR-7-5p exhibits differential expression levels in various invasive BC cell lines. miR-7-5p inhibits EMT through targeting RYK and decreasing the phosphorylation of JNK, thereby reducing the metastasis of BC (100).

Exosomal miRNAs and tumor immunity

Exosomal miRNAs play a significant role in mediating the communication between BC cells and immune cells, thereby influencing immune regulation. Results of previous studies demonstrated the involvement of exosomal miRNAs in modulating the polarization of macrophages and the secretion of proinflammatory cytokines (101,102). Specifically, M1 macrophages exhibit a pro-inflammatory phenotype, characterized by the expression of cytokines, such as IL-12. This may contribute to the destruction of cancer cells. On the other hand, M2 macrophages produce anti-inflammatory cytokines, such as IL-10, thus promoting tumor progression (103). BC is characterized by the presence of tumor-associated macrophages (TAMs), predominantly displaying the M2 phenotype (104). Results of previous studies demonstrated that BC-derived exosomal miR-16 and miR-33 inhibit the M1 polarization of TAMs, through suppressing the expression of epigenetic factors, while simultaneously stimulating M2 polarization (102,105). This mechanism ultimately facilitates the advancement of metastatic BC. Previous research has focused on the miRNA-mediated inhibition of mRNA translation in the regulation of endoplasmic reticulum stress and immune evasion in human tumors. Specifically, in the context of endoplasmic reticulum stress, BC exosomes containing miR-27a-3p, miR-25-93-106b and miR-92 enhance the expression of PD-L1 in macrophages, thereby facilitating immune evasion (106,107). These findings suggest that targeting exosomal miRNAs may exhibit potential in the treatment of BC.

Exosomal miRNAs and drug resistance

Primary breast tumors often respond well to initial treatment, and develop drug resistance after a few months (70). At present, research is focused on exosomal miRNA-mediated drug resistance mechanisms in BC cells (108). Results of a previous study highlighted that modulation of exosome miRNA expression may impact the responsiveness of BC cells to hormonal treatments, targeted therapies and chemotherapeutic agents via diverse signaling pathways (109). BC is categorized according to hormone receptor expression, with ~70% of BC cases being labelled as estrogen receptor-positive. These cases are treated with anti-estrogen drugs, such as tamoxifen or fluoxetine; however, cells may develop resistance. Notably, exosomal miR-101 and miR-301 may cause BC cells to become resistant to tamoxifen, through the inhibition of PTEN (110). Exosomal miR-221 and miR-222 increase the therapeutic resistance of sensitive MCF7 BC cells to tamoxifen through downregulation of p27 and ERα targets. Exosomal miR-221/222 also promote BC cell resistance to fluoxetine through dysfunction of TGF-β and β-catenin signaling networks; thus, impacting the survival of patients with ER-positive BC (111,112). Results of a previous study revealed a high correlation between changes in exosomal miRNA expression and adriamycin resistance. For example, upregulation of miR-145 may sensitize BC to doxorubicin chemotherapy (113). In conclusion, exosomal miRNAs may exhibit potential as targets for increasing the chemosensitivity of BC.

Crosstalk between exosomal miRNAs and Wnt signaling

The role of exosomal miRNAs in the regulation of Wnt Signaling

Exosomal miRNAs may impact various aspects of tumor progression through regulation of Wnt signaling. Patients with recurring BC exhibited significantly lower levels of exosomal miR-18a-5p (114). Results of a recent study demonstrated that exosomal miR-18a-5p promotes the EMT and metastasis of nasopharyngeal carcinoma cells through activating the Wnt/β-catenin signaling pathway (115). In addition, the migration and invasion of BC cells are promoted via the downregulation of exosomal miR-7-5p through WNT signaling (100). In addition, exosomal miR-1260b promotes cell invasion through the Wnt/β-catenin signaling pathway (116). miRNA-1260b also plays a role in promoting tumor invasion in BC (117). Exosomal miR-10527-5p inhibits migration, invasion, lymphangiogenesis and lymphatic metastasis via Wnt/β-catenin signaling (118). In cancer cells, exosome-derived miR-375 targets DIP2C and regulates Wnt signaling, thus promoting osteoblastic metastasis (119). Li et al (120) revealed that exosomal miR-92a promotes cytarabine resistance through activating the Wnt/β-catenin signaling pathway (120). In conclusion, exosomal miRNAs may influence tumor progression through regulating Wnt signaling.

Wnt signaling may impact the expression of exosomal miRNAs

Results of previous studies highlighted that activation of Wnt/β-catenin signaling may impact the expression of exosomal miR-301a and promote resistance to radiation (121,122). In addition, Wnt signaling impacts the release of exosomal miR-454 to maintain the biological properties of cancer stem cells through BC cells (123). Exosomal miR-1323 is involved in cervical cancer progression and resistance to radiation, which may exhibit potential in the treatment of cervical cancer (124). In addition, the chemosensitivity of bladder cancer cells was increased via Wnt/β-catenin pathway-mediated downregulation of exosomal miR-148b-3p in CAFs (125). In conclusion, the release of miRNA in exosomes may be influenced by the activation or deactivation of Wnt signaling. Thus, regulation of Wnt signaling may regulate the expression of exosomal miRNAs in BC.

