Next Article in Journal
Hypouricemia and Urate Transporters
Next Article in Special Issue
Impact of miR-1/miR-133 Clustered miRNAs: PFN2 Facilitates Malignant Phenotypes in Head and Neck Squamous Cell Carcinoma
Previous Article in Journal
The Antiplatelet Action of S-Nitroso Human Serum Albumin in Whole Blood
Previous Article in Special Issue
Competitive Endogenous RNA Network Involving miRNA and lncRNA in Non-Hodgkin Lymphoma: Current Advances and Clinical Perspectives
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Breast Cancer Subtype-Specific miRNAs: Networks, Impacts, and the Potential for Intervention

1
Department of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada
2
Department of Microbiology & Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada
3
Nova Scotia Health Authority, Halifax, NS B3H 2YN, Canada
*
Author to whom correspondence should be addressed.
Biomedicines 2022, 10(3), 651; https://doi.org/10.3390/biomedicines10030651
Submission received: 10 February 2022 / Revised: 7 March 2022 / Accepted: 9 March 2022 / Published: 11 March 2022
(This article belongs to the Special Issue MicroRNA in Solid Tumor and Hematological Diseases 2.0)

Abstract

:
The regulatory and functional roles of non-coding RNAs are increasingly demonstrated as critical in cancer. Among non-coding RNAs, microRNAs (miRNAs) are the most well-studied with direct regulation of biological signals through post-transcriptional repression of mRNAs. Like the transcriptome, which varies between tissue type and disease condition, the miRNA landscape is also similarly altered and shows disease-specific changes. The importance of individual tumor-promoting or suppressing miRNAs is well documented in breast cancer; however, the implications of miRNA networks is less defined. Some evidence suggests that breast cancer subtype-specific cellular effects are influenced by distinct miRNAs and a comprehensive network of subtype-specific miRNAs and mRNAs would allow us to better understand breast cancer signaling. In this review, we discuss the altered miRNA landscape in the context of breast cancer and propose that breast cancer subtypes have distinct miRNA dysregulation. Further, given that miRNAs can be used as diagnostic and/or prognostic biomarkers, their impact as novel targets for subtype-specific therapy is also possible and suggest important implications for subtype-specific miRNAs.

1. Introduction

Three decades ago, the first microRNA (miRNA), lin-4, was identified in Caenorhabditis elegans [1]. The seminal paper was the first to suggest the regulation of a protein (LIN-14) through complementary sequence interaction of the miRNA with the 3′untranslated region (UTR) of the lin-14 mRNA. Subsequent research has demonstrated the universal importance of miRNAs, which is reflected by their ubiquitous presence and abundance across all genera from viruses to mammals. Calin et al., first described a potential miRNA role in cancer; miR-15 and miR-16 were deleted in chromosome 13 in chronic lymphocytic leukaemia [2]. In the past two decades, miRNAs found to play critical roles in the regulation of cancer pathways have been labelled oncomirs and tumor suppressor mirs based on their prominent oncogenic and tumor suppressive effect [3]. Notable examples include miR-21, miR-17~92 and the miR-200 family [4].
Over 2000 miRNAs have been identified in humans [5]. In the genome, miRNA genes are intragenic or intergenic, and some miRNA genes are clustered into a polycistronic transcription unit which are expressed together in multiple conditions [6]. Micro RNAs have an established naming convention where they are named by sequential numbers, and if the same miRNAs are generated from distinct genetic locations, they are identified by hyphenated numbers (e.g., miR-10-1), whereas closely related mature miRNAs are indicated by a letter suffix (e.g., miR-200 family members; miR-200a, miR200b, and miR200c). Mature miRNAs are labelled with the 5p or 3p suffix to denote their origin arm of the stem loop [7].
Post-transcription, mature functional miRNAs are generated via a multi-step process. The primary pri-miRNA transcripts consist of single or multiple loop sections and single-stranded fragments, which are cleaved by the microprocessor complex involving RNaseIII Drosha and its cofactor subunit DiGeorge syndrome critical region 8 (DGCR8) in the nucleus, resulting in pre-miRNAs [6]. The Drosha and DGCR8 microprocessor complex binds to the stem loop section and cleaves at 11 bp from base of stem loop [8,9]. Post-export to the cytoplasm through the exportin5, the loop part is cleaved by Dicer to form duplex RNA, each strand ~20 nucleotide long. Dicer recognizes the single nucleotide overhangs of the stem loop pri-miRNA, and the Dicer–RNA complex is stabilized by the trans-activation-responsive RNA binding protein (TARBP2) [3,10]. This duplex RNA is bound by Argonaute protein AGO to form RNA induced silencing complex (RISC) and generate the mature miRNAs [11]. One of the mature miRNA strands (passenger strand) is degraded by the nuclease activity of AGO2, while the other strand (guide strand) is incorporated into RISC, and subsequently regulates post-transcriptional gene expression via binding to complementary sequences in mRNA targets. This typically results in the downregulation of target mRNAs by inhibiting their translation and/or mediating their degradation via RISC [4,5,7,8,9]. In specific conditions of serum or amino acid starvation and in quiescent cells, miRNA mediated upregulation of translation is also observed [12,13].
The process of post-transcriptional gene regulation by miRNAs is essential in the regulation of all biological processes including development and differentiation, immune responses, cell cycle progression, cell death, stress responses, and metabolism [14,15,16,17,18,19,20]. The essentiality of miRNAs is also reflected in cancer, where dysregulation of miRNAs is commonly paired with cancer development, progression, and treatment responses [21]. For breast cancer, dysregulation of miRNA happens early in the development of ductal carcinoma [22]. In the seminal 2005 paper, Iorio et al. described distinct miRNA signatures between normal and breast cancer cells with significant deregulation of miR-125b, miR-145, miR-21 and miR-155, which are mostly known for their tumor suppressive function [23]. Further, in breast cancer specifically, the importance of miRNAs is evident not only in the progression of the disease, but also in the specific breast subtypes which define the disease clinically and molecularly and is the focus of this review.

2. Breast Cancer Subtypes Defined by Gene and miRNA Expression

Breast cancer is one of the most common malignancies, with multiple subtypes, based on clinical parameters and molecular profiling. In addition to disease staging, the expression status of hormone receptors’ estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) in tumors define the prognosis of the cancer and treatment options [24,25,26]. Hence, clinically, breast cancers are defined as ER+/PR+, HER+, or triple-negative (i.e., those lacking expression of these three receptors). This classification system allows for the administration of endocrine therapies in the hormone expressing subtypes. Additionally, breast cancer subtyping is observed via transcriptome profiling, which has identified four major subtypes (luminal A, luminal B, HER2, and basal-like). The ER+/PR+ breast cancers are predominately luminal A/B and TNBCs are predominately basal-like. Clearly gene expression defines breast cancer [27]; it is then not surprising that the expression of miRNAs also displays subtype-specificity.
With the increasing availability of microarray and small RNA sequencing technologies, significant advancement has been possible in identifying global changes in miRNA dysregulation. Meta-analysis of patient sample repositories has also proven a versatile tool for the identification and study of miRNAs involved in breast cancer. Clustering analysis of all patient tumors based on miRNA expression show separation based on breast cancer subtype, demonstrating that miRNAs affect subtype phenotypes [22,28,29].
Most of the studies included in this manuscript are from patient cohorts that identify important miRNAs among specific patients; these studies compare cancer tissue with normal tissue to identify the cancer specific miRNAs. By comparing the enriched miRNAs among different breast cancer subtypes, the subtype specific miRNA signature and common breast cancer miRNAs are identified. Direct relative expression of miRNAs among different subtypes are studied in in vitro experiments, where the controlled environment allows for the comparison between two cancer subtypes instead of comparing against a standard control.
This variation in the abundance of specific miRNAs associated with specific subtypes and the potential influence this has on subtype-associated gene expression, signaling and clinical outcome is discussed here for each major subtype and summarized in Table 1.