Conclusions

At present, research is focused on the role of exosomal miRNAs in BC. Results of previous studies highlighted the involvement of multiple exosomal miRNAs in various aspects of BC progression, including apoptosis regulation, cell metastasis, tumor immunity, drug resistance and modulation of Wnt signaling pathways. Thus, targeting multiple biological processes of exosomal miRNAs may exhibit potential in the treatment of BC (Fig. 3). At present, the use of exosomal miRNAs is limited due to difficulties in batch isolation and the extraction of exosomes. However, further investigations into the role of exosomal miRNAs and Wnt signaling pathways in BC may enhance the current understanding of the pathogenesis of BC, and aid in the development of exosomal miRNA-based therapies. In conclusion, exosomal miRNAs and Wnt signaling exhibit potential as effective diagnostic and therapeutic targets for BC. Further investigations into the regulatory mechanisms of exosomal miRNAs in Wnt signaling are required for the development of novel diagnostic and therapeutic targets in BC.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

HL organized the manuscript and produced the figures. XL completed the exosome section of the manuscript. WD provided the outline of the present review and completed the ‘Conclusions’ section of the manuscript. All authors read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Harbeck N, Penault-Llorca F, Cortes J, Gnant M, Houssami N, Poortmans P, Ruddy K, Tsang J and Cardoso F: Breast cancer. Nat Rev Dis Primers. 5:662019. View Article : Google Scholar : PubMed/NCBI

2 

Hachey SJ, Hatch CJ, Gaebler D, Mocherla A, Nee K, Kessenbrock K and Hughes CCW: Targeting tumor-stromal interactions in triple-negative breast cancer using a human vascularized micro-tumor model. Breast Cancer Res. 26:52024. View Article : Google Scholar : PubMed/NCBI

3 

Abeni E, Grossi I, Marchina E, Coniglio A, Incardona P, Cavalli P, Zorzi F, Chiodera PL, Paties CT, Crosatti M, et al: DNA methylation variations in familial female and male breast cancer. Oncol Lett. 21:4682021. View Article : Google Scholar : PubMed/NCBI

4 

Siegel RL, Miller KD, Fuchs HE and Jemal A: Cancer statistics, 2021. CA Cancer J Clin. 71:7–33. 2021. View Article : Google Scholar : PubMed/NCBI

5 

McDonald ES, Clark AS, Tchou J, Zhang P and Freedman GM: Clinical diagnosis and management of breast cancer. J Nucl Med. 57 (Suppl 1):9S–16S. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Rim EY, Clevers H and Nusse R: The wnt pathway: From signaling mechanisms to synthetic modulators. Annu Rev Biochem. 91:571–598. 2022. View Article : Google Scholar : PubMed/NCBI

7 

Zhang Z, Lin X, Wei L, Wu Y, Xu L, Wu L, Wei X, Zhao S, Zhu X and Xu F: A framework for Frizzled-G protein coupling and implications to the PCP signaling pathways. Cell Discov. 10:32024. View Article : Google Scholar : PubMed/NCBI

8 

Wang K, Ma F, Arai S, Wang Y, Varkaris A, Poluben L, Voznesensky O, Xie F, Zhang X, Yuan X and Balk SP: WNT5a signaling through ROR2 activates the hippo pathway to suppress YAP1 activity and tumor growth. Cancer Res. 83:1016–1030. 2023. View Article : Google Scholar : PubMed/NCBI

9 

Neiheisel A, Kaur M, Ma N, Havard P and Shenoy AK: Wnt pathway modulators in cancer therapeutics: An update on completed and ongoing clinical trials. Int J Cancer. 150:727–740. 2022. View Article : Google Scholar : PubMed/NCBI

10 

Nusse R and Varmus HE: Many tumors induced by the mouse mammary tumor virus contain a provirus integrated in the same region of the host genome. Cell. 31:99–109. 1982. View Article : Google Scholar : PubMed/NCBI

11 

van Ooyen A and Nusse R: Structure and nucleotide sequence of the putative mammary oncogene int-1; proviral insertions leave the protein-encoding domain intact. Cell. 39:233–240. 1984. View Article : Google Scholar : PubMed/NCBI

12 

Wend P, Runke S, Wend K, Anchondo B, Yesayan M, Jardon M, Hardie N, Loddenkemper C, Ulasov I, LesniakM S, et al: WNT10B/β-catenin signalling induces HMGA2 and proliferation in metastatic triple-negative breast cancer. EMBO Mol Med. 5:264–279. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Katkat E, Demirci Y, Heger G, Karagulle D, Papatheodorou I, Brazma A and Ozhan G: Canonical Wnt and TGF-β/BMP signaling enhance melanocyte regeneration but suppress invasiveness, migration, and proliferation of melanoma cells. Front Cell Dev Biol. 11:12979102023. View Article : Google Scholar : PubMed/NCBI

14 

Luga V, Zhang L, Viloria-Petit AM, Ogunjimi AA, Inanlou MR, Chiu E, Buchanan M, Hosein AN, Basik M and Wrana JL: Exosomes mediate stromal mobilization of autocrine Wnt-PCP signaling in breast cancer cell migration. Cell. 151:1542–1556. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Harper KL, Sosa MS, Entenberg D, Hosseini H, Cheung JF, Nobre R, Avivar-Valderas A, Nagi C, Girnius N, Davis RJ, et al: Mechanism of early dissemination and metastasis in Her2(+) mammary cancer. Nature. 540:588–592. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Malladi S, Macalinao DG, Jin X, He L, Basnet H, Zou Y, de Stanchina E and Massagué J: Metastatic latency and immune evasion through autocrine inhibition of WNT. Cell. 165:45–60. 2016. View Article : Google Scholar : PubMed/NCBI

17 

Leung CON, Yang Y, Leung RWH, So KKH, Guo HJ, Lei MML, Muliawan GK, Gao Y, Yu QQ, Yun JP, et al: Broad-spectrum kinome profiling identifies CDK6 upregulation as a driver of lenvatinib resistance in hepatocellular carcinoma. Nat Commun. 14:66992023. View Article : Google Scholar : PubMed/NCBI