2.1. ER+/PR+ Breast Cancers

Estrogen and progesterone hormones influence normal breast development and in breast cancer they play important roles in the disease progression, which is governed by the presence of estrogen and progesterone receptors. The luminal subtypes consist predominately of ER+/PR+ breast cancers and comprise the majority of breast cancers [25]. ER-associated miRNAs are enriched in the luminal subtypes. A key distinguisher between the luminal A and B subtypes is the expression of the Ki67 proliferation marker; luminal A breast cancers have low Ki67, while luminal B breast cancers have high Ki67 [46]. The subtype-specific expression of Ki67 reflects the better prognosis and low tumor grade associated with the luminal A subtype, and the comparably worse prognosis and higher tumor grade associated with the luminal B subtype. Furthermore, in terms of miRNA dysregulations, we see distinguishing features in the two luminal subtypes. Clustering analysis including breast cancers of all subtypes showed that luminal A and luminal B are clustered close to each other but were distinct from one another [22]. Further, there is more prominent dysregulation in miRNAs in luminal B vs. luminal A breast cancers. Specifically, 657 miRNAs were found dysregulated in luminal B cancers and 67 miRNAs were dysregulated in luminal A cancers. Among the luminal-dysregulation miRNAs, miR-1290 is significantly reduced in luminal A Ki67 low tumors [35]. MiR-1290 is oncogenic in a context driven nature [47]; its reduction in luminal A correlates with its clinical outcome.
The distinct luminal A-miRNA signature is defined by miR-30c-5p, miR-30b-5p, and miR-99a/let-7c/miR-125b cluster, while the luminal B subtype is enriched with miR-182-5p, miR-200b-3p, miR-15b-3p, miR-149-5p, miR-193b-3p and miR-342-3p, 5p [22,30,32,33,34]. Haakensen et al. linked miR-30b-5p, miR-30c-5p, miR182-5p and miR-200b-3p to better prognosis in patients with luminal subtype breast cancer [22]. Given the prominence of miR-30 miRNAs among luminal A breast cancers, it important to note that miR-30 is a cluster of six miRNAs (miR-30a, -30b, 30-c1, -30c-2, -30d, 30e), and as whole the miR-30 family generally inhibit migration and growth [48] (Figure 1). For example, miR-29b/miR-30d regulate migration through lysyl oxidase-like 4 (LOXL4) inhibition [49]. Early evidence demonstrated that miR-30 inhibit cell division through cyclin D2 targeting [50], and inhibition of c-Myc-induced carcinogenesis [51]. The other prominent luminal A-associated miRNA cluster, miR-99a (miR-100)/let-7c/miR-125b), is also typically paired with tumor suppression and reducing growth rate and migration in breast cancer (Figure 1). This cluster reduces tumor growth by inhibiting proteins involved in important cellular processes, including homeobox A1 (HOXA1), mammalian target of rapamycin (mTOR), insulin-like growth factor binding protein 1 (IGFBP1), and fibroblast growth factor receptor 3 (FGFR3) [52,53,54,55]. The miR-99a cluster also inhibits oncogenes, including Yamaguchi sarcoma viral oncogene homolog 1 (YES1), ETS proto-oncogene 1 (ETS1) and ETS variant transcription factor 6 (ETV6) [23].
The increased expression of miRNAs associated with tumor suppression among luminal A breast cancers is consistent with the relatively slow growth of luminal A breast cancers; however, there is also evidence that in advanced cancer stages, miR-30 plays an oncogenic role. In patients with advanced tumors, miR-30b-5p is highly expressed in tissue and circulation [38], suggesting a potential change in function towards tumor progression. This is an important caveat when considering miRNA function in cancers; it is always context dependent and reliant on the abundance of relative target mRNAs, which is also shifting during cancer progression. The function of miRNAs is never in isolation and dependent upon the evolving transcriptome.
Additionally, differences in circulating levels of miR-29a, miR-181a, and miR-652 is also evident in the serum samples of luminal breast cancers. All three miRNAs are significantly downregulated in both the tumors and serum of patients with luminal A vs. luminal B breast cancers [33]. Treatment regimens also influence circulating miRNA levels, with tamoxifen treatment of ER+ tumors resulting in increased miR-221 in serum [31].

2.2. HER2 Overexpressing Breast Cancers

HER2 is a tyrosine kinase receptor that belongs to the family of epidermal growth factor receptors (EGFR). HER2 overexpression in breast cancers is present about 15–20% of breast tumors, and like TNBC, it is associated with worse patient prognosis and survival [56]. However, unlike TNBCs, HER2+ (i.e., HER2 overexpression) in breast cancers can be targeted. Paired with chemotherapy, the monoclonal antibody against HER2, trastuzumab, is used to treat HER2+ breast cancers. However, resistance and recurrence are common, so other drugs have been developed such as the tyrosine kinase inhibitor lapatinib and the monoclonal antibody pertuzumab, which prevents HER2 dimerization and signaling [57]. HER2 overexpression results in specific gene signatures in breast cancer, especially in the cancer-promoting pathways. The increased mitogen-activated protein kinase (MAPK), phosphatidyl-inositol-3 kinase (PI3K)/AKT and HER3 receptor signaling [58,59], characteristic of HER2+ breast cancers, result in the downstream effects on enhanced cell proliferation and the observed aggressive clinical phenotype.
HER2+ breast cancers also have a distinct miRNA expression profile. Lowery et al. profiled 453 miRNAs in 29 tumors and found that that the HER2+ subtype is associated with a number of miRNAs, including miR-302c, miR-520d, miR-181c, miR-376b and miR-30e [37]. Additionally, in a study that evaluated 221 breast cancer tumors and 49 normal tissue controls, miR-125b is reportedly upregulated in HER2+ breast cancers [36]. MiR-4728-3p expression is also associated with HER2+ breast cancers and its gene is encoded within a HER2 intron [60]. Other miRNAs identified in HER2+ breast cancers include upregulation of miR-21 and miR-146a-5p, while miR-181d and miR-195-5p are downregulated [61]. These altered miRNA expression profiles are specific to the HER2+ subtype and could be exploited in diagnostic/prognostic tools.
In terms of a connection with cancer phenotypes associated with the HER2+ breast cancer subtype, these miRNAs also have function beyond just expression associations. For example, miR-125b is specifically connected to metastasis of HER2+ breast cancers [62] and with worse patient outcomes [39]. However, the effects of the miRNA in cancer are again context dependent, as miR-125 has also been reported to have tumor suppressive effects in various cancers [63]. For HER2+ associated miR-4728-3p, its mRNA targets include downstream targets of HER2 signal transduction and the estrogen receptor alpha (ESR1) [64]. Recent investigation into miR-4728 indicate that when it is overexpressed in HER2-postive tumors, the efficacy of HER2 inhibitor laptinib is minimized (Figure 1) [65]. This was linked to decreased expression of pro-apoptotic NOXA59. Further, miR-4728-3p mediates stabilization of miR-21-5p in HER2+ breast cancer (Figure 1) [66], facilitating miR-21-5p mediated oncogenesis [67,68].
As demonstrated with miR-4728 [65], the effect of miRNA on drug treatments are not uncommon, likely because miRNAs are expressed in a context dependent manner. Identifying changes to the miRNA landscape before and after treatment will aid in the development of improved treatments for HER2+ breast cancers. Normann et al. utilized four HER2+ breast cancer cell lines to assess whether treatment with trastuzumab and lapatinib together or separately change the miRNA landscape [57]. Levels of miRNAs hsa-let-7b, miR-1236, miR-134, miR-25, miR-3656, miR-3663-3p, miR-3940 and miR-885-5p were significantly altered following drug treatment [57]. Importantly, treatment with miRNA mimics (e.g., miR-101-5p mimic) sensitized cells to treatment with the drugs. Additionally, miR-101-5p downregulates HER2 and MAPK1; its expression is associated with higher survival rates in HER2+ tumors. These findings together indicate that miR-101-5p acts as a tumor suppressor in HER2+ cancers.