18 

Piva M, Domenici G, Iriondo O, Rábano M, Simões BM, Comaills V, Barredo I, López-Ruiz JA, Zabalza I, Kypta R and Vivanco M: Sox2 promotes tamoxifen resistance in breast cancer cells. EMBO Mol Med. 6:66–79. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Shi J, Wang Y, Zeng L, Wu Y, Deng J, Zhang Q, Lin Y, Li J, Kang T, Tao M, et al: Disrupting the interaction of BRD4 with diacetylated Twist suppresses tumorigenesis in basal-like breast cancer. Cancer Cell. 25:210–225. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Kahn M: Can we safely target the WNT pathway? Nat Rev Drug Discov. 13:513–532. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Pegtel DM and Gould SJ: Exosomes. Annu Rev Biochem. 88:487–514. 2019. View Article : Google Scholar : PubMed/NCBI

22 

Chen XJ, Guo CH, Wang ZC, Yang Y, Pan YH, Liang JY, Sun MG, Fan LS, Liang L and Wang W: Hypoxia-induced ZEB1 promotes cervical cancer immune evasion by strengthening the CD47-SIRPα axis. Cell Commun Signal. 22:152024. View Article : Google Scholar : PubMed/NCBI

23 

Yu X, Odenthal M and Fries JW: Exosomes as miRNA carriers: Formation-function-future. Int J Mol Sci. 17:20282016. View Article : Google Scholar : PubMed/NCBI

24 

Zhu L, Sun HT, Wang S, Huang SL, Zheng Y, Wang CQ, Hu BY, Qin W, Zou TT, Fu Y, et al: Isolation and characterization of exosomes for cancer research. J Hematol Oncol. 13:1522020. View Article : Google Scholar : PubMed/NCBI

25 

Li X, Han Y, Meng Y and Yin L: Small RNA-big impact: Exosomal miRNAs in mitochondrial dysfunction in various disease. RNA Biol. 21:1–20. 2024. View Article : Google Scholar

26 

Sun Z, Shi K, Yang S, Liu J, Zhou Q, Wang G, Song J, Li Z, Zhang Z and Yuan W: Effect of exosomal miRNA on cancer biology and clinical applications. Mol Cancer. 17:1472018. View Article : Google Scholar : PubMed/NCBI

27 

Lakshmi S, Hughes TA and Priya S: Exosomes and exosomal RNAs in breast cancer: A status update. Eur J Cancer. 144:252–268. 2021. View Article : Google Scholar : PubMed/NCBI

28 

Zhao Y, Jin LJ and Zhang XY: Exosomal miRNA-205 promotes breast cancer chemoresistance and tumorigenesis through E2F1. Aging (Albany NY). 13:18498–18514. 2021. View Article : Google Scholar : PubMed/NCBI

29 

Scognamiglio I, Cocca L, Puoti I, Palma F, Ingenito F, Quintavalle C, Affinito A, Roscigno G, Nuzzo S, Chianese RV, et al: Exosomal microRNAs synergistically trigger stromal fibroblasts in breast cancer. Mol Ther Nucleic Acids. 28:17–31. 2022. View Article : Google Scholar : PubMed/NCBI

30 

Zhan T, Rindtorff N and Boutros M: Wnt signaling in cancer. Oncogene. 36:1461–1473. 2017. View Article : Google Scholar : PubMed/NCBI

31 

Wellenstein MD, Coffelt SB, Duits DEM, van Miltenburg MH, Slagter M, de Rink I, Henneman L, Kas SM, Prekovic S, Hau CS, et al: Loss of p53 triggers WNT-dependent systemic inflammation to drive breast cancer metastasis. Nature. 572:538–542. 2019. View Article : Google Scholar : PubMed/NCBI

32 

Staal FJ and Clevers HC: WNT signalling and haematopoiesis: A WNT-WNT situation. Nat Rev Immunol. 5:21–30. 2005. View Article : Google Scholar : PubMed/NCBI

33 

Sidaway P: Prostate cancer: Wnt signalling induces resistance. Nat Rev Urol. 12:5972015. View Article : Google Scholar : PubMed/NCBI

34 

Xu X, Zhang M, Xu F and Jiang S: Wnt signaling in breast cancer: Biological mechanisms, challenges and opportunities. Mol Cancer. 19:1652020. View Article : Google Scholar : PubMed/NCBI

35 

Xiao Q and Chen Z, Jin X, Mao R and Chen Z: The many postures of noncanonical Wnt signaling in development and diseases. Biomed Pharmacother. 93:359–369. 2017. View Article : Google Scholar : PubMed/NCBI

36 

Ozawa M, Baribault H and Kemler R: The cytoplasmic domain of the cell adhesion molecule uvomorulin associates with three independent proteins structurally related in different species. EMBO J. 8:1711–1717. 1989. View Article : Google Scholar : PubMed/NCBI

37 

McCrea PD and Gumbiner BM: Purification of a 92-kDa cytoplasmic protein tightly associated with the cell-cell adhesion molecule E-cadherin (uvomorulin). Characterization and extractability of the protein complex from the cell cytostructure. J Biol Chem. 266:4514–4520. 1991. View Article : Google Scholar : PubMed/NCBI

38 

Zhan T, Chen M, Liu W, Han Z, Zhu Q, Liu M, Tan J, Liu J, Chen X, Tian X and Huang X: MiR-455-3p inhibits gastric cancer progression by repressing Wnt/β-catenin signaling through binding to ARMC8. BMC Med Genomics. 16:1552023. View Article : Google Scholar : PubMed/NCBI