2.3. TNBC

TNBC is one of the major contributors of breast cancer mortality; nearly 25% of the overall breast cancer-related deaths are among patients with TNBC, despite representing only 10–15% of breast cancers [40]. TNBCs are aggressive in terms of proliferation, with a high mitotic index and high Ki67 staining in histology. They also recur more frequently than other breast cancer subtypes. Like the other above reviewed subtypes, TNBC also has reported correlations with specific miRNAs. For example, survival analysis of patients reveal correlation with multiple miRNAs in TNBC. Specifically, miR-27a/b, miR-210 and miR-30e are associated with worse survival, and miR-155 and miR-493 are associated with better survival in TNBC [41,42,43]. Further, miR-374a/b and miR-454 are associated with disease free survival [64]. Some miRNAs are reported to be associated with metabolic processes in TNBC; miR-210, miR-105-5p and miR-767-5p are essential for the Warburg effect, with miR-210 involved in glucose uptake, lactate production and extracellular acidification rate in TNBC [43].
TNBCs exhibit significant heterogeneity, both phenotypically and genotypically [44,69]. Hence, TNBCs can be further subdivided into multiple subtypes. Based on gene expression profiling, TNBC is divided into six major subtypes; basal-like 1 (BL1), basal-like 2 (BL2) immunomodulatory (IM), claudin-low mesenchymal (M), mesenchymal-stem like (MSL) and luminal androgen receptor positive (LAR), and each of these have unique clinical outcomes, phenotypes and drug sensitivities. For example, the two mesenchymal TNBC subtypes are associated with epithelial–mesenchymal–transition (EMT) gene signatures and pronounced migratory capacity. The individual miRNA signature for molecular subtypes has not been studied in detail for most of the TNBC molecular subtypes. However, significant information on basal-like TNBC are achieved through in silico analysis of patient cohort studies with miRNA studies. The molecular influences of some of the miRNAs are studied in detail in in vitro experiments detailing its function in TNBC.
For example, Rinaldis et al. identified a distinct four miRNA signature in TNBC and demonstrated that the miR-17~92 and miR-106b-25 clusters are significantly overexpressed in basal-like TNBCs [70]. It is noteworthy that the miR-17~92 cluster is of particular importance in the TNBC transcriptome and are among the most well-studied miRNAs (Figure 1). The miR-17~92 cluster comprises of miR-17, miR-18a, miR-19a, miR-20a, miR-19b-1 and miR-92a-1; together these miRNAs regulate expression of transcription factor E2F1, thrombospondin1 (THBS1), connective tissue growth factor (CTGF) and phosphate and tensin homolog (PTEN) [71,72,73]. Earlier, O’Donnell et al. showed that the proto-oncogene cMYC modulates the critical transcription factor E2F1 by regulating the miR-17~92 cluster, resulting in cancer proliferation [74]. This intricate oncogene signaling regulation has earned the miR-17~92 cluster the moniker oncomiR-1. Apart from involvement in cMYC oncogene signaling, the oncomiR-1 cluster also inhibits inositol polyphosphate-4-phosphatase type II B (INPP4B) and is distinctly associated with the BL1TNBC subtype [75]. Notably, INPP4B is a known inhibitor of the PI3K/AKT mediated growth pathway; knockdown experiments clearly showed the tumor suppressive role of INPP4B [76]. Further, Kalecky et al. found distinct differences between BL1 and BL2; miR17~92 cluster, miR-17, miR-18a and miR-19a were high in BL1 tumors, but not as high in BL2 tumors [75].
The BL1 subtype of TNBC is associated with low PTEN and overall decreased miRNA expression [77]. This group is also associated with worst survival among TNBCs. Recent evidence on structural mapping of miR-17~92 cluster showed a sub-optimal micro processing of primary miRNAs, resulting in unequal expression of the constituent miRNAs of the cluster [78]. This highlights the potential role of regulatory molecules of miRNA processing in the regulation of the miR-17~92 cluster [79,80]. This cluster may play a crucial role in the subtype characteristics associated with BL1-TNBCs and the distinction of TNBCs from ER+/PR+ and HER2+ breast cancers.
Like the miR-17~92 cluster, miR-135b shows molecular subtype specificity among TNBCs; miR-135b is upregulated in TNBCs in general but is most overexpressed in BL1 and BL2 TNBC subtypes [81]. Importantly, miR-135b regulates expression of the ER, androgen receptor (AR) and hypoxia inducible factor 1 alpha subunit inhibitor (HIF1AN) [82]; thereby, its expression in TNBCs may directly affect hormone receptor loss and contribute to this distinguishing feature of TNBCs. Interestingly, the AR significantly reduces miR-135b expression [81]. Further, miR-135-b also targets large tumor suppressor kinase 2 (LATS2), a intermediator of Hippo signaling, and promotes cell proliferation in unstratified breast cancer patient tumors [83]. In TNBC, miR-135-b targets adenomatosus polyposis coli (APC), a WNT signaling regulator [84]. Both these targets are important regulators of migratory process. These studies observed increased proliferation and migration in the cells treated with miR-135b mimics. However, in early-stage breast cancer, miR-135b-5p repression resulted in migration through the syndecan-binding protein (SDCBP) [85]. These findings strongly suggest that miR-135-5p has a role in the regulation of migration processes in breast cancer, with functional difference among different subtypes arising from context-specific signaling networks.
Currently, most of the TNBC molecular subtype data with respect to miRNA associations is predominately with the BL1 and BL2 subtypes. Given the importance of miRNA in gene expression regulation, it is likely that there are specific miRNAs associated with the other molecular subtypes; however, they just remain unstudied. The MSL and M molecular subtypes exhibit migratory phenotypes with pronounced expression of EMT genes. These genes are repressed in the basal-like cells; hence, the likelihood of a role for miRNA-mediated regulation is high in distinguishing these phenotypes.
Although the associations with specific molecular subtypes was not addressed in a study that compared miRNA expression among different breast cancer cell lines, Adams et al. reported downregulation of miR-34a and miR-200 in TNBCs compared to ER+ breast cancer and non-tumorigenic breast cell lines (Figure 1) [86,87]. These specific miRNAs are connected with EMT regulation in breast and other tumors, with effects in migration [88,89,90,91]. Inhibition of miR-34a promotes the migratory phenotype in MDA-MB-231 cells through the IL6 pathway [88]. Notably, MDA-MB-231 is classified as a mesenchymal TNBC cell line, and miR-34a is downregulated in the cell line [88].
The miR-200 family consists of miR-141, miR-200a, 200b, 200c and miR-429 [92]. It is one of the most well-studied miRNA families with extensive effects in the regulation of migration, via its multiple members and targets. Notably, inhibitors or antagomirs of miR-200a inhibited critical EMT genes N-cadherin, Snail and Twist [93], and other genes involved in migration, like proto-oncogene Jun, ETS1, brain-specific homeobox/POU domain protein 3A (BRN3) and zinc finger E-box binding homeobox 1 (ZEB1), were regulated by all miR-200 family members [93]. The miR-200 family associations with some TNBC cell lines may be DNA methylation dependent. Interestingly, TNBC cell lines with mesenchymal phenotypes, like MDA-MB-231 cells and lung metastasis sub-clones of MDA-MB-468 cells (MDA-MB-468 LN), have hypermethylated promoters of the miR-200 family [94,95]. Demethylation of the miR-200c promoter site induced its expression and reduced cell migration [96]. ZEB1 facilitates this epigenetic regulation of the miR-200 family [97].

2.4. Therapeutics of miRNA in Breast Cancer

The importance of miRNAs in breast cancer suggests they have therapeutic potential, and this could be achieved by either depleting oncogenic miRNAs or enriching tumor suppressive miRNAs [98,99]. Elimination of oncogenic miRNAs is possible by delivering an oligomer complementary to the target miRNA. These antagomirs bind to mature miRNAs, resulting in inhibition and degradation of the target miRNAs [98]. Enriching tumor suppressive miRNAs is done by delivering miRNA mimics (double stranded RNA sequences that have the same sequence as the miRNA) in cells; importantly, with mimics they are considered native to the cell [100]. By comparing published work on miRNA-based therapeutics, Krützfeldt et al. shortlisted twelve miRNAs being studied with therapeutic interest in breast cancers [98]. Three miRNAs in particular, miR-21, miR-10-b and miR-34a, have been extensively studied with profound preclinical therapeutic potential, with both antimetastatic and anti-proliferative properties. Reintroduction of miR-34a in TNBCs reduces both migration and proliferation in cell culture [86] and in vivo [101]. Combination therapy of doxorubicin and miR-34a delivered using nanoparticles was effective in TNBCs models [102]. Among antagomir studies on breast cancer, miR-21 and miR-10 antagomirs has shown promise in breast cancer [103,104,105]. It is important to note that both miR-21 and miR-34a appear to have critical influence on cancer signaling in specific breast cancer subtypes; hence, it is prudent to test the therapeutic or diagnostic/prognostic role of miRNAs in cell lines that have been clearly subtyped. Cell line classifications based on breast cancer subtypes have been summarized and should be carefully considered [106,107].

3. Conclusions

In this review, we have highlighted both the seminal papers in the field and the recent findings demonstrating miRNAs associations among certain breast cancer subtypes. We have focused our discussion on the miRNA examples that have functional consequences in the breast cancer subtypes. We have delineated important miRNA-initiated regulation of gene expression that leads to cancer phenotypes that are prominent within specific breast cancer subtypes. Importantly, the transcriptome network can be “enriched” in certain tumor-promoting or tumor suppressive phenotypes based on the abundance of certain miRNAs and their targets. Eventually these dynamics lead to effects on clinical fates. Furthermore, it is not one miRNA in isolation but the intricate signaling network of multiple miRNAs and their many gene targets that will result in specific tumor or subtype characteristics. We also reviewed the emerging evidence for a role in miRNAs in the molecular subtypes of TNBCs; this area needs further investigation.
Clearly, miRNAs play a role in the progression of breast cancer and the eventual patient outcomes. This can be exploited or intervened in new therapeutic strategies for breast cancer. Further analysis on the role of specific miRNAs and novel agents for manipulation of tumor-specific miRNAs is required. Likely, any effective intervention may require the consideration of multiple miRNAs or miRNA networks and be specifically targeted towards patients with tumors expressing the required miRNA-target profile. This review adds value to the importance of miRNAs in breast cancer subtypes and has showcased the need for further study.

Author Contributions

R.P.A. and H.F.C. jointly drafted the review. P.M., R.P.A. and H.F.C. revised the manuscript for final submission. All authors have read and agreed to the published version of the manuscript.

Funding

R.P.A. is funded by the Cancer Research Training Program from the Beatrice Hunter Cancer Research Institute (BHCRI) and Saunders Matthey. H.F.C. is funded by a Nova Scotia Graduate Studentship. R.P.A. and H.F.C are also supported by a project grant to P.M. from the Canadian Institutes of Health Research (CHIR, PJT 162313). P.M. is a senior scientist of the BHCRI and an affiliate scientist of the Nova Scotia Health Authority.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The figure in this manuscript is created with www.BioRender.com (accessed on 9 February 2022). Marie-Claire Wasson formatted the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Lee, R.C.; Feinbaum, R.L.; Ambros, V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 1993, 75, 843–854. [Google Scholar] [CrossRef]
  2. Calin, G.A.; Dumitru, C.D.; Shimizu, M.; Bichi, R.; Zupo, S.; Noch, E.; Aldler, H.; Rattan, S.; Keating, M.; Rai, K.; et al. Frequent deletions and down-regulation of micro-RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc. Natl. Acad. Sci. USA 2002, 99, 15524–15529. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Ali Syeda, Z.; Langden, S.S.S.; Munkhzul, C.; Lee, M.; Song, S.J. Regulatory Mechanism of MicroRNA Expression in Cancer. Int. J. Mol. Sci. 2020, 21, 1723. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Cui, M.; Wang, H.; Yao, X.; Zhang, D.; Xie, Y.; Cui, R.; Zhang, X. Circulating MicroRNAs in Cancer: Potential and Challenge. Front. Genet. 2019, 10, 626. [Google Scholar] [CrossRef] [Green Version]
  5. Plotnikova, O.; Baranova, A.; Skoblov, M. Comprehensive Analysis of Human microRNA—mRNA Interactome. Front. Genet. 2019, 10, 933. [Google Scholar] [CrossRef]
  6. Ha, M.; Kim, V.N. Regulation of microRNA biogenesis. Nat. Rev. Mol. Cell Biol. 2014, 15, 509–524. [Google Scholar] [CrossRef]
  7. Ambros, V.; Bartel, B.; Bartel, D.P.; Burge, C.B.; Carrington, J.C.; Chen, X.; Dreyfuss, G.; Eddy, S.R.; Griffiths-Jones, S.; Marshall, M.; et al. A uniform system for microRNA annotation. RNA 2003, 9, 277–279. [Google Scholar] [CrossRef] [Green Version]
  8. Han, J.; Lee, Y.; Yeom, K.-H.; Nam, J.-W.; Heo, I.; Rhee, J.-K.; Sohn, S.Y.; Cho, Y.; Zhang, B.-T.; Kim, V.N. Molecular Basis for the Recognition of Primary microRNAs by the Drosha-DGCR8 Complex. Cell 2006, 125, 887–901. [Google Scholar] [CrossRef] [Green Version]
  9. Gregory, R.I.; Yan, K.-P.; Amuthan, G.; Chendrimada, T.; Doratotaj, B.; Cooch, N.; Shiekhattar, R. The Microprocessor complex mediates the genesis of microRNAs. Nature 2004, 432, 235–240. [Google Scholar] [CrossRef]
  10. Park, J.-E.; Heo, I.; Tian, Y.; Simanshu, D.K.; Chang, H.; Jee, D.; Patel, D.J.; Kim, V.N. Dicer recognizes the 5′ end of RNA for efficient and accurate processing. Nature 2011, 475, 201–205. [Google Scholar] [CrossRef]
  11. Kawamata, T.; Tomari, Y. Making RISC. Trends Biochem. Sci. 2010, 35, 368–376. [Google Scholar] [CrossRef] [PubMed]
  12. Ørom, U.A.; Nielsen, F.C.; Lund, A.H. MicroRNA-10a Binds the 5′UTR of Ribosomal Protein mRNAs and Enhances Their Translation. Mol. Cell 2008, 30, 460–471. [Google Scholar] [CrossRef] [PubMed]
  13. Bukhari, S.I.A.; Vasudevan, S. FXR1a-associated microRNP: A driver of specialized non-canonical translation in quiescent conditions. RNA Biol. 2017, 14, 137–145. [Google Scholar] [CrossRef] [PubMed]
  14. Vidigal, J.A.; Ventura, A. The biological functions of miRNAs: Lessons from in vivo studies. Trends Cell Biol. 2015, 25, 137–147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Tüfekci, K.U.; Meuwissen, R.L.J.; Genç, Ş. The Role of MicroRNAs in Biological Processes. Methods Pharmacol. Toxicol. 2013, 1107, 15–31. [Google Scholar] [CrossRef]
  16. Osada, H.; Takahashi, T. MicroRNAs in biological processes and carcinogenesis. Carcinogenesis 2007, 28, 2–12. [Google Scholar] [CrossRef] [Green Version]
  17. Ebert, M.S.; Sharp, P.A. Roles for MicroRNAs in Conferring Robustness to Biological Processes. Cell 2012, 149, 515–524. [Google Scholar] [CrossRef] [Green Version]
  18. Bueno, M.J.; Malumbres, M. MicroRNAs and the cell cycle. Biochim. Biophys. Acta BBA Mol. Basis Dis. 2011, 1812, 592–601. [Google Scholar] [CrossRef] [Green Version]
  19. Saliminejad, K.; Khorram Khorshid, H.R.; Soleymani Fard, S.; Ghaffari, S.H. An overview of microRNAs: Biology, functions, therapeutics, and analysis methods. J. Cell. Physiol. 2019, 234, 5451–5465. [Google Scholar] [CrossRef]
  20. Yi, M.; Xu, L.; Jiao, Y.; Luo, S.; Li, A.; Wu, K. The role of cancer-derived microRNAs in cancer immune escape. J. Hematol. Oncol. 2020, 13, 25. [Google Scholar] [CrossRef] [Green Version]
  21. Wang, Z. Antisense RNA and Cancer. In Cancer and Noncoding RNAs; Chakrabarti, D.J., Mitra, D.S., Eds.; Translational Epigenetics; Academic Press: Boston, MA, USA, 2018; Volume 1, Chapter 12; pp. 203–227. [Google Scholar]
  22. Haakensen, V.D.; Nygaard, V.; Greger, L.; Aure, M.R.; Fromm, B.; Bukholm, I.R.; Lüders, T.; Chin, S.-F.; Git, A.; Caldas, C.; et al. Subtype-specific micro-RNA expression signatures in breast cancer progression. Int. J. Cancer 2016, 139, 1117–1128. [Google Scholar] [CrossRef]
  23. Iorio, M.; Ferracin, M.; Liu, C.-G.; Veronese, A.; Spizzo, R.; Sabbioni, S.; Magri, E.; Pedriali, M.; Fabbri, M.; Campiglio, M.; et al. MicroRNA Gene Expression Deregulation in Human Breast Cancer. Cancer Res. 2005, 65, 7065–7070. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Viale, G. The current state of breast cancer classification. Ann. Oncol. 2012, 23 (Suppl. S10), x207–x210. [Google Scholar] [CrossRef] [PubMed]
  25. Althoubaity, F.K. Molecular classification of breast cancer: A retrospective cohort study. Ann. Med. Surg. 2019, 49, 44–48. [Google Scholar] [CrossRef] [PubMed]
  26. Perou, C.M.; Sørlie, T.; Eisen, M.B.; Van De Rijn, M.; Jeffrey, S.S.; Rees, C.A.; Pollack, J.R.; Ross, D.T.; Johnsen, H.; Akslen, L.A.; et al. Molecular portraits of human breast tumours. Nature 2000, 406, 747–752. [Google Scholar] [CrossRef]
  27. Van’T Veer, L.J.; Dai, H.; Van De Vijver, M.J.; He, Y.D.; Hart, A.A.M.; Mao, M.; Peterse, H.L.; Van Der Kooy, K.; Marton, M.J.; Witteveen, A.T.; et al. Gene expression profiling predicts clinical outcome of breast cancer. Nature 2002, 415, 530–536. [Google Scholar] [CrossRef] [Green Version]
  28. Kandettu, A.; Radhakrishnan, R.; Chakrabarty, S.; Sriharikrishnaa, S.; Kabekkodu, S.P. The emerging role of miRNA clusters in breast cancer progression. Biochim. Biophys. Acta-Rev. Cancer 2020, 1874, 188413. [Google Scholar] [CrossRef]
  29. Cantini, L.; Bertoli, G.; Cava, C.; Dubois, T.; Zinovyev, A.; Caselle, M.; Castiglioni, I.; Barillot, E.; Martignetti, L. Identification of microRNA clusters cooperatively acting on epithelial to mesenchymal transition in triple negative breast cancer. Nucleic Acids Res. 2019, 47, 2205–2215. [Google Scholar] [CrossRef] [Green Version]
  30. Amorim, M.; Lobo, J.; Fontes-Sousa, M.; Estevão-Pereira, H.; Salta, S.; Lopes, P.; Coimbra, N.; Antunes, L.; De Sousa, S.P.; Henrique, R.; et al. Predictive and Prognostic Value of Selected MicroRNAs in Luminal Breast Cancer. Front. Genet. 2019, 10, 815. [Google Scholar] [CrossRef] [Green Version]