39 

Yang Y and Mlodzik M: Wnt-Frizzled/planar cell polarity signaling: Cellular orientation by facing the wind (Wnt). Annu Rev Cell Dev Biol. 31:623–646. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Katoh M: WNT/PCP signaling pathway and human cancer (review). Oncol Rep. 14:1583–1588. 2005.PubMed/NCBI

41 

Saneyoshi T, Kume S, Amasaki Y and Mikoshiba K: The Wnt/calcium pathway activates NF-AT and promotes ventral cell fate in Xenopus embryos. Nature. 417:295–299. 2002. View Article : Google Scholar : PubMed/NCBI

42 

Liang H, Chen Q, Coles AH, Anderson SJ, Pihan G, Bradley A, Gerstein R, Jurecic R and Jones SN: Wnt5a inhibits B cell proliferation and functions as a tumor suppressor in hematopoietic tissue. Cancer Cell. 4:349–360. 2003. View Article : Google Scholar : PubMed/NCBI

43 

Zhuang X, Zhang H, Li X, Li X, Cong M, Peng F, Yu J, Zhang X, Yang Q and Hu G: Differential effects on lung and bone metastasis of breast cancer by Wnt signalling inhibitor DKK1. Nat Cell Biol. 19:1274–1285. 2017. View Article : Google Scholar : PubMed/NCBI

44 

Mahdi T, Hänzelmann S, Salehi A, Muhammed SJ, Reinbothe TM, Tang Y, Axelsson AS, Zhou Y, Jing X, Almgren P, et al: Secreted frizzled-related protein 4 reduces insulin secretion and is overexpressed in type 2 diabetes. Cell Metab. 16:625–633. 2012. View Article : Google Scholar : PubMed/NCBI

45 

Slusarski DC, Corces VG and Moon RT: Interaction of wnt and a frizzled homologue triggers g-protein-linked phosphatidylinositol signalling. Nature. 390:410–413. 1997. View Article : Google Scholar : PubMed/NCBI

46 

Fang Y, Xiao X, Wang J, Dasari S, Pepin D, Nephew KP, Zamarin D and Mitra AK: Cancer associated fibroblasts serve as an ovarian cancer stem cell niche through noncanonical Wnt5a signaling. NPJ Precis Oncol. 8:72024. View Article : Google Scholar : PubMed/NCBI

47 

Ge J, Yu YJ, Li JY, Li MY, Xia SM, Xue K, WangS Y and Yang C: Activating Wnt/β-catenin signaling by autophagic degradation of APC contributes to the osteoblast differentiation effect of soy isoflavone on osteoporotic mesenchymal stem cells. Acta Pharmacol Sin. 44:1841–1855. 2023. View Article : Google Scholar : PubMed/NCBI

48 

Zhu Y, Zhang E, Gao H, Shang C, Yin M, Ma M, Liu Y, Zhang X and Li X: Resistomycin inhibits Wnt/β-catenin signaling to induce the apoptotic death of human colorectal cancer cells. Mar Drugs. 21:6222023. View Article : Google Scholar : PubMed/NCBI

49 

Rui Q, Dong S, Jiang W and Wang D: Response of canonical Wnt/β-catenin signaling pathway in the intestine to microgravity stress in Caenorhabditis elegans. Ecotoxicol Environ Saf. 186:1097822019. View Article : Google Scholar : PubMed/NCBI

50 

Šopin T, Liška F, Kučera T, Cmarko D and Vacík T: Lysine demethylase KDM2A promotes proteasomal degradation of TCF/LEF transcription factors in a neddylation-dependent manner. Cells. 12:26202023. View Article : Google Scholar : PubMed/NCBI

51 

Xu Y, Yang Z, Yuan H, Li Z, Li Y, Liu Q and Chen J: PCDH10 inhibits cell proliferation of multiple myeloma via the negative regulation of the Wnt/β-catenin/BCL-9 signaling pathway. Oncol Rep. 34:747–754. 2015. View Article : Google Scholar : PubMed/NCBI

52 

Wang C, Zhang R, Wang X, Zheng Y, Jia H, Li H, Wang J, Wang N, Xiang F and Li Y: Silencing of KIF3B suppresses breast cancer progression by regulating EMT and Wnt/β-catenin signaling. Front Oncol. 10:5974642020. View Article : Google Scholar : PubMed/NCBI

53 

Malla RR and Kiran P: Tumor microenvironment pathways: Cross regulation in breast cancer metastasis. Genes Dis. 9:310–324. 2022. View Article : Google Scholar : PubMed/NCBI

54 

Wang L, Jin Z, Master RP, Maharjan CK, Carelock ME, Reccoppa TBA, Kim MC, Kolb R and Zhang W: Breast cancer stem cells: Signaling pathways, cellular interactions, and therapeutic implications. Cancers (Basel). 14:32872022. View Article : Google Scholar : PubMed/NCBI

55 

Pastushenko I and Blanpain C: EMT transition states during tumor progression and metastasis. Trends Cell Biol. 29:212–226. 2019. View Article : Google Scholar : PubMed/NCBI

56 

Dri A, Arpino G, Bianchini G, Curigliano G, Danesi R, De Laurentiis M, Del Mastro L, Fabi A, Generali D, Gennari A, et al: Puglisi, Breaking barriers in triple negative breast cancer (TNBC)-Unleashing the power of antibody-drug conjugates (ADCs). Cancer Treat Rev. 123:1026722024. View Article : Google Scholar : PubMed/NCBI

57 

Park M, Kim D, Ko S, Kim A, Mo K and Yoon H: Breast cancer metastasis: Mechanisms and therapeutic implications. Int J Mol Sci. 23:68062022. View Article : Google Scholar : PubMed/NCBI

58 

Li Y, Jin K, van Pelt GW, van Dam H, Yu X, Mesker WE, Dijke PT, Zhou F and Zhang L: c-Myb enhances breast cancer invasion and metastasis through the Wnt/β-catenin/Axin2 pathway. Cancer Res. 76:3364–3375. 2016. View Article : Google Scholar : PubMed/NCBI

59 

Lamouille S, Xu J and Derynck R: Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol. 15:178–196. 2014. View Article : Google Scholar : PubMed/NCBI

60 

Yang J, Mani SA, Donaher JL, Ramaswamy S, Itzykson RA, Come C, Savagner P, Gitelman I, Richardson A and Weinberg RA: Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell. 117:927–939. 2004. View Article : Google Scholar : PubMed/NCBI

61 

Pai SG, Carneiro BA, Mota JM, Costa R, Leite CA, Barroso-Sousa R, Kaplan JB, Chae YK and Giles FJ: Wnt/beta-catenin pathway: Modulating anticancer immune response. J Hematol Oncol. 10:1012017. View Article : Google Scholar : PubMed/NCBI

62 

Wang Q, Chen F, Yang N, Xu L, Yu X, Wu M and Zhou Y: DEPDC1B-mediated USP5 deubiquitination of β-catenin promotes breast cancer metastasis by activating the wnt/β-catenin pathway. Am J Physiol Cell Physiol. 325:C833–C848. 2023. View Article : Google Scholar : PubMed/NCBI

63 

Barkal AA, Brewer RE, Markovic M, Kowarsky M, Barkal SA, Zaro BW, Krishnan V, Hatakeyama J, Dorigo O, Barkal LJ and Weissman IL: CD24 signalling through macrophage Siglec-10 is a target for cancer immunotherapy. Nature. 572:392–396. 2019. View Article : Google Scholar : PubMed/NCBI

64 

Oldenborg PA, Zheleznyak A, Fang YF, Lagenaur CF, Gresham HD and Lindberg FP: Role of CD47 as a marker of self on red blood cells. Science. 288:2051–2054. 2000. View Article : Google Scholar : PubMed/NCBI

65 

Shulewitz M, Soloviev I, Wu T, Koeppen H, Polakis P and Sakanaka C: Repressor roles for TCF-4 and Sfrp1 in Wnt signaling in breast cancer. Oncogene. 25:4361–4369. 2006. View Article : Google Scholar : PubMed/NCBI

66 

Noman MZ, Van Moer K, Marani V, Gemmill RM, Tranchevent LC, Azuaje F, Muller A, Chouaib S, Thiery JP, Berchem G and Janji B: CD47 is a direct target of SNAI1 and ZEB1 and its blockade activates the phagocytosis of breast cancer cells undergoing EMT. Oncoimmunology. 7:e13454152018. View Article : Google Scholar : PubMed/NCBI

67 

Blondeaux E, Arecco L, Punie K, Graffeo R, Toss A, De Angelis C, Trevisan L, Buzzatti G, Linn SC, Dubsky P, et al: Germline TP53 pathogenic variants and breast cancer: A narrative review. Cancer Treat Rev. 114:1025222023. View Article : Google Scholar : PubMed/NCBI

68 

Huang X, Shi D, Zou X, Wu X, Huang S, Kong L, Yang M, Xiao Y, Chen B, Chen X, et al: BAG2 drives chemoresistance of breast cancer by exacerbating mutant p53 aggregate. Theranostics. 13:339–354. 2023. View Article : Google Scholar : PubMed/NCBI

69 

Grote I, Bartels S, Kandt L, Bollmann L, Christgen H, Gronewold M, Raap M, Lehmann U, Gluz O, Nitz U, et al: TP53 mutations are associated with primary endocrine resistance in luminal early breast cancer. Cancer Med. 10:8581–8594. 2021. View Article : Google Scholar : PubMed/NCBI

70 

Vasan N, Baselga J and Hyman DM: A view on drug resistance in cancer. Nature. 575:299–309. 2019. View Article : Google Scholar : PubMed/NCBI

71 

Bai X, Ni J, Beretov J, Graham PA and Li Y: Cancer stem cell in breast cancer therapeutic resistance. Cancer Treat Rev. 69:152–163. 2018. View Article : Google Scholar : PubMed/NCBI

72 

VanderVorst K, Dreyer CA, Hatakeyama J, Bell GRR, Learn JA, Berg AL, Hernandez M, Lee H, Collins SR and Carraway KL III: Vangl-dependent Wnt/planar cell polarity signaling mediates collective breast carcinoma motility and distant metastasis. Breast Cancer Res. 25:522023. View Article : Google Scholar : PubMed/NCBI

73 

Puvirajesinghe TM, Bertucci F, Jain A, Scerbo P, Belotti E, Audebert S, Sebbagh M, Lopez M, Brech A, Finetti P, et al: Identification of p62/SQSTM1 as a component of non-canonical Wnt VANGL2-JNK signalling in breast cancer. Nat Commun. 7:103182016. View Article : Google Scholar : PubMed/NCBI

74 

Courtwright A, Siamakpour-Reihani S, Arbiser JL, Banet N, Hilliard E, Fried L, Livasy C, Ketelsen D, Nepal DB, Perou CM, et al: Secreted frizzle-related protein 2 stimulates angiogenesis via a calcineurin/NFAT signaling pathway. Cancer Res. 69:4621–4628. 2009. View Article : Google Scholar : PubMed/NCBI

75 

Kalluri R and LeBleu VS: The biology, function, and biomedical applications of exosomes. Science. 367:eaau69772020. View Article : Google Scholar : PubMed/NCBI