  31. Amiruddin, A.; Massi, M.N.; Islam, A.A.; Patellongi, I.; Pratama, M.Y.; Sutandyo, N.; Natzir, R.; Hatta, M.; Latar, N.H.M.; Wahid, S. microRNA-221 and tamoxifen resistance in luminal-subtype breast cancer patients: A case-control study. Ann. Med. Surg. 2021, 73, 103092. [Google Scholar] [CrossRef]
  32. Søkilde, R.; Persson, H.; Ehinger, A.; Pirona, A.C.; Fernö, M.; Hegardt, C.; Larsson, C.; Loman, N.; Malmberg, M.; Rydén, L.; et al. Refinement of breast cancer molecular classification by miRNA expression profiles. BMC Genom. 2019, 20, 503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. McDermott, A.M.; Miller, N.; Wall, D.; Martyn, L.M.; Ball, G.; Sweeney, K.J.; Kerin, M. Identification and Validation of Oncologic miRNA Biomarkers for Luminal A-like Breast Cancer. PLoS ONE 2014, 9, e87032. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Arabkari, V.; Clancy, E.; Dwyer, R.; Kerin, M.; Kalinina, O.; Holian, E.; Newell, J.; Smith, T.J. Relative and Absolute Expression Analysis of MicroRNAs Associated with Luminal a Breast Cancer—A Comparison. Pathol. Oncol. Res. 2019, 26, 833–844. [Google Scholar] [CrossRef] [PubMed]
  35. Endo, Y.; Toyama, T.; Takahashi, S.; Yoshimoto, N.; Iwasa, M.; Asano, T.; Fujii, Y.; Yamashita, H. miR-1290 and its potential targets are associated with characteristics of estrogen receptor α-positive breast cancer. Endocr.-Relat. Cancer 2012, 20, 91–102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Luo, Y.; Wang, X.; Niu, W.; Wang, H.; Wen, Q.; Fan, S.; Zhao, R.; Li, Z.; Xiong, W.; Peng, S.; et al. Elevated microRNA-125b levels predict a worse prognosis in HER2-positive breast cancer patients. Oncol. Lett. 2017, 13, 867–874. [Google Scholar] [CrossRef]
  37. Lowery, A.J.; Miller, N.; Devaney, A.; E McNeill, R.; A Davoren, P.; Lemetre, C.; Benes, V.; Schmidt, S.; Blake, J.; Ball, G.; et al. MicroRNA signatures predict oestrogen receptor, progesterone receptor and HER2/neureceptor status in breast cancer. Breast Cancer Res. 2009, 11, R27. [Google Scholar] [CrossRef]
  38. Estevão-Pereira, H.; Lobo, J.; Salta, S.; Amorim, M.; Lopes, P.; Cantante, M.; Reis, B.; Antunes, L.; Castro, F.; De Sousa, S.P.; et al. Overexpression of circulating MiR-30b-5p identifies advanced breast cancer. J. Transl. Med. 2019, 17, 435. [Google Scholar] [CrossRef] [Green Version]
  39. Bailey, S.T.; Westerling, T.; Brown, M. Loss of estrogen-regulated microRNA expression increases HER2 signaling and is prognostic of poor outcome in luminal breast cancer. Cancer Res. 2014, 75, 436–445. [Google Scholar] [CrossRef] [Green Version]
  40. Metzger-Filho, O.; Tutt, A.M.; de Azambuja, E.; Saini, K.S.; Viale, G.; Loi, S.; Bradbury, I.; Bliss, J.; Azim, H.H.A.; Ellis, P.; et al. Dissecting the Heterogeneity of Triple-Negative Breast Cancer. J. Clin. Oncol. 2012, 30, 1879–1887. [Google Scholar] [CrossRef] [Green Version]
  41. Lü, L.; Mao, X.; Shi, P.; He, B.; Xu, K.; Zhang, S.; Wang, J. MicroRNAs in the Prognosis of Triple-Negative Breast Cancer: A Systematic Review and Meta-Analysis. Medicine 2017, 96, e7085. [Google Scholar] [CrossRef]
  42. Gasparini, P.; Cascione, L.; Fassan, M.; Lovat, F.; Guler, G.; Balci, S.; Irkkan, C.; Morrison, C.; Croce, C.M.; Shapiro, C.L.; et al. microRNA expression profiling identifies a four microRNA signature as a novel diagnostic and prognostic biomarker in triple negative breast cancers. Oncotarget 2014, 5, 1174–1184. [Google Scholar] [CrossRef] [PubMed]
  43. Du, Y.; Wei, N.; Ma, R.; Jiang, S.; Song, D. A miR-210-3p regulon that controls the Warburg effect by modulating HIF-1α and p53 activity in triple-negative breast cancer. Cell Death Dis. 2020, 11, 731. [Google Scholar] [CrossRef] [PubMed]
  44. Sanges, F.; Floris, M.; Cossu-Rocca, P.; Muroni, M.R.; Pira, G.; Urru, S.A.M.; Barrocu, R.; Gallus, S.; Bosetti, C.; D’Incalci, M.; et al. Histologic subtyping affecting outcome of triple negative breast cancer: A large Sardinian population-based analysis. BMC Cancer 2020, 20, 491. [Google Scholar] [CrossRef] [PubMed]
  45. Moi, L.; Braaten, T.; Al-Shibli, K.; Lund, E.; Busund, L.-T.R. Differential expression of the miR-17-92 cluster and miR-17 family in breast cancer according to tumor type; results from the Norwegian Women and Cancer (NOWAC) study. J. Transl. Med. 2019, 17, 334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Liang, Q.; Ma, D.; Gao, R.-F.; Yu, K.-D. Effect of Ki-67 Expression Levels and Histological Grade on Breast Cancer Early Relapse in Patients with Different Immunohistochemical-based Subtypes. Sci. Rep. 2020, 10, 7648. [Google Scholar] [CrossRef]
  47. Ghafouri-Fard, S.; Khoshbakht, T.; Hussen, B.M.; Taheri, M.; Samadian, M. A Review on the Role of miR-1290 in Cell Proliferation, Apoptosis and Invasion. Front. Mol. Biosci. 2021, 8, 8. [Google Scholar] [CrossRef]
  48. Croset, M.; Pantano, F.; Kan, C.W.S.; Bonnelye, E.; Descotes, F.; Alix-Panabières, C.; Lecellier, C.-H.; Bachelier, R.; Allioli, N.; Hong, S.-S.; et al. miRNA-30 Family Members Inhibit Breast Cancer Invasion, Osteomimicry, and Bone Destruction by Directly Targeting Multiple Bone Metastasis—Associated Genes. Cancer Res. 2018, 78, 5259–5273. [Google Scholar] [CrossRef] [Green Version]
  49. Yin, H.; Wang, Y.; Wu, Y.; Zhang, X.; Zhang, X.; Liu, J.; Wang, T.; Fan, J.; Sun, J.; Yang, A.; et al. EZH2-mediated Epigenetic Silencing of miR-29/miR-30 targets LOXL4 and contributes to Tumorigenesis, Metastasis, and Immune Microenvironment Remodeling in Breast Cancer. Theranostics 2020, 10, 8494–8512. [Google Scholar] [CrossRef]
  50. Poudel, S.; Song, J.; Jin, E.-J.; Song, K. Sulfuretin-induced miR-30C selectively downregulates cyclin D1 and D2 and triggers cell death in human cancer cell lines. Biochem. Biophys. Res. Commun. 2013, 431, 572–578. [Google Scholar] [CrossRef]
  51. Chang, T.-C.; Yu, D.; Lee, Y.-S.; Wentzel, E.A.; Arking, D.E.; West, K.M.; Dang, C.V.; Thomas-Tikhonenko, A.; Mendell, J.T. Widespread microRNA repression by Myc contributes to tumorigenesis. Nat. Genet. 2008, 40, 43–50. [Google Scholar] [CrossRef] [Green Version]
  52. Wang, X.; Li, Y.; Qi, W.; Zhang, N.; Sun, M.; Huo, Q.; Cai, C.; Lv, S.; Yang, Q. MicroRNA-99a inhibits tumor aggressive phenotypes through regulating HOXA1 in breast cancer cells. Oncotarget 2015, 6, 32737–32747. [Google Scholar] [CrossRef] [Green Version]
  53. Hu, Y.; Zhu, Q.; Tang, L. MiR-99a Antitumor Activity in Human Breast Cancer Cells through Targeting of mTOR Expression. PLoS ONE 2014, 9, e92099. [Google Scholar] [CrossRef] [Green Version]
  54. Xia, M.; Li, H.; Wang, J.-J.; Zeng, H.-J.; Wang, S.-H. MiR-99a Suppress Proliferation, Migration and Invasion through Regu-lating Insulin-like Growth Factor 1 Receptor in Breast Cancer. Eur. Rev. Med. Pharmacol. Sci. 2016, 20, 1755–1763. [Google Scholar] [PubMed]
  55. Long, X.; Shi, Y.; Ye, P.; Guo, J.; Zhou, Q.; Tang, Y. MicroRNA-99a Suppresses Breast Cancer Progression by Targeting FGFR3. Front. Oncol. 2020, 9, 1473. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Loibl, S.; Gianni, L. HER2-positive breast cancer. Lancet 2017, 389, 2415–2429. [Google Scholar] [CrossRef]
  57. Normann, L.S.; Aure, M.R.; Leivonen, S.-K.; Haugen, M.H.; Hongisto, V.; Kristensen, V.N.; Mælandsmo, G.M.; Sahlberg, K.K. MicroRNA in combination with HER2-targeting drugs reduces breast cancer cell viability in vitro. Sci. Rep. 2021, 11, 10893. [Google Scholar] [CrossRef] [PubMed]