76 

Hu JL, Wang W, Lan XL, Zeng ZC, Liang YS, Yan YR, Song FY, Wang FF, Zhu XH, Liao WJ, et al: CAFs secreted exosomes promote metastasis and chemotherapy resistance by enhancing cell stemness and epithelial-mesenchymal transition in colorectal cancer. Mol Cancer. 18:912019. View Article : Google Scholar : PubMed/NCBI

77 

Li BL, Lu W, Qu JJ, Ye L, Du GQ and Wan XP: Loss of exosomal miR-148b from cancer-associated fibroblasts promotes endometrial cancer cell invasion and cancer metastasis. J Cell Physiol. 234:2943–2953. 2019. View Article : Google Scholar : PubMed/NCBI

78 

Kim CK and Pak TR: miRNA degradation in the mammalian brain. Am J Physiol Cell Physiol. 319:C624–C629. 2020. View Article : Google Scholar : PubMed/NCBI

79 

Califf RM: Biomarker definitions and their applications. Exp Biol Med (Maywood). 243:213–221. 2018. View Article : Google Scholar : PubMed/NCBI

80 

Petroušková P, Hudáková N, Maloveská M, Humeník F and Cizkova D: Non-Exosomal and exosome-derived miRNAs as promising biomarkers in canine mammary cancer. Life (Basel). 12:5242022.PubMed/NCBI

81 

Li H and Tie XJ: Exploring research progress in studying serum exosomal miRNA-21 as a molecular diagnostic marker for breast cancer. Clin Transl Oncol. 11:10.1007/s12094–024-03454-z. 2024.

82 

Liu M, Mo F, Song X, He Y, Yuan Y, Yan J, Yang Y, Huang J and Zhang S: Exosomal hsa-miR-21-5p is a biomarker for breast cancer diagnosis. PeerJ. 9:e121472021. View Article : Google Scholar : PubMed/NCBI

83 

Li S, Zhang M, Xu F, Wang Y and Leng D: Detection significance of miR-3662, miR-146a, and miR-1290 in serum exosomes of breast cancer patients. J Cancer Res Ther. 17:749–755. 2021. View Article : Google Scholar : PubMed/NCBI

84 

Wang W and Luo YP: MicroRNAs in breast cancer: Oncogene and tumor suppressors with clinical potential. J Zhejiang Univ Sci B. 16:18–31. 2015. View Article : Google Scholar : PubMed/NCBI

85 

Wang B, Mao JH, Wang BY, Wang LX, Wen HY, Xu LJ, Fu JX and Yang H: Exosomal miR-1910-3p promotes proliferation, metastasis, and autophagy of breast cancer cells by targeting MTMR3 and activating the NF-κB signaling pathway. Cancer Lett. 489:87–99. 2020. View Article : Google Scholar : PubMed/NCBI

86 

Wei Y, Li M, Cui S, Wang D, Zhang CY, Zen K and Li L: Shikonin inhibits the proliferation of human breast cancer cells by reducing tumor-derived exosomes. Molecules. 21:7772016. View Article : Google Scholar : PubMed/NCBI

87 

Viallard C and Larrivée B: Tumor angiogenesis and vascular normalization: Alternative therapeutic targets. Angiogenesis. 20:409–426. 2017. View Article : Google Scholar : PubMed/NCBI

88 

Jung KO, Youn H, Lee CH, Kang KW and Chung JK: Visualization of exosome-mediated miR-210 transfer from hypoxic tumor cells. Oncotarget. 8:9899–9910. 2017. View Article : Google Scholar : PubMed/NCBI

89 

Baroni S, Romero-Cordoba S, Plantamura I, Dugo M, D'Ippolito E, Cataldo A, Cosentino G, Angeloni V, Rossini A, Daidone MG and Iorio MV: Exosome-mediated delivery of miR-9 induces cancer-associated fibroblast-like properties in human breast fibroblasts. Cell Death Dis. 7:e23122016. View Article : Google Scholar : PubMed/NCBI

90 

Kong W, He L, Richards EJ, Challa S, Xu CX, Permuth-Wey J, Lancaster JM, Coppola D, Sellers TA, Djeu JY and Cheng JQ: Upregulation of miRNA-155 promotes tumour angiogenesis by targeting VHL and is associated with poor prognosis and triple-negative breast cancer. Oncogene. 33:679–689. 2014. View Article : Google Scholar : PubMed/NCBI

91 

Kontomanolis E, Mitrakas A, Giatromanolaki A, Kareli D, Panteliadou M, Pouliliou S and Koukourakis MI: A pilot study on plasma levels of micro-RNAs involved in angiogenesis and vascular maturation in patients with breast cancer. Med Oncol. 34:202017. View Article : Google Scholar : PubMed/NCBI

92 

Luengo-Gil G, Gonzalez-Billalabeitia E, Perez-Henarejos SA, Manzano EN, Chaves-Benito A, Garcia-Martinez E, Garcia-Garre E, Vicente V and Ayala de la Peña F: Angiogenic role of miR-20a in breast cancer. PLoS One. 13:e01946382018. View Article : Google Scholar : PubMed/NCBI

93 

Lee JK, Park SR, Jung BK, Jeon YK, Lee YS, Kim MK, Kim YG, Jang JY and Kim CW: Exosomes derived from mesenchymal stem cells suppress angiogenesis by down-regulating VEGF expression in breast cancer cells. PLoS One. 8:e842562013. View Article : Google Scholar : PubMed/NCBI

94 

Donnarumma E, Fiore D, Nappa M, Roscigno G, Adamo A, Iaboni M, Russo V, Affinito A, Puoti I, Quintavalle C, et al: Cancer-associated fibroblasts release exosomal microRNAs that dictate an aggressive phenotype in breast cancer. Oncotarget. 8:19592–19608. 2017. View Article : Google Scholar : PubMed/NCBI

95 

Yan Z, Sheng Z, Zheng Y, Feng R, Xiao Q, Shi L, Li H, Yin C, Luo H, Hao C, et al: Cancer-associated fibroblast-derived exosomal miR-18b promotes breast cancer invasion and metastasis by regulating TCEAL7. Cell Death Dis. 12:11202021. View Article : Google Scholar : PubMed/NCBI

96 

Wang H, Wei H, Wang J, Li L, Chen A and Li Z: MicroRNA-181d-5p-containing exosomes derived from CAFs promote EMT by regulating CDX2/HOXA5 in breast cancer. Mol Ther Nucleic Acids. 19:654–667. 2020. View Article : Google Scholar : PubMed/NCBI

97 

Martello G, Rosato A, Ferrari F, Manfrin A, Cordenonsi M, Dupont S, Enzo E, Guzzardo V, Rondina M, Spruce T, et al: A MicroRNA targeting dicer for metastasis control. Cell. 141:1195–1207. 2010. View Article : Google Scholar : PubMed/NCBI

98 

Weng YS, Tseng HY, Chen YA, Shen PC, Al Haq AT, Chen LM, Tung YC and Hsu HL: MCT-1/miR-34a/IL-6/IL-6R signaling axis promotes EMT progression, cancer stemness and M2 macrophage polarization in triple-negative breast cancer. Mol Cancer. 18:422019. View Article : Google Scholar : PubMed/NCBI

99 

Zhang Y, Lai X, Yue Q, Cao F, Zhang Y, Sun Y, Tian J, Lu Y, He L, Bai J and Wei Y: Bone marrow mesenchymal stem cells-derived exosomal microRNA-16-5p restrains epithelial-mesenchymal transition in breast cancer cells via EPHA1/NF-κB signaling axis. Genomics. 114:1103412022. View Article : Google Scholar : PubMed/NCBI

100 

Liang Z, Liu L, Gao R, Che C and Yang G: Downregulation of exosomal miR-7-5p promotes breast cancer migration and invasion by targeting RYK and participating in the atypical WNT signalling pathway. Cell Mol Biol Lett. 27:882022. View Article : Google Scholar : PubMed/NCBI

101 

Wang X, Luo G, Zhang K, Cao J, Huang C, Jiang T, Liu B, Su L and Qiu Z: Correction: Hypoxic tumor-derived exosomal miR-301a mediates M2 macrophage polarization via PTEN/PI3Kγ to promote pancreatic cancer metastasis. Cancer Res. 80:9222020. View Article : Google Scholar : PubMed/NCBI

102 

Chen WX, Wang DD, Zhu B, Zhu YZ, Zheng L, Feng ZQ and Qin XH: Exosomal miR-222 from adriamycin-resistant MCF-7 breast cancer cells promote macrophages M2 polarization via PTEN/Akt to induce tumor progression. Aging (Albany NY). 13:10415–10430. 2021. View Article : Google Scholar : PubMed/NCBI

103 

Gordon S and Martinez FO: Alternative activation of macrophages: Mechanism and functions. Immunity. 32:593–604. 2010. View Article : Google Scholar : PubMed/NCBI

104 

Pakravan K, Mossahebi-Mohammadi M, Ghazimoradi MH, Cho WC, Sadeghizadeh M and Babashah S: Monocytes educated by cancer-associated fibroblasts secrete exosomal miR-181a to activate AKT signaling in breast cancer cells. J Transl Med. 20:5592022. View Article : Google Scholar : PubMed/NCBI

105 

Hao C, Sheng Z, Wang W, Feng R, Zheng Y, Xiao Q and Zhang B: Tumor-derived exosomal miR-148b-3p mediates M2 macrophage polarization via TSC2/mTORC1 to promote breast cancer migration and invasion. Thorac Cancer. 14:1477–1491. 2023. View Article : Google Scholar : PubMed/NCBI

106 

Yao X, Tu Y, Xu Y, Guo Y, Yao F and Zhang X: Endoplasmic reticulum stress-induced exosomal miR-27a-3p promotes immune escape in breast cancer via regulating PD-L1 expression in macrophages. J Cell Mol Med. 24:9560–9573. 2020. View Article : Google Scholar : PubMed/NCBI

107 

Jiang M, Zhang W, Zhang R, Liu P, Ye Y, Yu W, Guo X and Yu J: Cancer exosome-derived miR-9 and miR-181a promote the development of early-stage MDSCs via interfering with SOCS3 and PIAS3 respectively in breast cancer. Oncogene. 39:4681–4694. 2020. View Article : Google Scholar : PubMed/NCBI

108 

Salehi M, Vafadar A, Khatami SH, Taheri-Anganeh M, Vakili O, Savardashtaki A, Negahdari B, Naeli P, Behrouj H, Ghasemi H and Movahedpour A: Gastrointestinal cancer drug resistance: the role of exosomal miRNAs. Mol Biol Rep. 49:2421–2432. 2022. View Article : Google Scholar : PubMed/NCBI

109 

Hu W, Tan C, He Y, Zhang G, Xu Y and Tang J: Functional miRNAs in breast cancer drug resistance. Onco Targets Ther. 11:1529–1541. 2018. View Article : Google Scholar : PubMed/NCBI

110 

Sachdeva M, Wu H, Ru P, Hwang L, Trieu V and Mo YY: MicroRNA-101-mediated Akt activation and estrogen-independent growth. Oncogene. 30:822–831. 2011. View Article : Google Scholar : PubMed/NCBI

111 

Miller TE, Ghoshal K, Ramaswamy B, Roy S, Datta J, Shapiro CL, Jacob S and Majumder S: MicroRNA-221/222 confers tamoxifen resistance in breast cancer by targeting p27Kip1. J Biol Chem. 283:29897–29903. 2008. View Article : Google Scholar : PubMed/NCBI

112 

Wei Y, Lai X, Yu S, Chen S, Ma Y, Zhang Y, Li H, Zhu X, Yao L and Zhang J: Exosomal miR-221/222 enhances tamoxifen resistance in recipient ER-positive breast cancer cells. Breast Cancer Res Treat. 147:423–431. 2014. View Article : Google Scholar : PubMed/NCBI

113 

Gao M, Miao L, Liu M, Li C, Yu C, Yan H, Yin Y, Wang Y, Qi X and Ren J: miR-145 sensitizes breast cancer to doxorubicin by targeting multidrug resistance-associated protein-1. Oncotarget. 7:59714–59726. 2016. View Article : Google Scholar : PubMed/NCBI

114 

Sueta A, Yamamoto Y, Tomiguchi M, Takeshita T, Yamamoto-Ibusuki M and Iwase H: Differential expression of exosomal miRNAs between breast cancer patients with and without recurrence. Oncotarget. 8:69934–69944. 2017. View Article : Google Scholar : PubMed/NCBI

115 

Zhong Q, Nie Q, Wu R and Huang Y: Exosomal miR-18a-5p promotes EMT and metastasis of NPC cells via targeting BTG3 and activating the Wnt/β-catenin signaling pathway. Cell Cycle. 22:1544–1562. 2023. View Article : Google Scholar : PubMed/NCBI

116 

Xia Y, Wei K, Hu LQ, Zhou CE, Lu ZB, Zhan GS, Pan XL, Pan CF, Wang J, Wen W, et al: Exosome-mediated transfer of miR-1260b promotes cell invasion through Wnt/β-catenin signaling pathway in lung adenocarcinoma. J Cell Physiol. 235:6843–6853. 2020. View Article : Google Scholar : PubMed/NCBI

117 

Huang Z, Zhen S, Jin L, Chen J, Han Y, Lei W and Zhang F: miRNA-1260b promotes breast cancer cell migration and invasion by downregulating CCDC134. Curr Gene Ther. 23:60–71. 2023. View Article : Google Scholar : PubMed/NCBI

118 

Xiao Z, Feng X, Zhou Y, Li P, Luo J, Zhang W, Zhou J, Zhao J, Wang D, Wang Y, et al: Exosomal miR-10527-5p inhibits migration, invasion, lymphangiogenesis and lymphatic metastasis by affecting Wnt/β-catenin signaling via Rab10 in esophageal squamous cell carcinoma. Int J Nanomedicine. 18:95–114. 2023. View Article : Google Scholar : PubMed/NCBI

119 

Liu Y, Yang C, Chen S, Liu W, Liang J, He S and Hui J: Cancer-derived exosomal miR-375 targets DIP2C and promotes osteoblastic metastasis and prostate cancer progression by regulating the Wnt signaling pathway. Cancer Gene Ther. 30:437–449. 2023.PubMed/NCBI

120 

Li H, Xie C, Lu Y, Chang K, Guan F and Li X: Exosomal mir92a promotes cytarabine resistance in myelodysplastic syndromes by activating Wnt/β-catenin signal pathway. Biomolecules. 12:14482022. View Article : Google Scholar : PubMed/NCBI

121 

Yue X, Lan F and Xia T: Hypoxic glioma cell-secreted exosomal miR-301a activates Wnt/β-catenin signaling and promotes radiation resistance by targeting TCEAL7. Mol Ther. 27:1939–1949. 2019. View Article : Google Scholar : PubMed/NCBI

122 

Yue X, Cao D, Lan F, Pan Q, Xia T and Yu H: MiR-301a is activated by the Wnt/β-catenin pathway and promotes glioma cell invasion by suppressing SEPT7. Neuro Oncol. 18:1288–1296. 2016. View Article : Google Scholar : PubMed/NCBI

123 

Wang L, He M, Fu L and 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. 257:1180242020. View Article : Google Scholar : PubMed/NCBI

124 

Fang F, Guo C, Zheng W, Wang Q and Zhou L: Exosome-mediated transfer of miR-1323 from cancer-associated fibroblasts confers radioresistance of c33a cells by targeting PABPN1 and activating Wnt/β-catenin signaling pathway in cervical cancer. Reprod Sci. 29:1809–1821. 2022. View Article : Google Scholar : PubMed/NCBI

125 

Shan G, Zhou X, Gu J, Zhou D, Cheng W, Wu H, Wang Y, Tang T and Wang X: Downregulated exosomal microRNA-148b-3p in cancer associated fibroblasts enhance chemosensitivity of bladder cancer cells by downregulating the Wnt/β-catenin pathway and upregulating PTEN. Cell Oncol (Dordr). 44:45–59. 2021. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2024
Volume 52 Issue 2

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li H, Li X and Du W: Interplay between Wnt signaling molecules and exosomal miRNAs in breast cancer (Review). Oncol Rep 52: 107, 2024
APA
Li, H., Li, X., & Du, W. (2024). Interplay between Wnt signaling molecules and exosomal miRNAs in breast cancer (Review). Oncology Reports, 52, 107. https://doi.org/10.3892/or.2024.8766
MLA
Li, H., Li, X., Du, W."Interplay between Wnt signaling molecules and exosomal miRNAs in breast cancer (Review)". Oncology Reports 52.2 (2024): 107.
Chicago
Li, H., Li, X., Du, W."Interplay between Wnt signaling molecules and exosomal miRNAs in breast cancer (Review)". Oncology Reports 52, no. 2 (2024): 107. https://doi.org/10.3892/or.2024.8766