  58. Baselga, J. Why the Epidermal Growth Factor Receptor? The Rationale for Cancer Therapy. Oncologist 2002, 7 (Suppl. S4), 2–8. [Google Scholar] [CrossRef] [PubMed]
  59. Baselga, J.; Swain, S.M. Novel anticancer targets: Revisiting ERBB2 and discovering ERBB3. Nat. Cancer 2009, 9, 463–475. [Google Scholar] [CrossRef] [PubMed]
  60. Persson, H.; Kvist, A.; Rego, N.; Staaf, J.; Vallon-Christersson, J.; Luts, L.; Loman, N.; Jönsson, G.B.; Naya, H.; Hoglund, M.; et al. Identification of New MicroRNAs in Paired Normal and Tumor Breast Tissue Suggests a Dual Role for the ERBB2/Her2 Gene. Cancer Res. 2011, 71, 78–86. [Google Scholar] [CrossRef] [Green Version]
  61. Tashkandi, H.; Shah, N.; Patel, Y.; Chen, H. Identification of new miRNA biomarkers associated with HER2-positive breast cancers. Oncoscience 2015, 2, 924–929. [Google Scholar] [CrossRef] [Green Version]
  62. Ferracin, M.; Bassi, C.; Pedriali, M.; Pagotto, S.; D’Abundo, L.; Zagatti, B.; Corrà, F.; Musa, G.; Callegari, E.; Lupini, L.; et al. miR-125b targets erythropoietin and its receptor and their expression correlates with metastatic potential and ERBB2/HER2 expression. Mol. Cancer 2013, 12, 130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Banzhaf-Strathmann, J.; Edbauer, D. Good guy or bad guy: The opposing roles of microRNA 125b in cancer. Cell Commun. Signal. 2014, 12, 30. [Google Scholar] [CrossRef] [Green Version]
  64. Newie, I.; Søkilde, R.; Persson, H.; Grabau, D.; Rego, N.; Kvist, A.; Von Stedingk, K.; Axelson, H.; Borg, A.; Vallon-Christersson, J.; et al. The HER2-Encoded miR-4728-3p Regulates ESR1 through a Non-Canonical Internal Seed Interaction. PLoS ONE 2014, 9, e97200. [Google Scholar] [CrossRef] [PubMed]
  65. Floros, K.V.; Lochmann, T.L.; Hu, B.; Monterrubio, C.; Hughes, M.T.; Wells, J.D.; Morales, C.B.; Ghotra, M.S.; Costa, C.; Souers, A.J.; et al. Coamplification of miR-4728 protects HER2 -amplified breast cancers from targeted therapy. Proc. Natl. Acad. Sci. USA 2018, 115, E2594–E2603. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Newie, I.; Søkilde, R.; Persson, H.; Jacomasso, T.; Gorbatenko, A.; Borg, Å.; De Hoon, M.; Pedersen, S.F.; Rovira, C. HER2-encoded mir-4728 forms a receptor-independent circuit with miR-21-5p through the non-canonical poly(A) polymerase PAPD5. Sci. Rep. 2016, 6, 35664. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Kumarswamy, R.; Volkmann, I.; Thum, T. Regulation and function of miRNA-21 in health and disease. RNA Biol. 2011, 8, 706–713. [Google Scholar] [CrossRef] [Green Version]
  68. Jenike, A.E.; Halushka, M.K. miR-21: A non-specific biomarker of all maladies. Biomark. Res. 2021, 9, 18. [Google Scholar] [CrossRef] [PubMed]
  69. Pareja, F.; Geyer, F.C.; Marchiò, C.; Burke, K.A.; Weigelt, B.; Reis-Filho, J.S. Triple-negative breast cancer: The importance of molecular and histologic subtyping, and recognition of low-grade variants. npj Breast Cancer 2016, 2, 16036. [Google Scholar] [CrossRef]
  70. De Rinaldis, E.; Gazinska, P.; Mera, A.; Modrusan, Z.; Fedorowicz, G.M.; Burford, B.; Gillett, C.; Marra, P.; Grigoriadis, A.; Dornan, D.; et al. Integrated genomic analysis of triple-negative breast cancers reveals novel microRNAs associated with clinical and molecular phenotypes and sheds light on the pathways they control. BMC Genom. 2013, 14, 643. [Google Scholar] [CrossRef] [Green Version]
  71. Ventura, A.; Young, A.G.; Winslow, M.M.; Lintault, L.; Meissner, A.; Erkeland, S.J.; Newman, J.; Bronson, R.T.; Crowley, D.; Stone, J.R.; et al. Targeted Deletion Reveals Essential and Overlapping Functions of the miR-17~92 Family of miRNA Clusters. Cell 2008, 132, 875–886. [Google Scholar] [CrossRef] [Green Version]
  72. Mogilyansky, E.; Rigoutsos, I. The miR-17/92 cluster: A comprehensive update on its genomics, genetics, functions and increasingly important and numerous roles in health and disease. Cell Death Differ. 2013, 20, 1603–1614. [Google Scholar] [CrossRef] [PubMed]
  73. Dews, M.; Homayouni, A.; Yu, D.; Murphy, D.; Sevignani, C.; Wentzel, E.; E Furth, E.; Lee, W.M.; Enders, G.H.; Mendell, J.T.; et al. Augmentation of tumor angiogenesis by a Myc-activated microRNA cluster. Nat. Genet. 2006, 38, 1060–1065. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. O’Donnell, K.A.; Wentzel, E.A.; Zeller, K.I.; Dang, C.; Mendell, J.T. c-Myc-regulated microRNAs modulate E2F1 expression. Nature 2005, 435, 839–843. [Google Scholar] [CrossRef] [PubMed]
  75. Kalecky, K.; Modisette, R.; Pena, S.; Cho, Y.-R.; Taube, J. Integrative analysis of breast cancer profiles in TCGA by TNBC subgrouping reveals novel microRNA-specific clusters, including miR-17-92a, distinguishing basal-like 1 and basal-like 2 TNBC subtypes. BMC Cancer 2020, 20, 141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Liu, H.; Paddock, M.N.; Wang, H.; Murphy, C.J.; Geck, R.C.; Navarro, A.J.; Wulf, G.M.; Elemento, O.; Haucke, V.; Cantley, L.C.; et al. The INPP4B Tumor Suppressor Modulates EGFR Trafficking and Promotes Triple-Negative Breast Cancer. Cancer Discov. 2020, 10, 1226–1239. [Google Scholar] [CrossRef] [PubMed]
  77. Wang, D.-Y.; Jiang, Z.; Ben-David, Y.; Woodgett, J.R.; Zacksenhaus, E. Molecular stratification within triple-negative breast cancer subtypes. Sci. Rep. 2019, 9, 19107. [Google Scholar] [CrossRef] [Green Version]
  78. Chakraborty, S.; Krishnan, Y. A structural map of oncomiR-1 at single-nucleotide resolution. Nucleic Acids Res. 2017, 45, 9694–9705. [Google Scholar] [CrossRef] [Green Version]
  79. Donayo, A.O.; Johnson, R.M.; Tseng, H.-W.; Izreig, S.; Gariepy, A.; Mayya, V.K.; Wu, E.; Alam, R.; Lussier, C.; Jones, R.G.; et al. Oncogenic Biogenesis of pri-miR-17~92 Reveals Hierarchy and Competition among Polycistronic MicroRNAs. Mol. Cell 2019, 75, 340–356. [Google Scholar] [CrossRef]
  80. Fuziwara, C.S.; Kimura, E.T. Insights into Regulation of the miR-17-92 Cluster of miRNAs in Cancer. Front. Med. 2015, 2, 64. [Google Scholar] [CrossRef] [Green Version]
  81. Uva, P.; Cossu-Rocca, P.; Loi, F.; Pira, G.; Murgia, L.; Orrù, S.; Floris, M.; Muroni, M.R.; Sanges, F.; Carru, C.; et al. miRNA-135b Contributes to Triple Negative Breast Cancer Molecular Heterogeneity: Different Expression Profile in Basal-like Versus non-Basal-like Phenotypes. Int. J. Med Sci. 2018, 15, 536–548. [Google Scholar] [CrossRef] [Green Version]
  82. Aakula, A.; Leivonen, S.-K.; Hintsanen, P.; Aittokallio, T.; Ceder, Y.; Børresen-Dale, A.-L.; Perälä, M.; Östling, P.; Kallioniemi, O. MicroRNA-135b regulates ERα, AR and HIF1AN and affects breast and prostate cancer cell growth. Mol. Oncol. 2015, 9, 1287–1300. [Google Scholar] [CrossRef]
  83. Hua, K.; Jin, J.; Zhao, J.; Song, J.; Song, H.; Li, D.; Maskey, N.; Zhao, B.; Wu, C.; Xu, H.; et al. miR-135b, upregulated in breast cancer, promotes cell growth and disrupts the cell cycle by regulating LATS2. Int. J. Oncol. 2016, 48, 1997–2006. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Lv, Z.; Xin, H.; Yang, Z.; Wang, W.; Dong, J.; Jin, L.; Li, F. miR-135b promotes proliferation and metastasis by targeting APC in triple-negative breast cancer. J. Cell. Physiol. 2019, 234, 10819–10826. [Google Scholar] [CrossRef] [PubMed]
  85. Pu, T.; Shen, M.; Li, S.; Yang, L.; Gao, H.; Xiao, L.; Zhong, X.; Zheng, H.; Liu, Y.; Ye, F.; et al. Repression of miR-135b-5p promotes metastasis of early-stage breast cancer by regulating downstream target SDCBP. Lab. Investig. 2019, 99, 1296–1308. [Google Scholar] [CrossRef]
  86. Adams, B.D.; Wali, V.B.; Cheng, C.J.; Inukai, S.; Booth, C.J.; Agarwal, S.; Rimm, D.L.; Győrffy, B.; Santarpia, L.; Pusztai, L.; et al. miR-34a Silences c-SRC to Attenuate Tumor Growth in Triple-Negative Breast Cancer. Cancer Res. 2016, 76, 927–939. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Kato, M.; Paranjape, T.; Ullrich, R.; Nallur, S.; Gillespie, E.; Keane, K.; Esquela-Kerscher, A.; Weidhaas, J.B.; Slack, F.J. The mir-34 microRNA is required for the DNA damage response in vivo in C. elegans and in vitro in human breast cancer cells. Oncogene 2009, 28, 2419–2424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Weng, Y.-S.; Tseng, H.-Y.; Chen, Y.-A.; Shen, P.-C.; Al Haq, A.T.; Chen, L.-M.; Tung, Y.-C.; Hsu, H.-L. 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 2019, 18, 42. [Google Scholar] [CrossRef] [Green Version]
  89. Koutsaki, M.; Spandidos, D.; Zaravinos, A. Epithelial—Mesenchymal transition-associated miRNAs in ovarian carcinoma, with highlight on the miR-200 family: Prognostic value and prospective role in ovarian cancer therapeutics. Cancer Lett. 2014, 351, 173–181. [Google Scholar] [CrossRef]
  90. Zhang, Q.; Wang, J.; Li, N.; Liu, Z.; Chen, Z.; Li, Z.; Lai, Y.; Shen, L.; Gao, J. miR-34a increases the sensitivity of colorectal cancer cells to 5-fluorouracil in vitro and in vivo. Am. J. Cancer Res. 2018, 8, 280–290. [Google Scholar]
  91. Hur, K.; Toiyama, Y.; Takahashi, M.; Balaguer, F.; Nagasaka, T.; Koike, J.; Hemmi, H.; Koi, M.; Boland, C.R.; Goel, A. MicroRNA-200c modulates epithelial-to-mesenchymal transition (EMT) in human colorectal cancer metastasis. Gut 2013, 62, 1315–1326. [Google Scholar] [CrossRef] [Green Version]
  92. Senfter, D.; Madlener, S.; Krupitza, G.; Mader, R.M. The microRNA-200 family: Still much to discover. Biomol. Concepts 2016, 7, 311–319. [Google Scholar] [CrossRef] [PubMed]
  93. Cui, J.; Gong, M.; Fang, S.; Hu, C.; Wang, Y.; Zhang, J.; Tang, N.; He, Y. All-trans retinoic acid reverses malignant biological behavior of hepatocarcinoma cells by regulating miR-200 family members. Genes Dis. 2021, 8, 509–520. [Google Scholar] [CrossRef] [PubMed]
  94. Neves, R.; Scheel, C.; Weinhold, S.; Honisch, E.; Iwaniuk, K.M.; Trompeter, H.-I.; Niederacher, D.; Wernet, P.; Santourlidis, S.; Uhrberg, M. Role of DNA methylation in miR-200c/141 cluster silencing in invasive breast cancer cells. BMC Res. Notes 2010, 3, 219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Davalos, V.; Moutinho, C.; Villanueva, A.; Boque, R.; Silva, P.; Carneiro, F.; Esteller, M. Dynamic epigenetic regulation of the microRNA-200 family mediates epithelial and mesenchymal transitions in human tumorigenesis. Oncogene 2011, 31, 2062–2074. [Google Scholar] [CrossRef] [PubMed]
  96. Wang, Q.; Cheng, Y.; Wang, Y.; Fan, Y.; Li, C.; Zhang, Y.; Wang, Y.; Dong, Q.; Ma, Y.; Teng, Y.-E.; et al. Tamoxifen reverses epithelial—mesenchymal transition by demethylating miR-200c in triple-negative breast cancer cells. BMC Cancer 2017, 17, 492. [Google Scholar] [CrossRef]
  97. Damiano, V.; Brisotto, G.; Borgna, S.; di Gennaro, A.; Armellin, M.; Perin, T.; Guardascione, M.; Maestro, R.; Santarosa, M. Epigenetic silencing of miR-200c in breast cancer is associated with aggressiveness and is modulated by ZEB1. Genes Chromosom. Cancer 2017, 56, 147–158. [Google Scholar] [CrossRef]
  98. Krützfeldt, J.; Rajewsky, N.; Braich, R.; Rajeev, K.G.; Tuschl, T.; Manoharan, M.; Stoffel, M. Silencing of microRNAs in vivo with ‘antagomirs’. Nature 2005, 438, 685–689. [Google Scholar] [CrossRef]
  99. Kaboli, P.J.; Rahmat, A.; Ismail, P.; Ling, K.-H. MicroRNA-based therapy and breast cancer: A comprehensive review of novel therapeutic strategies from diagnosis to treatment. Pharmacol. Res. 2015, 97, 104–121. [Google Scholar] [CrossRef]
  100. Bader, A.G.; Brown, D.L.; Stoudemire, J.; Lammers, P.J. Developing therapeutic microRNAs for cancer. Gene Ther. 2011, 18, 1121–1126. [Google Scholar] [CrossRef] [Green Version]
  101. Li, L.; Xie, X.; Luo, J.; Liu, M.; Xi, S.; Guo, J.; Kong, Y.; Wu, M.; Gao, J.; Xie, Z.; et al. Targeted Expression of miR-34a Using the T-VISA System Suppresses Breast Cancer Cell Growth and Invasion. Mol. Ther. 2012, 20, 2326–2334. [Google Scholar] [CrossRef] [Green Version]
  102. Deng, X.; Cao, M.; Zhang, J.; Hu, K.; Yin, Z.; Zhou, Z.; Xiao, X.; Yang, Y.; Sheng, W.; Wu, Y.; et al. Hyaluronic acid-chitosan nanoparticles for co-delivery of MiR-34a and doxorubicin in therapy against triple negative breast cancer. Biomaterials 2014, 35, 4333–4344. [Google Scholar] [CrossRef] [PubMed]
  103. Ma, L.; Reinhardt, F.; Pan, E.; Soutschek, J.; Bhat, B.; Marcusson, E.G.; Teruya-Feldstein, J.; Bell, G.W.; A Weinberg, R. Therapeutic silencing of miR-10b inhibits metastasis in a mouse mammary tumor model. Nat. Biotechnol. 2010, 28, 341–347. [Google Scholar] [CrossRef] [PubMed]
  104. Devulapally, R.; Sekar, N.M.; Sekar, T.V.; Foygel, K.; Massoud, T.F.; Willmann, J.K.; Paulmurugan, R. Polymer Nanoparticles Mediated Codelivery of AntimiR-10b and AntimiR-21 for Achieving Triple Negative Breast Cancer Therapy. ACS Nano 2015, 9, 2290–2302. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Yin, H.; Xiong, G.; Guo, S.; Xu, C.; Xu, R.; Guo, P.; Shu, D. Delivery of Anti-miRNA for Triple-Negative Breast Cancer Therapy Using RNA Nanoparticles Targeting Stem Cell Marker CD133. Mol. Ther. 2019, 27, 1252–1261. [Google Scholar] [CrossRef] [Green Version]
  106. Holliday, D.L.; Speirs, V. Choosing the right cell line for breast cancer research. Breast Cancer Res. 2011, 13, 215. [Google Scholar] [CrossRef] [Green Version]
  107. Dai, X.; Cheng, H.; Bai, Z.; Li, J. Breast Cancer Cell Line Classification and Its Relevance with Breast Tumor Subtyping. J. Cancer 2017, 8, 3131–3141. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Examples of significant microRNAs (miRNAs) associated with specific breast cancer subtypes and their effects on cell phenotypes. In estrogen receptor positive/progesterone receptor positive (ER+/PR+) breast cancers (blue box), we note the role of miR-100 and miR-30 families and the distinction between the luminal A and luminal B molecular subtypes. In human epidermal growth factor receptor 2 positive (HER2+) breast cancers (yellow box), we have noted miR-4728-3p, which is present in the intronic region of HER2 and is co-expressed with HER2. The miRNA is involved in feedback regulation of HER2 and oncogenic miR-21-5p. This facilitates several oncogenic processes in later stage tumors. In triple negative breast cancers (TNBCs) (purple box), we have noted the cMYC oncogene driven miR-17~92 cluster, which is overexpressed in TNBCs, specifically the BL1 molecular subtype. The miRNA cluster promotes proliferation through its direct targets, which include phosphate and tensin homolog (PTEN) and inositol polyphosphate-4-phosphatase type II B (INPP4B) and are inhibitors of the proliferation mediator AKT. The migratory phenotype of TNBC cell lines that fall within the mesenchymal stem like (MSL) and mesenchymal (M) molecular subtypes is facilitated by inhibition of the miR-200 family through epigenetic changes that allows expression of epithelial to mesenchymal transition (EMT) and migration genes, resulting in the migratory phenotype.
Figure 1. Examples of significant microRNAs (miRNAs) associated with specific breast cancer subtypes and their effects on cell phenotypes. In estrogen receptor positive/progesterone receptor positive (ER+/PR+) breast cancers (blue box), we note the role of miR-100 and miR-30 families and the distinction between the luminal A and luminal B molecular subtypes. In human epidermal growth factor receptor 2 positive (HER2+) breast cancers (yellow box), we have noted miR-4728-3p, which is present in the intronic region of HER2 and is co-expressed with HER2. The miRNA is involved in feedback regulation of HER2 and oncogenic miR-21-5p. This facilitates several oncogenic processes in later stage tumors. In triple negative breast cancers (TNBCs) (purple box), we have noted the cMYC oncogene driven miR-17~92 cluster, which is overexpressed in TNBCs, specifically the BL1 molecular subtype. The miRNA cluster promotes proliferation through its direct targets, which include phosphate and tensin homolog (PTEN) and inositol polyphosphate-4-phosphatase type II B (INPP4B) and are inhibitors of the proliferation mediator AKT. The migratory phenotype of TNBC cell lines that fall within the mesenchymal stem like (MSL) and mesenchymal (M) molecular subtypes is facilitated by inhibition of the miR-200 family through epigenetic changes that allows expression of epithelial to mesenchymal transition (EMT) and migration genes, resulting in the migratory phenotype.
Biomedicines 10 00651 g001
Table 1. Distinct microRNA (miRNA) signatures and individual miRNAs associated with specific breast cancer subtypes. Breast cancer gene (BRCA), estrogen receptor (ER), progesterone receptor (PR), triple negative breast cancer (TNBC), human epidermal growth factor receptor 2 (HER2), quantitative real time polymerase chain reaction (qPCR), The Cancer Genome Atlas (TCGA), gene expression omnibus (GEO), formalin-fixed paraffin-embedded (FFPE).
Table 1. Distinct microRNA (miRNA) signatures and individual miRNAs associated with specific breast cancer subtypes. Breast cancer gene (BRCA), estrogen receptor (ER), progesterone receptor (PR), triple negative breast cancer (TNBC), human epidermal growth factor receptor 2 (HER2), quantitative real time polymerase chain reaction (qPCR), The Cancer Genome Atlas (TCGA), gene expression omnibus (GEO), formalin-fixed paraffin-embedded (FFPE).
Analysis TypeFindingsmiRNA SignatureSubtypeReference
microRNA (miRNA)quantitative real-time polymerase chain reaction (qPCR) panel on 139 breast cancer (BRCA) patient
tissues compared against 26 normal tissues
patients with miR-182-5p and
miR-200b-3p expression show
better prognosis
miR-30c-5p,
miR-30b-5p,
miR-182-5p, and
miR-200b-3p
ER/PR +[30]
case control study of estrogen receptor/progesterone receptor positive (ER/PR +) patients with tamoxifen treatmentmiR-221 expression is high in
ER/PR + patients and is not changed by KI67/PR levels
miR-221ER/PR +[31]
clustering analysis of RNA seq from patient cohort of 186 patientsmiR-99a/let-7c/miR-125b cluster is high in luminal A compared to luminal BmiR-99a/let-7c/
miR-125b
ER/PR +[32]
qPCR of 54 luminal A type patients against 56 controlsstudy identified miRNAs specifically downregulated in luminal A type patientsmiR-29a, miR-625, miR-181aER/PR +[33]
miRNA qPCR of luminal A patients compared against controlsdiagnostic markers for luminal AmiR-145, miR-195 and miR-486ER/PR+[34]
immunohistochemistry of miR-1290
targets among 256 ER positive breast cancer
miR-1290 is a prognostic marker for luminal breast cancersmiR-1290ER/PR+[35]
meta-analysis of patient datasetsspecific miRNA signature between
luminal A and luminal B breast cancer subtypes
miR-30b-5p,
miR-30c-5p high in luminal A,
miR-182-5p,
miR-200b-3p,
miR-15b-3p,
miR-149-5p,
miR-193b-3p and miR-342-3p, 5p high in luminal B
ER/PR+[22]
mimic transfection, luciferase activity and qPCRmiR-125b functions as a competitive endogenous RNA with EPOR and ERBB2miR-125bHER2+[36]
in silico and qPCR analysis of 300 miRNAsupregulated miRNA biomarkers for human epidermal growth factor receptor 2 (HER2) subtypemiR-146a-5pHER2+[37]
in silico and qPCR analysis of 300 miRNAsdownregulated miRNA biomarkers for HER2 subtypemiR-181d and
miR-195-5p
HER2+[38]
miRNA screen (1626) in combination with targeted treatments lapatinib and trastuzumabtumor suppressive miRNA signature identified, treatment with mimics sensitize cells to trastuzumab and lapatinibmiR-101-5p,
mir-518a-5p,
miR-19b-2-5p,
miR-1237-3p,
miR-29a-3p,
miR-29c-3p,
miR-106a-5p, and miR-744-3p
HER2+[39]
protein expression and The Cancer Genome Atlas (TCGA) data analysisoverexpression of miR-4728 in HER2 minimizes the effect of laptinibmiR-4728
triple negative breast cancer (TNBC) vs. non-TNBC patient samplesdiagnostic markers of TNBCshsa-miR-10a, hsa-miR-18a, hsa-miR-135b and hsa-miR-577TNBC[40]
miR arrays from stored tissuesdistinct in TNBCs compared to ER negative patientsmiR-10a, miR-18a, miR-135b and miR-577TNBC[41]
miR arrays from stored tissuesbasal-like subtype has overexpression of both clusters, derived from copy numbermiR-17-92 and
miR-106b-25 cluster overexpression
TNBC[42]
upregulated in TNBCs, prognostic signaturemiR-455-3p,
miR-107,
miR-146b-5p,
miR-17-5p,
miR-324-5p,
miR-20a-5p and
miR-142-3p,
downregulated in TNBCsmiR-139-5p, miR-10b-5p, miR-486-5p
regression analysis of patient data compared against gene expression omnibus (GEO) datasetsupregulated in TNBCsmiR-455-3p,
miR-107,
miR-146b-5p,
miR-17-5p,
miR-324-5p,
miR-20a-5p and
miR-142-3p
TNBC[43]
clustering of miRNAs significantly different between TNBCs and
HER2+ subtypes
miR-139-5p, miR-10b-5p, miR-486-5p
meta-analysis of published research articlesupregulated in TNBCs, prognostic signaturemiR-10b, miR-21, miR-29, miR-9, miR-221/222, miR-373TNBC[44]
downregulated in TNBCsmiR-145,
miR-199a-5p,
miR-200 family,
miR-203, miR-205
miRNA extraction and microarray from formalin-fixed paraffin-embedded (FFPE) tissuesTNBC-specific four miRNA signature which is reduced in other subtypesmiR-17-5p,
miR-20a-5p,
miR-92a-3p,
miR-106b-5p
TNBC[45]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Arun, R.P.; Cahill, H.F.; Marcato, P. Breast Cancer Subtype-Specific miRNAs: Networks, Impacts, and the Potential for Intervention. Biomedicines 2022, 10, 651. https://doi.org/10.3390/biomedicines10030651

AMA Style

Arun RP, Cahill HF, Marcato P. Breast Cancer Subtype-Specific miRNAs: Networks, Impacts, and the Potential for Intervention. Biomedicines. 2022; 10(3):651. https://doi.org/10.3390/biomedicines10030651

Chicago/Turabian Style

Arun, Raj Pranap, Hannah F. Cahill, and Paola Marcato. 2022. "Breast Cancer Subtype-Specific miRNAs: Networks, Impacts, and the Potential for Intervention" Biomedicines 10, no. 3: 651. https://doi.org/10.3390/biomedicines10030651

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop