Next Article in Journal
Integrated Transcriptome and Metabolome Dissecting Interaction between Vitis vinifera L. and Grapevine Fabavirus
Next Article in Special Issue
Comparative Proteomics Analysis of Exosomes Identifies Key Pathways and Protein Markers Related to Breast Cancer Metastasis
Previous Article in Journal
Mesoporous Bioactive Nanoparticles for Bone Tissue Applications
Previous Article in Special Issue
Genetic Heterogeneity, Tumor Microenvironment and Immunotherapy in Triple-Negative Breast Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Regulation of Inflammasome by microRNAs in Triple-Negative Breast Cancer: New Opportunities for Therapy

by
Liliana-Roxana Balahura (Stămat)
1,
Sorina Dinescu
1,2,* and
Marieta Costache
1,2
1
Department of Biochemistry and Molecular Biology, University of Bucharest, 050095 Bucharest, Romania
2
Research Institute of the University of Bucharest, 050663 Bucharest, Romania
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(4), 3245; https://doi.org/10.3390/ijms24043245
Submission received: 21 December 2022 / Revised: 28 January 2023 / Accepted: 2 February 2023 / Published: 7 February 2023
(This article belongs to the Special Issue Targeting Breast Cancer: Strategies and Hopes)

Abstract

:
During the past decade, researchers have investigated the molecular mechanisms of breast cancer initiation and progression, especially triple-negative breast cancer (TNBC), in order to identify specific biomarkers that could serve as feasible targets for innovative therapeutic strategies development. TNBC is characterized by a dynamic and aggressive nature, due to the absence of estrogen, progesterone and human epidermal growth factor 2 receptors. TNBC progression is associated with the dysregulation of nucleotide-binding oligomerization domain-like receptor and pyrin domain-containing protein 3 (NLRP3) inflammasome, followed by the release of pro-inflammatory cytokines and caspase-1 dependent cell death, termed pyroptosis. The heterogeneity of the breast tumor microenvironment triggers the interest of non-coding RNAs’ involvement in NLRP3 inflammasome assembly, TNBC progression and metastasis. Non-coding RNAs are paramount regulators of carcinogenesis and inflammasome pathways, which could help in the development of efficient treatments. This review aims to highlight the contribution of non-coding RNAs that support inflammasome activation and TNBC progression, pointing up their potential for clinical applications as biomarkers for diagnosis and therapy.

1. Introduction

The breast is a complex structure, which is organized in 15 to 25 lobes of different sizes that are connected with lactiferous ducts that terminate in the nipple, ductal formations and glandular tissue, all of these being surrounded by fibro-adipose tissue [1]. Specifically, the adult breast is a tissue highly variable in conformation, adiposity and volume, due to the different proportions between adipose, fibrous and glandular tissue, among which are also found blood and lymphatic vessels. The corresponding distribution of fat and collagenous components differs among women and is influenced by hormonal, physiologic and environmental factors [2].
Breast cancer (BC) is a dynamic, aggressive and heterogeneous disease that is the principle cause of death among women worldwide, but that also affects men [3]. This neoplasm has the potential to be determined by exposure to both genetic and non-genetic risk factors such as gender, age, menopause, nulliparity, obesity, alcohol abuse and exposure to hormones, radiation or therapy [1]. The early detection and diagnosis of BC is based on screening techniques (ultrasound, mammography, contrast-enhanced digital mammography, magnetic resonance imaging and positron emission tomography), microwave imaging techniques (microwave tomographic, radar-based microwave imaging and radiometry), biomarker-based techniques (radioimmunoassay, immunohistochemistry, enzyme-linked immunosorbent assay and fluoroimmunoassay) and breast tissue biopsies, which are used to differentiate between malign and benign tumors [4].
Following a diagnosis of BC, it is necessary to stage it according to the American Joint Committee on Cancer (AJCC) tumor, nodes, and metastasis (TNM) system, as TNM staging is used to define and stratify the size of the tumors (T), the status of regional lymph nodes (N), and distant metastasis (M) [5]. TNM staging is divided into four classes: (I) clinical staging, which includes information from clinical examination; (II) pathological staging, which includes the affected anatomical formations; (III) post-therapy staging, which includes clinical and pathologic information; and (IV) restaging, if necessary [6].
In order to establish the most efficient treatment for patients, the histological classification is completed via the molecular classification of BC that is based on the expression profiles of the estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 2 (HER2) and Ki67 antigen. The main molecular subtypes of breast cancer are luminal A (ER positive, PR positive and HER2 negative), luminal B (ER positive, PR negative and HER2 negative), HER2-enriched (ER negative, PR negative and HER2 positive), triple-negative breast cancer (TNBC), basal-like (ER negative, PR negative and HER2 negative), claudin low (ER negative, claudin negative, vimentin positive, E-cadherin low) and normal breast-like (adipose tissue gene signature) [7]. Luminal and HER2-enriched subtypes are associated with good prognoses and are highly responsive to therapy, resulting in a greatly improved outcome, while TNBC is the most aggressive subtype of BC, which is characterized by a high cell proliferation rate and a tendency to relapse [8]. Experimental studies indicate the presence of two main pathways involved in low-grade and high-grade breast tumorigenesis; low-grade BCs are regularly ER positive, PR positive and HER2 negative, while high-grade BCs are ER negative, PR negative and HER2 positive [9].
The mechanism of BC is dependent on the genetic modification and molecular processes that determine initiation, transformation and progression from normal tissue to tumor tissue [10]. More than 90% of diagnosed BCs are associated with the mutation of specific genes, such as breast cancer genes 1 and 2 (BRCA1 and BRCA2), TP53, phosphatase and tensin homolog (PTEN), serine/threonine kinase 11 (STK11), ataxia-telangiectasia mutated (ATM), BRCA1 interacting protein 1 (BRIP1) or the partner and localizer of BRCA2 (PALB2), etc. [11].
In addition, BC aggressiveness is supported by chronic inflammation considering the up-regulation of pro-inflammatory cytokines, such as interleukin (IL)-1β, IL-6, IL-18 or tumor necrosis factor-α (TNF-α), growth factors or free radicals [12]. The secretion and release of pro-inflammatory cytokines are associated with inflammasome complex activation. Inflammasome is a cytoplasmic multiprotein complex, composed of a sensor (NOD-like receptor protein (NLRP)), an adaptor (an apoptosis-associated speck-like protein containing a caspase-activation and recruitment domain (CARD) (ASC)) and effector molecules (pro-caspase). The inflammasome complex is involved in numerous physiological and pathological mechanisms, including different types of cancer, although the activation of this complex can have both a positive or negative impact on carcinogenesis [13]. First of all, inflammasome activity stimulates epithelial mesenchymal transition (EMT), metastasis and angiogenesis, while inhibiting apoptosis and enhancing tumor development. On the other hand, the inflammasome pathway is also associated with immune reactions and the programmed death of tumor cells through pyroptosis [14].
The dynamic interaction between BC and inflammasome is regulated by a complex molecular network, including non-coding RNAs (ncRNAs). NcRNAs can be classified into short non-coding RNAs (sncRNAs) or long non-coding RNAs (lncRNAs), based on their number of nucleotides [15]. Among the most studied sncRNAs are microRNAs (miRNAs), which are single-stranded ribonucleic acid molecules involved in diverse cellular processes (cell survival, proliferation and adhesion, motility, cell death, inflammation, carcinogenesis, etc.). According to their implication in tumorigenic mechanisms and metastasis, miRNA molecules can be classified into oncogenic miRNAs and suppressor miRNAs [16]. Increasing evidence has indicated that many miRNAs are involved in the regulation of the inflammasome complex (miR-7, miR-9, miR-20a, miR-21, miR-23a, miR-30e, miR-33, miR-132, miR-133, miR-146, miR-155, miR-223, miR-296, miR-377, miR-711, etc.) and that these molecules represent a link between inflammasome activity and BC, especially TNBC [17]. Recently, researchers’ attention has been directed towards targeting these molecules as possible therapeutic strategies for the treatment of BC, due to their significance in post-transcriptional regulation [18].
Considering the importance of this topic and the ongoing expertise, this review aims to highlight the molecular mechanisms involved in BC correlated with the significance of the inflammasome pathway and miRNAs. Furthermore, another target is to accentuate the complex network and dual role of miRNAs (oncogenic and suppressor), in doing so emphasizing RNA-based techniques’ potential for clinical applications for diagnosis and therapy.

2. Molecular Characteristics of TNBC

TNBC is a highly invasive and aggressive type of BC, which is characterized by the absence of ER, PR and HER2 expression. TNBC represents almost 20% of all diagnosed BC, being associated with resistance to chemotherapy, a predisposition to metastasis, poor prognoses and reduced survival rates [19]. Over the years, researchers have studied the molecular signatures of TNBC using advanced techniques and, according to gene expression profiles, Lehmann et al. [20], Burstein et al. [21] and Jezequel et al. [22] have all proposed different classification of TNBC in order to investigate possible therapeutic targets to improve the outcomes of TNBC (Table 1). Moreover, according to Lehmann et al., TNBC can be subdivided into six groups: basal-like 1, basal-like 2, mesenchymal, mesenchymal stem-like, immunomodulatory, and luminal androgen receptor TNBC [20]. Burstein et al. proposed a TNBC classification divided into four subtypes: basal-like immune-activated, basal-like immune-suppressed, mesenchymal, and luminal androgen receptor TNBC [21], while Jezequel et al. divided triple-negative tumors into three clusters: C1 (22.4%), C2 (44.9%) and C3 (32.7%) [22]. Classifying and understanding the particularities of TNBC allow for the development of personalized medicine, because each subtype has different characteristics and responses to anti-tumor therapy [23].
The breast tumor microenvironment (TME) is associated with chronic inflammatory reactions, with inflammatory infiltrate being a prognostic marker of cancer. Pro-inflammatory molecules together with their specific receptors are involved in the development of TNBC due to their capability to promote cell differentiation and angiogenesis, to recruit immune cells and to influence the immune system [46]. Lymphocytes, macrophages and fibroblasts are the most common cell types of the TME and secrete different factors (such as cytokines, chemokines, enzymes, growth factors, etc.), which are associated with an aggressive malignancy and high risk of metastasis. Pro-inflammatory mediators (IL-6, CCL2, COX2, and TNF-α) are generally up-regulated in breast tumor stroma; IL-1, IL-6, IL-8, IL-11, IL-18 and IL-23 are among the most studied inflammatory factors involved in inflammation, TNBC, invasion and metastasis [47]. In close correlation with the development of inflammatory reactions and TNBC is the activation of the NLRP3 inflammasome complex, with IL-1β and IL-18 being the two major cytokines activated by NLRP3 inflammasome, which promote tumor cell proliferation and invasion [48].
Over the years, new signaling pathways within breast TME have been discovered, which have opened up new perspectives for the development of strategies that aim to block these signals that promote tumorigenesis, angiogenesis and metastasis. Specifically, conventional therapeutic strategies are the gold standard for neoadjuvant treatment represented by a combination between anthracyclines and taxanes, capecitabine and taxane and ixabepilone monotherapy (paclitaxel, 5-fluorouracil, doxorubicin, cisplatin, carboplatin, abraxane, bevacizumab, cyclophosphamide, ixabepilone, capecitabine, etc.), adjuvant treatment (anthracycline-based drugs), surgery (mastectomy and lumpectomy) and radiotherapy [49].
Despite the heterogeneity and aggressiveness of TNBC, the corresponding standard of care (SOC) requires neoadjuvant chemotherapy, followed by surgery and adjuvant chemotherapy (Table 2). There are several treatment schemes for early diagnosed TNBC that are based on anthracycline and taxane administration, but which cause numerous adverse effects (nausea, fatigue, gastrointestinal toxicity, myelosuppression, alopecia, hypothyroidism, hyperthyroidism, pneumonitis, skin reactions, adrenal insufficiency, peripheral neuropathy, neutropenia, pyrexia, anemia, thrombocytopenia, electrolyte abnormalities, infection, etc.) and do not lower the relapse rate [50].
Although neoadjuvant chemotherapy is the standard treatment approach for diagnosed TNBC, the therapeutic potential of other agents that interfere with various molecular mechanisms (such as angiogenesis, the immune response, the cell cycle, etc.) have been tested or are currently under testing and approval (Table 3). The optimal treatment scheme for TNBC is a great challenge due to the disease’s heterogeneity and increased risk of relapse, so the researchers have explored several promising anti-tumor agents (pembrolizumab, bevacizumab, olaparib, sacituzumab govitecan, etc.) [53]. Other therapeutic approaches are represented by platinum salts (carboplatin, cisplatin, etc.) immunotherapies that target the programmed cell death-1 (PD-1) receptor/PD-L1 pathway, immune-checkpoint inhibitors, poly-adenosine diphosphate ribose polymerase (PARP) inhibitors or AKT inhibitors [54].
The molecular characteristics of TNBC tumors (heterogeneity and the chemoresistance mechanism) raise difficulties in establishing an effective conventional therapeutic strategy; therefore advanced therapeutic strategies have been developed. There are two main types of advanced therapeutic treatments: passive transport (or enhanced permeability and retention (nanoparticles)) and active transport (miRNA and aptamers) [49].

3. Molecular Mechanisms of Inflammasome Activation during TNBC

Discovered in 2002 by Martinon et al., inflammasome represents the perfect structure bridging inflammation, tumorigenesis, angiogenesis and metastasis [45]. The inflammasome complex is a cytoplasmic multiproteic platform that comprises three components: the sensor molecule, the adaptor protein and the effector (Figure 1). The inflammasome pathway is activated by the recognition of specific stimuli, danger-associated molecular patterns (DAMPs), pathogen-associated molecular patterns (PAMPs), microbe-associated molecular patterns (MAMPs), and homeostasis-altering molecular processes (HAMPs) (such as ATP, toxins, K+ efflux, reactive oxygen species, etc.) by toll-like receptors (TLRs), the nucleotide-binding oligomerization domain (NOD) and leucine-rich repeat (LRR)-containing receptors (NLRs), the absent in melanoma-2 (AIM2)-like receptors (ALRs)), retinoic acid-inducible gene (RIG)-I-like receptors (RLRs) or C-type lectin receptors (CLRs). Inflammasome-related sensor molecules consist of pathogen recognition receptors (PRRs), such as TLRs, NLRs, ALRs, RLRs, CLRs and proteins that contain a tripartite motif, such as pyrin (also known as marenostrin or TRIM20) [45].
The activation of inflammasome is carried out by two main signals: priming and activation. The priming stimuli of the inflammasome are activate TLRs, the FAS-associated death domain protein and IL-1 receptor ligands, which activate the NF-kB signaling pathway, causing the up-regulation of inflammasome expression. In normal conditions, the expression of inflammasome components is relatively low and unable to initiate inflammasome activation. In this stage, two signaling molecules from the NF-kB pathway, MyD88 and TIR-domain-containing adapter-inducing interferon-β (TRIF), are involved in inflammasome regulation and pro-IL-1β secretion, but the expression of ASC, pro-caspase-1 and pro-IL-18 remains unmodified. Following the priming signal, the second signal, which is induced by the presence of DAMPs or PAMPs, is required in order to promote the ASC oligomerization, inflammasome assembly and caspase-1 (CASP1) activation [67].
These sensors can determine the oligomerization of the adapter protein ASC, also known as PYCARD, and the activation of the cysteine protease CASP1 [68]. After the detection of specific stimuli by the receptors, the ASC adapter proteins are assembled and form speck-like structures, which recruit pro-CASP1 that is cleaved into mature CASP1. Active CASP1 cleaves the pro-inflammatory cytokines (pro-IL-1β and pro-IL-18) and pore-forming gasdermin D (GSDMD) in order to promote the CASP1-dependent cell death, termed pyroptosis, with the pyroptosis’ mechanism being directed by the gasdermin family (GSDMA, GSDMB, GSDMC, GSDMD, GSDME and DFNB59) [69].
The classification of inflammasome is based on the structure of the receptor activating the signaling pathway, meaning that therefore this complex is divided into: canonical inflammasome-forming NLRs (NLRP1, NLRP2, NLRP3, NLRP6, NLRP7, NLRP9 and NOD-like receptor C4 (NLRC4) and NLRC5), non-canonical inflammasome-forming AIM2 (AIM2 and IFI16) and inflammasome-forming pyrin (MEFV) [70]. The canonical NLRP3 inflammasome is the most studied in the BC context, although several molecular aspects have yet to be investigated. Unlike canonical inflammasome, which involves CASP1 activity, non-canonical inflammasome requires the activation of human caspase-4/5 or murine caspase-11. Moreover, the study of non-canonical signaling is still ongoing and no details are yet known about the affected molecular mechanisms [71].
Breast tumor growth, progression and aggressiveness are promoted by NLRP3 inflammasome activation, pro-inflammatory cytokine secretion and the pyroptotic death of cancer cells. The overexpression of NLRP3, IL-1β, IL-18 and CASP1 is correlated with inflammatory signaling pathways, such as NF-kB, STAT1, STAT3, IL-1β or Wnt/β-catenin, developing an invasive TME, characterized by the release of reactive oxygen species (ROS), reactive nitrogen species (RNS), angiogenic factors, cytokines or chemokines [72].
Aggressive factors from the extracellular environment can cause alterations of cellular structures, leading to the release of pro-inflammatory mediators (cytokines, DAMPs or PAMPs, etc.). The presence of an inflammatory response in the TME is favorable for the initiation of programmed cell death (apoptosis, necroptosis, pyroptosis, etc.) [73].
Pyroptosis, or CASP1-dependent cell death, is characterized by cellular swelling, cell lysis, nuclear condensation and DNA fragmentation. Pyroptosis can have both pro-tumorigenic and anti-tumorigenic roles. The pro-tumorigenic role of pyroptosis is correlated with the NLRP3 and ASC oligomerization mechanism, while the anti-tumorigenic effects of pyroptosis are associated with the activation of the immunogenic signals and immune responses [74].
Following the activation of the NLRP3 inflammasome and the maturation of CASP1, the proteolytic cleavage of GSDMD into N-terminal and C-terminal domains is stimulated. The N-terminal domains of GSDMD attach to the cell membrane and form pores of 10–20 nm in diameter, with the GSDMD-derived pores allowing the influx of calcium or sodium ions and efflux of potassium, thus stimulating the inflammatory process [75].
Over the last years, there has been a special interest in the research field of the contribution of inflammasome complexes to the onset and progression of various diseases, such as obesity, diabetes or cancer. Advanced molecular studies have exposed the dynamic role of inflammasomes and the overlapping with the immune system and inflammatory-associated mechanisms. The connection between inflammasome complexes and the immune system is represented by the secretion and maturation of pro-inflammatory IL-1β, which activates the NF-kB signaling pathway and angiogenic signals, leading to tumor development [76].
The complex relationship among TNBC, inflammasome signaling, the immunosuppressive TME and resistance to chemotherapy requires meticulous investigation. Specifically, breast TME is characterized by the interaction between infiltrated immune cells, inflammatory cells, cytokines, chemokines and growth factors. According to Table 1, two of the criteria for the TNBC classification are represented by the involvement of the immune system and inflammation in the tumorigenic mechanisms, which implies personalized treatment strategies based on inflammatory and immune particularities [77]. In breast malignancies, an immunosuppressive TME is closely correlated with inflammasome signaling. Activation of immune mechanisms (cytokine and chemokine signaling pathways and T-cell receptor or B-cell receptor signaling, etc.) stimulates NOD-like receptor signaling leading to inflammasome activation. In addition, the activation of immune pathways (such as TLRs) stimulates the detection of DAMPs and PAMPs and induces intracellular signals that contribute to the recruitment of ASC adaptor protein and the oligomerization of inflammasome [78,79].
Over the years, clinical trials have revealed that the increased expression of NLRP3 components is associated with unfavorable prognoses and lower survival rates. The up-regulation of NLRP3 inflammasome in TNBC promotes cell migration and invasion, being associated with epithelial–mesenchymal transition (EMT) mechanisms and metastasis through CASP1-dependent mechanisms and the autocrine production of pro-inflammatory IL-1β [80].
Although the function of pyroptosis in the TME of TNBC has not yet been fully elucidated, studies have revealed that pyroptosis plays a dual role (both anti- and pro-tumorigenic). The activation of pyroptosis can thereby generate inflammatory reactions and immune responses that could enhance the sensitivity to immunotherapy [78]. Recent studies have further indicated that the activation of NLRP3 inflammasome stimulates the myeloid cells’ infiltration in breast TME [80]. On the other hand, pro-inflammatory cytokines are released into the TME and promote tumor growth. Additionally, the up-regulation of gasdermin B (GSDMB) is associated with poor prognoses and less sensitivity to HER2-targeted therapy [81].
Considering that inflammasome is implicated in TNBC development and progression, numerous studies have focused on discovering the potential anti-cancer strategies of inflammasome components, such as the identification of possible NLRP3, IL-1β or CASP1 inhibitors (MCC950 or AcYVAD-cmk) or the identification of ncRNA molecules that intervene at the transcriptional level. For example, anakinra, canakinumab or rilonacept are inhibitors of the IL-1 signaling pathway, which contribute to the initiation of apoptosis and blockade of tumor cells’ cycle progression [82]. As mentioned previously, inflammasome opens up new therapeutic perspectives for TNBC, which must be studied thoroughly.

4. Non-Coding RNAs in Oncology

The sequencing of the human genome indicated that approximately 98% of genes are non-functional or “junk DNA” while about 2% of them encode proteins. Moreover, technological advances in sequencing have revealed that the non-coding sequences are copied into RNA molecules that can assist fundamental biological functions, such as regulating gene expression, cell growth and organs’ development, or play an important role in the evolution of a wide variety of human diseases, including cancer, autoimmune disease, metabolic diseases or inflammation. Regarding the implication of ncRNAs in cancer, mutation or aberrations in the promoters of ncRNAs and the dysregulation of the enzymes involved in their biogenesis (Drosha, Dicer) can lead to altered gene expression levels, the normal expression levels of which are crucial for the normal functioning of the human body. Furthermore, up- or down-regulation of ncRNA expression is associated with cancer progression and metastasis [83].
As mentioned previously, ncRNAs can be classified into sncRNAs or lncRNAs, based on their number of nucleotides [15]. Among the most studied sncRNAs are miRNAs, small interfering RNAs (siRNAs), small nucleolar RNAs (snoRNAs), transfer RNA (tRNA)-derived small RNAs (tsRNAs), and PIWI-interacting RNAs (piRNAs). MiRNAs are single-stranded ribonucleic acid molecules that incorporate between 19 and 25 nucleotides and regulate the expression of the target messenger RNAs (mRNAs). In the eukaryotic cells, a primary transcript (pri-miRNA) with multiple secondary structures is formed and is cleaved by two enzymes (Drosha and Dicer) in order to form a single hairpin structure precursor of microRNA (pre-miRNA) and miRNA duplex. Subsequently, these molecules are incorporated into the RNA-induced silencing complex (RISC) and are associated with Argonaute RISC catalytic component 2 (AGO2), leading to the cleavage of the complementary mRNA or suppression of translation (Figure 2) [15].
Among the most studied oncomiRs are miR-373, miR-135b, miR-210, miR-638, miR-301b, miR-663a, etc. (Figure 2). MiR-373 was first identified as a metastasis-promoting miRNA in breast cancer, being involved in tumor-cell proliferation, migration, invasion and metastasis via CD44 targeting [84]. The up-regulation of miR-373 determines the secretion and maturation of caspase-8, which is mainly responsible for the non-canonical activation of the inflammasome complex [85]. MiR-135b is an oncomiR that targets TGF-β in order to enhance cell proliferation and invasion [86], at the same time being involved in the inhibition of CASP1 expression and inflammatory reactions [87]. MiR-210 is also known to play a significant role in breast tumor biological functions, including tumor proliferation and metastasis via regulation of hypoxia and programmed cell death [88]. In the aberrant miR expression, miR-301 is up-regulated in TNBC and promotes tumor growth, cell proliferation, migration, invasion and tamoxifen resistance via the TLR4/NF-kB signaling pathway [89]. Moreover, recent studies have revealed that miR-663a is an inflammation-related miRNA that is frequently deregulated in TNBC, being involved in both apoptotic pathway and inflammasome activity through secretion and maturation of pro-inflammatory cytokines [90,91]. The findings of these studies discovered that miR-638 up-regulation might play a critical role in TNBC progression and inflammasome assembly via modulation of the ATP synthesis mechanism [92,93].
MiRNAs have been described as important regulators in tumor suppression, with miR-217 having been acknowledged as an inhibitor of tumor growth, inflammation and inflammasome, through its regulation of the inflammatory response and the induction of oxidative stress and programmed cell death [94]. Recent studies have indicated that miR-340 functions as a tumor suppressor and cell cycle regulator and that miR-340 could affect pyroptosis through NLRP3 inflammasome and the TLR4/NF-kB signaling pathway, subsequently leading to decreased inflammatory signaling [95,96]. MiR-31 is among the most frequently altered microRNAs in breast malignancies, and the altered expression of miR-31 is associated with the negative regulation of the NF-kB signaling pathway, inflammasome activation, the release of cytokines and cell death [97,98]. Additionally, miR-223 is an anti-inflammatory microRNA that acts as a tumor suppressor in TNBC. The published literature has demonstrated that miRNA-223 expression is associated with NLRP3 inflammasome inhibition and the inhibition of cell cycle progression [99,100].
The main anti-cancer strategy is to activate the programmed death of cancer cells. For example, pyroptosis is a caspase-and-gasdermins-mediated cell death, which can be regulated by drugs or genetic interventions. Recently, special interest has been given to the potential of pyroptosis to suppress TNBC through the recruitment of tumor-suppressed immune cells and the activation of anti-tumor immunity. Studies have indicated that CASP1-induced pyroptosis combined with immune checkpoint inhibitors (ICIs) can enhance anti-tumor immunity in vitro and in vivo [101]. In TNBC conditions, immune cells (CD8+ T cells, NK cells, etc.) stimulate the expression of gasdermins, then coordinate M1 macrophages and T lymphocytes infiltration, in order to induce pyroptosis to stimulate cell sensitivity and to activate tumor clearance [102]. In addition, the activation of pyroptotic pathway by anti-tumor drugs, such as paclitaxel and cisplatin, determines the inhibition of tumor proliferation and metastasis [103].
In the field of oncology, miRNA, tsRNA and piRNA are among the most studied sncRNAs, with numerous studies having focused on the investigation of the mechanisms of miRNAs regarding the molecular and cellular processes involved in cancer conditions. Their results have demonstrated that miRNAs are regulators that guide the expression of both coding and non-coding molecules, such as oncogenes (RAS, MYC, etc.) and tumor suppressors (TP53, BRCA1, etc.). More than that, miRNAs can function as oncogenes, inhibiting the tumor suppressor molecules’ activity, promoting accelerated cancer cell growth and finally encouraging the development of tumor formations. Tumors are subordinate to oncogenic miRNAs (oncomiR) expression for their survival and growth, due to the phenomenon of oncomiR addiction, making them promising potential therapeutic targets in anti-cancer treatment, thereby clearing the way to miRNA-based therapy [104,105]. On the other hand, miRNAs can also act either as tumor suppressors or oncogenes, depending on the progression of associated mechanisms [106]. Additionally, miRNAs can be used as molecular biomarkers because the aberrant expression of several miRNAs has been detected in multiple types of cancer. The expression level of a specific miRNA can be used to indicate the state of the cancer, to differentiate malignant tumors from benign ones and to indicate the type of cancer. Additionally, the molecular signatures of miRNAs outline the prognosis, anticipating the potential for metastasis and the response to therapy [107].

5. MicroRNAs Interplay with TNBC and NLRP3

MiRNA molecules are important regulators that target and interfere with mRNA translation, thus influencing gene expression. The aberrant expression of miRNAs disturbs normal biological mechanisms, such as cell growth, proliferation and migration, programmed cell death, and immune or inflammatory responses. The progression of BC (especially TNBC) is promoted by the abnormal expression of several miRNAs (Table 4). Involved in the development of TNBC are numerous up-regulated (miR-135b, miR-21, miR-301b, miR-221, miR-298, miR-449, miR-302b, miR-663a, miR-638, miR-137, miR-140, miR-155, miR-210, miR-9, miR-181, miR-373, etc.) and down-regulated miRNAs (miR-217, miR-211-5p, miR-185, miR-204-5p, miR-128, miR-340, miR-384, miR-200c, miR-31, miR-200a, miR-141, miR-27b-3p, miR-223, etc.) [108,109].
MiRNAs are the main modulators of gene expression, being involved in numerous normal and pathological mechanisms. Certain miRNAs are directly involved in inflammasome complex modulation into breast TME and also indirectly alter the inflammasome-specific markers by modulating other signaling pathways, such as mTOR, MAPK, cAMP/PKA, AMPK or IFN signaling pathways [146]. Additionally, recent studies have identified the potential of miRNAs to regulate pyroptosis of breast tumor cell through activation or suppression of various molecular networks [147]. Several research groups have carried out relevant studies that have contributed to the understanding of miRNA-mediated regulation of inflammasome assembly and pyroptosis activation in order to develop therapies that target the inflammasome [148].
Wang et al. investigated the influence of miR-200b on NLRP3, ASC, GSDMD, CASP1, IL-1β and IL-18 expression. Their results indicated that up-regulation of miR-200b determined the pyroptosis activation via regulation of the JAZF1/NF-kB signaling pathway. The tumor promoter JAZF zinc finger 1 (JAZF1) is a repressor of TGF-β-activated kinase 1 (TAK1), which is involved in NF-kB signaling cascade. The research group also observed that high expression of JAZF1 constrained the effect of miR-200b on the pyroptosis mechanism [149].
MiR-155-5p is another miRNA molecule that was found to be up-regulated in TNBC. Xu et al. evaluated the capacity of miR-155-5p antagomir to enhance the cytotoxic effects of cetuximab on tumor cells in vitro. The results demonstrated that the combination between miR-155-5p antagomir and cetuximab regulates GSMDE and CASP1 in order to stimulate the pyroptotic cell death and to decrease tumor growth. The in vivo experiments also confirmed that down-regulation of miR-155-5p increased the cytotoxicity of cetuximab, leading to pyroptosis and apoptosis activation in TNBC cells [124].
Bioinformatic analysis of miRNAs’ role in inflammasome-mediated breast tumorigenesis has revealed that certain miRNAs that are associated with the NLR family and consequently influence the prospect of TNBC. For example, NLRP3 is associated with miR-125, miR-146b, miR-200, miR-223-3p, miR-373, miR-520 or miR-548 [150,151], NLRP1 regulation is realized by miR-143-5p, miR-210 [152], and NLRP8 is modulated by miR-181 [153]. The modulation of inflammasome by these miRNAs has effects on TNBC through different signaling pathways, such as NF-κB, TGF-β or IL-6, which are also regulated by the same miRNAs (miR-146b, miR-520, miR-373, etc.) and in normal conditions inhibit TNBC cell growth and proliferation [154,155]. MiRNAs function as master modulators of inflammasome and pyroptosis-related TNBC by regulating the expression of multiple genes and mechanisms. Although some studies have revealed a piece of miRNA-associated networks, it is compulsory to expand the knowledge about inflammasomes and the molecular mechanisms of pyroptosis into the context of TNBC.
MiRNA molecules are involved in the regulation of NLRP3 components, leading to the overexpression of pro-inflammatory markers. MiR-223 is a non-coding molecule and NLRP3 regulator that can bind to the 3′UTR region of NLRP3 mRNA, inhibit the translation mechanism and alter the release and maturation of pro-inflammatory cytokines [156]. Among the numerous other microRNAs involved in NLRP3 regulation is miR-22, which acts as a tumor suppressor. The increased expression of miR-22 is correlated with NLRP3 assembly difficulties, decreased cell proliferation rate and metalloproteinase 2 (MMP2), MMP9, vimentin, and N-cadherin low-expression profiles [157]. The aggressive behavior of cancer is due to the miR-21 activity, which inhibits the NLRP3 assembly by regulating NLRP3 phosphorylation and lysine 63-ubiquitination and alters the normal functions of the innate immune system [158].
Among the microRNA molecules that regulate NLRP3 inflammasome is miR-9. As well, miR-9 has been reported to perform important functions in tumor initiation, inflammation and immune regulation [159]. MiR-9 can suppress the activation of the NLPR3 inflammasome and NF-kB signaling, thereby determining the inhibition of CASP1 expression and the secretion of pro-inflammatory factors (IL-1β and IL-18). The up-regulation of miR-9 is also associated with embryonic lethal abnormal vision-like protein 1 (ELAVL1) regulation, anti-inflammatory mechanisms and the inhibition of pyroptosis cell death [126].
MiRNAs have great potential to become effective therapeutic targets in TNBC treatment, considering the critical functions of microRNAs in TNBC progression and the modulation of NLRP3 components’ expression. Patients diagnosed with TNBC are still lacking an effective therapeutic response to chemotherapy, radiotherapy, surgery and immunotherapy. Therefore, it is necessary to develop new powerful and modern therapies that target the molecular mechanisms involved in the development of triple-negative breast tumors.
MiRNA molecules are decisive for oncogenic processes that have been intensively studied in recent years. For example, numerous studies have focused on the investigation of targeted therapies to silence (for oncogenes) or overexpress (for tumor suppressors) specific miRNAs [111,160]. However, miRNA-based therapeutic applications represent a subject that has not yet been fully elucidated, although these types of treatments have been evaluated in several pre-clinical studies, with the yielded results being deemed satisfactory. MiRNA-derived therapies are based on nucleotide hybridization, so the advantage of using them is that targeted sequences can be easily approached and that they can easily target combinations of RNAs. Although miRNA-based therapy indeed offers many advantages, the method for their delivery still remains a challenging subject [161,162,163].
MiRNA-based therapeutic strategies used in TNBC are either nucleic-acid-based strategies or drug-based strategies. Nucleic-acid-based treatments are divided into two groups: miRNA replacement (miRNA mimics) and anti-miRNA therapy, with the anti-miRNA treatment also being subdivided into two categories depending on the mechanism of action (miRNA antagonists or antagomiRs and miRNA sponges) [129].
MiRNA replacement or miRNA mimic is used in order to restore the defective tumor suppressor miRNA expression and to inhibit breast tumorigenesis progression. The delivery system of miRNA mimics is well received by normal cells [163]. For example, Li and her team explored the mechanism and role of miR-1290 in a radio-treated TNBC context, employing the transfection method for both miR-1290 mimic and miR-1290 inhibitor. Specifically, miR-1290 is a critical mediator that induces tumor proliferation and metastasis, being a potential biomarker for the diagnosis and prognosis of BC. The results of the first experiments carried out indicated that after radiotherapy the levels of NLRP3, ACS and CASP1 did not significantly differ between the radiosensitive and radioresistant MDA-MB-231 cells. Subsequently, the researchers evaluated the profile of miR-1290 in radiosensitive and radioresistant cells and concluded that the expression of miR-1290 was higher in the resistant MDA-MB-231 cells than in those that were sensitive. In the final part of their experiments, the transfection of the miR-1290 mimic and inhibitor demonstrated that the miR-1290 mimic had a positive impact on cell viability and inhibited NLRP3 inflammasome assembly via ASC and CASP1 suppression. Regarding pyroptosis, the miR-1290 inhibitor determined the up-regulation of CASP1, IL-1β and IL-18 by targeting NLRP3. One conclusion of this complex study was that miR-1290 promoted the radioresistance of MDA-MB-231 cells [163]. In addition to this study, which highlights the potential of miR-1290 as a key regulator of pyroptosis, other studies have revealed that many miRNAs are involved in positive regulation of pyroptosis through NLRP3 inflammasome activation, such as miR-21 [164] or miR-155-5p [124].
Anti-miRNA therapy is based on three main approaches: genetic knockout, anti-sense oligonucleotides (meaning antagomiRs) and miRNA sponges. AntagomiRs are modified anti-sense oligonucleotides that consist of ribose residues 2′-O-methylation, cholesterol residues 3′-conjugated and phosphorothioate linkages. AntagomiRs affect miRNA-related pathways by binding and inhibiting the activity of specific oncomiRs involved in TNBC development and progression [165], and the effectiveness of antagomiR therapy varies depending on tissue type and the method of delivery to the targeted tissue. This approach can be used as complementary sequences attached to the miRNAs or in the form of locked nucleic acid oligonucleotides, which contain a bond between the 2′-oxygen and 4′-carbon at each nucleotide, in order to inhibit TNBC progression and metastasis [166]. In 2016, Conde and their research team developed a RNA-triple-helix structure comprised of a miR-205 mimic and antagomiR-221 in order to evaluate a new platform for local anti-cancer therapy using both oncomiR inhibition and tumor suppressor miR replacement approaches. After miR delivery, the obtained data suggested that miR-205 and miR-221 targeted laminin subunit gamma 1 (LAMC1) protein and influenced TNBC by stimulating cell adhesion, proliferation, migration and metastasis, in both in vitro and in vivo conditions; on the other hand, miR-205 up-regulation was a consequence of an increased p53 profile. The concluding remarks of this study revealed that miR-221 and miR-205 inhibition affected E-cadherin, Slug and Snail expression, reducing TNBC cell migration and invasion. This new strategy also determined VEGF down-regulation, suppressing angiogenesis in TNBC contexts [167]. MiR-205 supports breast tumors’ growth and inhibits apoptosis due to its capacity to mediate the assembly mechanism of the NLRP3 complex. The activation of NLRP3 is possible due to the increase in NLRP3, ASC and CASP1 expression and the development of an inflammatory response under the control of miR-205 [168]. The up-regulation of miR-205 has also been reported to decrease steroid receptor co-activator (SRC) expression, which is involved in inflammation through the regulation of pro-inflammatory IL-18 expression [169,170]. On the other hand, miR-221 is a key modulator of breast tumorigenesis and inflammatory responses [171]. The up-regulation of miR-221 has anti-inflammatory effects, and suppresses oxidative stress and programmed cell death in TNBC, due to the decrease in Bcl-2 expression and the suppression of the NLRP3/ASC/CASP1 signaling pathway, as well as inflammatory reaction [114].
MiRNA sponges represent another anti-miRNA strategy, which contain multiple binding sites (between four and sixteen) complementary to the target miRNAs and which have the capacity to inhibit other miRNAs that contain between two and seven specific sequential nucleotides [172]. Advanced studies indicate that miRNA sponges can be transcribed by both RNA polymerase II and III promoters, because their transcripts are stable as a result of capped 5′ and 3′ polyadenylated tails [173]. Additionally, numerous endogenous miRNA sponges that influence the profiles of TNBC-specific biomarkers have been studied. For example, Zhang et al. designed a self-assembled DNA nanosponge embedded with doxorubicin, which mimics the role of miRNA sponge transcripts for the adsorption and clearance of miR-21. The obtained sponge-like delivery system determined an increased efficiency of anti-tumor therapy due to cell apoptosis-specific gene regulation [174]. More interestingly, the potential of such a miR-21 sponge-based strategy can also be used to prevent metastasis through the negative regulation of colony stimulating factor-1 (CSF-1), a biomarker for malignancy and metastasis [175].
On the other hand, drug-based strategies focus on the regulation of miRNAs through the activity of certain intercalating agents (stilbenes, curcumin, flavonoids, histone deacetylases inhibitors, etc.) that bind to miRNA [176]. The regulation of miRNAs by flavonoids has a negative impact on tumor cells’ proliferation, metastasis and EMT, but also modulates important mechanisms, such as angiogenesis, inflammation, inflammasome, immune responses, etc. [177]. The research activities carried out in this field have demonstrated that quercetin can influence the expression of certain oncomiRs, such as miR-21, miR-155 or let-7, leading to BC regression by programmed cell-death stimulation and increasing the sensitivity of tumor cells to chemotherapy [178]. In addition, miR-146a, miR-146b, miR-605 and miR-381 expression is modulated by quercetin, determining tumor growth inhibition and apoptosome assembly [179].
Over the years, the role of miRNA in TNBC progression has been intensively studied, with the results of these studies having highlighted the existence of numerous miRNAs involved in the modulation of the inflammasome complex, such as miR-223-3p, miR-7-5p, miR-22-3p, miR-33, miR-9, miR-155, etc. Their mechanism of action is based on binding 3′UTR of NLRP3 mRNA conserved regions and interfering with translation [180]. For example, Xie et al. studied the influence of miR-33 on NLRP3 inflammasome activation. The obtained results confirmed that miR-33 attenuates mitochondrial oxygen consumption and increases the production of cellular ROS, given that it is responsible for the secretion of IL-1β and the increased expression of NLRP3 and caspase-1 in macrophages [181]. On the contrary, Zhang et al. studied the possibility of inactivating the inflammasome and implicitly the growth of breast tumor cells targeting miR-233. Firstly, the researchers demonstrated the NLRP3 knockdown results in a decrease in breast tumor cell proliferation and migration, due to apoptosis initiation. Secondly, they used miR-233 mimics in order to suppress NLRP3 expression and NLRP3-dependent mechanisms, such as expression levels of ASC, IL-1β, IL-18 and CASP1. At the end of their study, the researchers stated that miR-233 activity inhibits the NLRP3 activation pathway, leading to the suppression of tumor cell growth, migration and invasion [18]. Another study, conducted by Hu et al. highlighted the role of miR-155 in the aggressiveness of TNBC due to the NLRP3 inflammatory pathway. Additionally, miR-155 interference decreased the MDA-MB-231 cell proliferation and ability to form colonies, stimulated the apoptosis of tumor cells and inhibited secretion of pro-inflammatory mediators [182].

6. Conclusions

In summary, this review integrates miRNAs and their functions into the inflammasome-dependent TNBC progression, as miRNAs are key regulators of gene expression and tumorigenesis, being involved in NLRP3 inflammasome assembly and pyroptosis cell death initiation. Due to the heterogeneity and aggressiveness of TNBC caused by genetic and environmental factors, the development and validation of effective targeted strategies has been challenging. Over the past few years, wider attention has been paid to the activation of NLRP3 in oncogenic pathways and to the targeted therapeutic approaches based on miRNAs combined with anti-tumor drugs, in order to increase the survival rate of patients. The results of the original experiments are encouraging, although the elucidation of the molecular manipulation of inflammasome via microRNAs in order to improve TNBC treatment is imperative.

Author Contributions

Conceptualization, L.-R.B. and S.D.; investigation, L.-R.B.; resources, S.D.; writing—original draft preparation, L.-R.B.; writing—review and editing, S.D. and M.C.; visualization, L.-R.B. and S.D.; supervision, M.C.; project administration, S.D. and M.C.; funding acquisition, S.D. and M.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors acknowledge the support from 41PFE/30.12.2021 project, financed by the Ministry of Research, Innovation and Digitalization through Program 1-Development of the National R&D System, Subprogram 1.2. Institutional performance- Financing projects for excellence in RDI.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Khan, N.A.J.; Tirona, M. An updated review of epidemiology, risk factors, and management of male breast cancer. Med. Oncol. 2021, 38, 39–46. [Google Scholar] [CrossRef] [PubMed]
  2. Oskar, S.; Kehm, R.; Terry, M.B. Breast Tissue Composition—Why It Matters and How Can We Measure It More Accurately in Epidemiology Studies. Cancer Epidemiol. Biomarkers Prev. 2021, 30, 590–592. [Google Scholar] [CrossRef] [PubMed]
  3. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  4. Wang, L. Early Diagnosis of Breast Cancer. Sensors 2017, 17, 1572. [Google Scholar] [CrossRef] [PubMed]
  5. Teichgraeber, D.C.; Guirguis, M.S.; Whitman, G.J. Breast Cancer Staging: Updates in the AJCC Cancer Staging Manual, 8th Edition, and Current Challenges for Radiologists, From the AJR Special Series on Cancer Staging. Am. J. Roentgenol. 2021, 217, 278–290. [Google Scholar] [CrossRef]
  6. Kalli, S.; Semine, A.; Cohen, S.; Naber, S.P.; Makim, S.S.; Bahl, M. American Joint Committee on Cancer’s Staging System for Breast Cancer, Eighth Edition: What the Radiologist Needs to Know. Radiographics 2018, 38, 1921–1933. [Google Scholar] [CrossRef]
  7. Tsang, J.Y.S.; Tse, G.M. Molecular Classification of Breast Cancer. Adv. Anat. Pathol. 2019, 27, 27–35. [Google Scholar] [CrossRef]
  8. Kumar, P.; Aggarwal, R. An overview of triple-negative breast cancer. Arch. Gynecol. Obstet. 2016, 293, 247–269. [Google Scholar] [CrossRef]
  9. Jackson, H.W.; Fischer, J.R.; Zanotelli, V.R.T.; Ali, H.R.; Mechera, R.; Soysal, S.D.; Moch, H.; Muenst, S.; Varga, Z.; Weber, W.P.; et al. The single-cell pathology landscape of breast cancer. Nature 2020, 578, 615–620. [Google Scholar] [CrossRef]
  10. Yeo, S.K.; Guan, J.-L. Breast Cancer: Multiple Subtypes within a Tumor? Trends Cancer 2017, 3, 753–760. [Google Scholar] [CrossRef]
  11. Sukumar, J.; Gast, K.; Quiroga, D.; Lustberg, M.; Williams, N. Triple-negative breast cancer: Promising prognostic biomarkers currently in development. Expert Rev. Anticancer. Ther. 2020, 21, 135–148. [Google Scholar] [CrossRef] [PubMed]
  12. Saponaro, C.; Scarpi, E.; Sonnessa, M.; Cioffi, A.; Buccino, F.; Giotta, F.; Pastena, M.I.; Zito, F.A.; Mangia, A. Prognostic Value of NLRP3 Inflammasome and TLR4 Expression in Breast Cancer Patients. Front. Oncol. 2021, 11, 705331. [Google Scholar] [CrossRef] [PubMed]
  13. Faria, S.S.; Costantini, S.; de Lima, V.C.C.; de Andrade, V.P.; Rialland, M.; Cedric, R.; Budillon, A.; Magalhaes, K.G. NLRP3 inflammasome-mediated cytokine production and pyroptosis cell death in breast cancer. J. Biomed. Sci. 2021, 28, 26. [Google Scholar] [CrossRef] [PubMed]
  14. Tezcan, G.; Martynova, E.V.; Gilazieva, Z.E.; McIntyre, A.; Rizvanov, A.A.; Khaiboullina, S.F. MicroRNA Post-transcriptional Regulation of the NLRP3 Inflammasome in Immunopathologies. Front. Pharmacol. 2019, 10, 451. [Google Scholar] [CrossRef] [PubMed]
  15. Liu, J.; Zhao, G.; Liu, X.-L.; Zhang, G.; Zhao, S.-Q.; Zhang, S.-L.; Luo, L.-H.; Yin, D.-C.; Zhang, C.-Y. Progress of non-coding RNAs in triple-negative breast cancer. Life Sci. 2021, 272, 119238. [Google Scholar] [CrossRef]
  16. Ding, X.-F. Roles of miRNA and IncRNA in triple-negative breast cancer. J. Zhejiang Univ. Sci. B 2020, 21, 673–689. [Google Scholar] [CrossRef]
  17. Zamani, P.; Oskuee, R.K.; Atkin, S.L.; Navashenaq, J.G.; Sahebkar, A. MicroRNAs as important regulators of the NLRP3 inflammasome. Prog. Biophys. Mol. Biol. 2019, 150, 50–64. [Google Scholar] [CrossRef]
  18. Zhang, L.; Li, H.; Zang, Y.; Wang, F. NLRP3 inflammasome inactivation driven by miR-223-3p reduces tumor growth and increases anticancer immunity in breast cancer. Mol. Med. Rep. 2019, 19, 2180–2188. [Google Scholar] [CrossRef]
  19. Lyons, T.G. Targeted Therapies for Triple-Negative Breast Cancer. Curr. Treat. Options Oncol. 2019, 20, 82. [Google Scholar] [CrossRef]
  20. Lehmann, B.D.; Bauer, J.A.; Chen, X.; Sanders, M.E.; Chakravarthy, A.B.; Shyr, Y.; Pietenpol, J.A. Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J. Clin. Investig. 2011, 121, 2750–2767. [Google Scholar] [CrossRef] [Green Version]
  21. Burstein, M.D.; Tsimelzon, A.; Poage, G.M.; Covington, K.R.; Contreras, A.; Fuqua, S.A.; Savage, M.I.; Osborne, C.K.; Hilsenbeck, S.G.; Chang, J.C.; et al. Comprehensive genomic analysis identifies novel subtypes and targets of triple-negative breast cancer. Clin. Cancer Res. 2015, 21, 1688–1698. [Google Scholar] [CrossRef] [PubMed]
  22. Jézéquel, P.; Loussouarn, D.; Guérin-Charbonnel, C.; Campion, L.; Vanier, A.; Gouraud, W.; Lasla, H.; Guette, C.; Valo, I.; Verrièle, V.; et al. Gene-expression molecular subtyping of triple-negative breast cancer tumours: Importance of immune response. Breast Cancer Res. 2015, 17, 43. [Google Scholar] [CrossRef] [PubMed]
  23. Hubalek, M.; Czech, T.; Müller, H. Biological Subtypes of Triple-Negative Breast Cancer. Breast Care 2017, 12, 8–14. [Google Scholar] [CrossRef] [PubMed]
  24. Yin, L.; Duan, J.-J.; Bian, X.-W.; Yu, S.-C. Triple-negative breast cancer molecular subtyping and treatment progress. Breast Cancer Res. 2020, 22, 61. [Google Scholar] [CrossRef] [PubMed]
  25. Abramson, V.G.; Lehmann, B.D.; Ballinger, T.J.; Pietenpol, J.A. Subtyping of triple-negative breast cancer: Implications for therapy. Cancer 2015, 121, 8–16. [Google Scholar] [CrossRef]
  26. Bareche, Y.; Buisseret, L.; Gruosso, T.; Girard, E.; Venet, D.; Dupont, F.; Desmedt, C.; Larsimont, D.; Park, M.; Rothé, F.; et al. Unraveling Triple-Negative Breast Cancer Tumor Microenvironment Heterogeneity: Towards an Optimized Treatment Approach. J. Natl. Cancer Inst. 2020, 112, 708–719. [Google Scholar] [CrossRef]
  27. Uscanga-Peralesa, G.I.; Santuario-Facio, S.K.; Ortiz-López, R. Triple negative breast cancer: Deciphering the biology and heterogeneity. Med. Univ. 2016, 18, 105–114. [Google Scholar] [CrossRef]
  28. Gibson, G.R.; Qian, D.; Ku, J.K.; Lai, L.L. Metaplastic Breast Cancer: Clinical Features and Outcomes. Am. Surg. 2005, 71, 725–730. [Google Scholar] [CrossRef]
  29. Hayes, M.J.; Thomas, D.; Emmons, A.; Giordano, T.J.; Kleer, C.G. Genetic Changes of Wnt Pathway Genes Are Common Events in Metaplastic Carcinomas of the Breast. Clin. Cancer Res. 2008, 14, 4038–4044. [Google Scholar] [CrossRef]
  30. Gerratana, L.; Basile, D.; Buono, G.; De Placido, S.; Giuliano, M.; Minichillo, S.; Coinu, A.; Martorana, F.; De Santo, I.; Del Mastro, L.; et al. Androgen receptor in triple negative breast cancer: A potential target for the targetless subtype. Cancer Treat. Rev. 2018, 68, 102–110. [Google Scholar] [CrossRef]
  31. Ahn, S.G.; Kim, S.J.; Kim, C.; Jeong, J. Molecular Classification of Triple-Negative Breast Cancer. J. Breast Cancer 2016, 19, 223–230. [Google Scholar] [CrossRef] [PubMed]
  32. Jiang, Y.-Z.; Ma, D.; Suo, C.; Shi, J.; Xue, M.; Hu, X.; Xiao, Y.; Yu, K.-D.; Liu, Y.-R.; Yu, Y.; et al. Genomic and Transcriptomic Landscape of Triple-Negative Breast Cancers: Subtypes and Treatment Strategies. Cancer Cell 2019, 35, 428–440.e5. [Google Scholar] [CrossRef]
  33. Mahmoud, R.; Ordonez-Moran, P.; Allegrucci, C. Challenges for Triple Negative Breast Cancer Treatment: Defeating Heterogeneity and Cancer Stemness. Cancers 2022, 14, 4280. [Google Scholar] [CrossRef]
  34. Molchanov, O.; Maistrenko, D.; Granov, D.; Vasina, L.; Popova, A.; Vasilevskaya, I.; Mikolaichuk, O.; Shemchuk, O.; Protas, A.; Sharoyko, V.; et al. Biomarkers and potential targets for immune and cellular therapy in triple negative breast cancer. Cell. Ther. Transplant. 2022, 11, 16–30. [Google Scholar] [CrossRef]
  35. Lehmann, B.D.; Pietenpol, J.A. Identification and use of biomarkers in treatment strategies for triple-negative breast cancer subtypes. J. Pathol. 2014, 232, 142–150. [Google Scholar] [CrossRef]
  36. Li, Y.; Zhang, H.; Merkher, Y.; Chen, L.; Liu, N.; Leonov, S.; Chen, Y. Recent advances in therapeutic strategies for triple-negative breast cancer. J. Hematol. Oncol. 2022, 29, 121. [Google Scholar] [CrossRef]
  37. Ghafouri, S.R.; Guvvala, S.; Jones, C.; Philipovskiy, A. Recently approved treatment options for patients with metastatic triple-negative and HER2-neu-positive breast cancer. J. Investig. Med. 2022, 70, 1329–1341. [Google Scholar] [CrossRef]
  38. Xiao, Y.; Ma, D.; Zhao, S.; Suo, C.; Shi, J.; Xue, M.-Z.; Ruan, M.; Wang, H.; Zhao, J.; Li, Q.; et al. Multi-Omics Profiling Reveals Distinct Microenvironment Characterization and Suggests Immune Escape Mechanisms of Triple-Negative Breast Cancer. Clin. Cancer Res. 2019, 25, 5002–5014. [Google Scholar] [CrossRef] [PubMed]
  39. Heusinkveld, M.; van der Burg, S.H. Identification and manipulation of tumor associated macrophages in human cancers. J. Transl. Med. 2011, 9, 216. [Google Scholar] [CrossRef] [PubMed]
  40. Luo, Y.; Zhou, H.; Krueger, J.; Kaplan, C.; Lee, S.H.; Dolman, C.; Markowitz, D.; Wu, W.; Liu, C.; Reisfeld, R.A.; et al. Targeting tumor-associated macrophages as a novel strategy against breast cancer. J. Clin. Investig. 2006, 116, 2132–2141. [Google Scholar] [CrossRef] [Green Version]
  41. Prasmickaite, L.; Tenstad, E.M.; Pettersen, S.; Jabeen, S.; Egeland, E.V.; Nord, S.; Pandya, A.; Haugen, M.H.; Kristensen, V.N.; Børresen-Dale, A.; et al. Basal-like breast cancer engages tumor-supportive macrophages via secreted factors induced by extracellular S100A4. Mol. Oncol. 2018, 12, 1540–1558. [Google Scholar] [CrossRef] [PubMed]
  42. Pe, K.C.S.; Saetung, R.; Yodsurang, V.; Chaotham, C.; Suppipat, K.; Chanvorachote, P.; Tawinwung, S. Triple-negative breast cancer influences a mixed M1/M2 macrophage phenotype associated with tumor aggressiveness. PLoS ONE 2022, 17, e0273044. [Google Scholar] [CrossRef] [PubMed]
  43. Mehta, A.K.; Kadel, S.; Townsend, M.G.; Oliwa, M.; Guerriero, J.L. Macrophage Biology and Mechanisms of Immune Suppression in Breast Cancer. Front. Immunol. 2021, 12, 643771. [Google Scholar] [CrossRef] [PubMed]
  44. Lee, K.Y. M1 and M2 polarization of macrophages: A mini-review. Med. Biol. Sci. Eng. 2019, 2, 1–5. [Google Scholar] [CrossRef]
  45. Martinon, F.; Burns, K.; Tschopp, J. The Inflammasome: A molecular platform triggering activation of inflammatory caspases and processing of proIL-β. Mol. Cell 2002, 10, 417–426. [Google Scholar] [CrossRef]
  46. Ershaid, N.; Sharon, Y.; Doron, H.; Raz, Y.; Shani, O.; Cohen, N.; Monteran, L.; Leider-Trejo, L.; Ben-Shmuel, A.; Yassin, M.; et al. NLRP3 inflammasome in fibroblasts links tissue damage with inflammation in breast cancer progression and metastasis. Nat. Commun. 2019, 10, 4375. [Google Scholar] [CrossRef]
  47. George, M.A.; Lustberg, M.B.; Orchard, T.S. Psychoneurological symptom cluster in breast cancer: The role of inflammation and diet. Breast Cancer Res. Treat. 2020, 184, 1–9. [Google Scholar] [CrossRef]
  48. Sonnessa, M.; Cioffi, A.; Brunetti, O.; Silvestris, N.; Zito, F.A.; Saponaro, C.; Mangia, A. NLRP3 Inflammasome From Bench to Bedside: New Perspectives for Triple Negative Breast Cancer. Front. Oncol. 2020, 10, 1587. [Google Scholar] [CrossRef]
  49. Medina, M.A.; Oza, G.; Sharma, A.; Arriaga, L.G.; Hernandez, J.M.; Rotello, V.M.; Ramirez, J.T. Triple-Negative Breast Cancer: A Review of Conventional and Advanced Therapeutic Strategies. Int. J. Environ. Res. Public Health 2020, 17, 2078. [Google Scholar] [CrossRef]
  50. Pauls, M.; Chia, S.; LeVasseur, N. Current and New Novel Combination Treatments for Metastatic Triple-Negative Breast Cancer. Curr. Oncol. 2022, 29, 4748–4767. [Google Scholar] [CrossRef]
  51. Carey, L.A.; Dees, E.C.; Sawyer, L.; Gatti, L.; Moore, D.T.; Collichio, F.; Ollila, D.W.; Sartor, C.I.; Graham, M.L.; Perou, C.M. The Triple Negative Paradox: Primary Tumor Chemosensitivity of Breast Cancer Subtypes. Clin. Cancer Res. 2007, 13, 2329–2334. [Google Scholar] [CrossRef] [PubMed]
  52. Martín, M.; Ruiz, A.; Ruiz Borrego, M.; Barnadas, A.; González, S.; Calvo, L.; Vila, M.M.; Antón, A.; Rodríguez-Lescure, A.; Seguí-Palmer, M.A.; et al. Fluorouracil, Doxorubicin, and Cyclophosphamide (FAC) Versus FAC Followed by Weekly Paclitaxel As Adjuvant Therapy for High-Risk, Node-Negative Breast Cancer: Results From the GEICAM/2003-02 Study. J. Clin. Oncol. 2013, 31, 2593–2599. [Google Scholar] [CrossRef] [PubMed]
  53. Park, J.H.; Ahn, J.-H.; Kim, S.-B. How shall we treat early triple-negative breast cancer (TNBC): From the current standard to upcoming immuno-molecular strategies. ESMO Open 2018, 3, e000357. [Google Scholar] [CrossRef] [PubMed]
  54. Bou Zerdan, M.; Ghorayeb, T.; Saliba, F.; Allam, S.; Bou Zerdan, M.; Yaghi, M.; Bilani, N.; Jaafar, R.; Nahleh, Z. Triple Negative Breast Cancer: Updates on Classification and Treatment in 2021. Cancers 2022, 14, 1253. [Google Scholar] [CrossRef]
  55. Emens, L.A. Breast Cancer Immunotherapy: Facts and Hopes. Clin. Cancer Res. 2018, 24, 511–520. [Google Scholar] [CrossRef] [PubMed]
  56. Pandy, J.G.P.; Balolong-Garcia, J.C.; Cruz-Ordinario, M.V.B.; Que, F.V.F. Triple negative breast cancer and platinum-based systemic treatment: A meta-analysis and systematic review. BMC Cancer 2019, 19, 1253. [Google Scholar] [CrossRef]
  57. Liu, M.; Mo, Q.-G.; Wei, C.-Y.; Qin, Q.-H.; Huang, Z.; He, J. Platinum-based chemotherapy in triple-negative breast cancer: A meta-analysis. Oncol. Lett. 2012, 5, 983–991. [Google Scholar] [CrossRef]
  58. Bian, L.; Yu, P.; Wen, J.; Na Li, N.; Huang, W.; Xie, X.; Ye, F. Survival benefit of platinum-based regimen in early stage triple negative breast cancer: A meta-analysis of randomized controlled trials. npj Breast Cancer 2021, 7, 157. [Google Scholar] [CrossRef]
  59. Thu, K.L.; Soria-Bretones, I.; Mak, T.W.; Cescon, D.W. Targeting the cell cycle in breast cancer: Towards the next phase. Cell Cycle 2018, 17, 1871–1885. [Google Scholar] [CrossRef]
  60. Sun, H.; Zhang, D.; Yao, Z.; Lin, X.; Liu, J.; Gu, Q.; Dong, X.; Liu, F.; Wang, Y.; Yao, N.; et al. Anti-angiogenic treatment promotes triple-negative breast cancer invasion via vasculogenic mimicry. Cancer Biol. Ther. 2017, 18, 205–213. [Google Scholar] [CrossRef] [Green Version]
  61. Zhu, K.; Wu, Y.; He, P.; Fan, Y.; Zhong, X.; Zheng, H.; Luo, T. PI3K/AKT/mTOR-Targeted Therapy for Breast Cancer. Cells 2022, 11, 2508. [Google Scholar] [CrossRef] [PubMed]
  62. Costa, R.L.B.; Han, H.S.; Gradishar, W.J. Targeting the PI3K/AKT/mTOR pathway in triple-negative breast cancer: A review. Breast Cancer Res. Treat. 2018, 169, 397–406. [Google Scholar] [CrossRef]
  63. Khan, M.A.; Jain, V.K.; Rizwanullah, M.; Ahmad, J.; Jain, K. PI3K/AKT/mTOR pathway inhibitors in triple-negative breast cancer: A review on drug discovery and future challenges. Drug Discov. Today 2019, 24, 2181–2191. [Google Scholar] [CrossRef]
  64. Singh, D.D.; Parveen, A.; Yadav, D.K. Role of PARP in TNBC: Mechanism of Inhibition, Clinical Applications, and Resistance. Biomedicines 2021, 9, 1512. [Google Scholar] [CrossRef]
  65. Maennling, A.E.; Tur, M.K.; Niebert, M.; Klockenbring, T.; Zeppernick, F.; Gattenlöhner, S.; Meinhold-Heerlein, I.; Hussain, A.F. Molecular Targeting Therapy against EGFR Family in Breast Cancer: Progress and Future Potentials. Cancers 2019, 11, 1826. [Google Scholar] [CrossRef]
  66. Liu, C.-Y.; Lau, K.-Y.; Hsu, C.-C.; Chen, J.-L.; Lee, C.-H.; Huang, T.-T.; Chen, Y.-T.; Huang, C.-T.; Lin, P.-H.; Tseng, L.-M. Combination of palbociclib with enzalutamide shows in vitro activity in RB proficient and androgen receptor positive triple negative breast cancer cells. PLoS ONE 2017, 12, e0189007. [Google Scholar] [CrossRef] [PubMed]
  67. Kelley, N.; Jeltema, D.; Duan, Y.; He, Y. The NLRP3 Inflammasome: An Overview of Mechanisms of Activation and Regulation. Int. J. Mol. Sci. 2019, 20, 3328. [Google Scholar] [CrossRef] [PubMed]
  68. Kumar, S.; Jain, S. Immune signalling by supramolecular assemblies. Immunology 2018, 155, 435–445. [Google Scholar] [CrossRef] [PubMed]
  69. Malik, A.; Kanneganti, T.-D. Inflammasome activation and assembly at a glance. J. Cell Sci. 2017, 130, 3955–3963. [Google Scholar] [CrossRef]
  70. Komada, T.; Muruve, D.A. The role of inflammasomes in kidney disease. Nat. Rev. Nephrol. 2019, 15, 501–520. [Google Scholar] [CrossRef]
  71. Yi, Y.-S. Regulatory Roles of Caspase-11 Non-Canonical Inflammasome in Inflammatory Liver Diseases. Int. J. Mol. Sci. 2022, 23, 4986. [Google Scholar] [CrossRef]
  72. Kantono, M.; Guo, B. Inflammasomes and Cancer: The Dynamic Role of the Inflammasome in Tumor Development. Front. Immunol. 2017, 8, 1132. [Google Scholar] [CrossRef] [PubMed]
  73. Montico, B.; Nigro, A.; Casolaro, V.; Col, J.D. Immunogenic Apoptosis as a Novel Tool for Anticancer Vaccine Development. Int. J. Mol. Sci. 2018, 19, 594. [Google Scholar] [CrossRef] [PubMed]
  74. Yu, P.; Zhang, X.; Liu, N.; Tang, L.; Peng, C.; Chen, X. Pyroptosis: Mechanisms and diseases. Signal Transduct. Target. Ther. 2021, 6, 128. [Google Scholar] [CrossRef]
  75. Wang, Q.; Wang, Y.; Ding, J.; Wang, C.; Zhou, X.; Gao, W.; Huang, H.; Shao, F.; Liu, Z. A bioorthogonal system reveals antitumour immune function of pyroptosis. Nature 2020, 579, 421–426. [Google Scholar] [CrossRef]
  76. Poli, G.; Fabi, C.; Bellet, M.M.; Costantini, C.; Nunziangeli, L.; Romani, L.; Brancorsini, S. Epigenetic Mechanisms of Inflammasome Regulation. Int. J. Mol. Sci. 2020, 21, 5758. [Google Scholar] [CrossRef]
  77. Rodríguez-Bautista, R.; Caro-Sánchez, C.H.; Cabrera-Galeana, P.; Alanis-Funes, G.J.; Gutierrez-Millán, E.; Ávila-Ríos, S.; Matías-Florentino, M.; Reyes-Terán, G.; Díaz-Chávez, J.; Villarreal-Garza, C.; et al. Immune Milieu and Genomic Alterations Set the Triple-Negative Breast Cancer Immunomodulatory Subtype Tumor Behavior. Cancers 2021, 13, 6256. [Google Scholar] [CrossRef] [PubMed]
  78. Luo, L.; Wei, Q.; Xu, C.; Dong, M.; Zhao, W. Immune landscape and risk prediction based on pyroptosis-related molecular subtypes in triple-negative breast cancer. Front. Immunol. 2022, 13, 933703. [Google Scholar] [CrossRef]
  79. Deets, K.A.; Vance, R.E. Inflammasomes and adaptive immune responses. Nat. Immunol. 2021, 22, 412–422. [Google Scholar] [CrossRef]
  80. Wang, Y.; Zhang, H.; Xu, Y.; Peng, T.; Meng, X.; Zou, F. NLRP3 induces the autocrine secretion of IL-1β to promote epithelial–mesenchymal transition and metastasis in breast cancer. Biochem. Biophys. Res. Commun. 2021, 560, 72–79. [Google Scholar] [CrossRef]
  81. Hamarsheh, S.; Zeiser, R. NLRP3 Inflammasome Activation in Cancer: A Double-Edged Sword. Front. Immunol. 2020, 11, 1444. [Google Scholar] [CrossRef] [PubMed]
  82. Huang, Y.; Wang, H.; Hao, Y.; Lin, H.; Dong, M.; Ye, J.; Song, L.; Wang, Y.; Li, Q.; Shan, B.; et al. Myeloid PTEN promotes chemotherapy-induced NLRP3-inflammasome activation and antitumour immunity. Nat. Cell Biol. 2020, 22, 716–727. [Google Scholar] [CrossRef] [PubMed]
  83. Slack, F.J.; Chinnaiyan, A.M. The Role of Non-coding RNAs in Oncology. Cell 2019, 179, 1033–1055. [Google Scholar] [CrossRef]
  84. Bakr, N.M.; Mahmoud, M.S.; Nabil, R.; Boushnak, H.; Swellam, M. Impact of circulating miRNA-373 on breast cancer diagnosis through targeting VEGF and cyclin D1 genes. J. Genet. Eng. Biotechnol. 2021, 19, 84. [Google Scholar] [CrossRef] [PubMed]
  85. Gurung, P.; Anand, P.K.; Malireddi, R.K.; Vande Walle, L.; Van Opdenbosch, N.; Dillon, C.P.; Weinlich, R.; Green, D.; Lamkanfi, M.; Kanneganti, T.-D. FADD and Caspase-8 Mediate Priming and Activation of the Canonical and Noncanonical Nlrp3 Inflammasomes. J. Immunol. 2014, 192, 1835–1846. [Google Scholar] [CrossRef]
  86. 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]
  87. Halappanavar, S.; Nikota, J.; Wu, D.; Williams, A.; Yauk, C.L.; Stampfli, M. IL-1 Receptor Regulates microRNA-135b Expression in a Negative Feedback Mechanism during Cigarette Smoke–Induced Inflammation. J. Immunol. 2013, 190, 3679–3686. [Google Scholar] [CrossRef]
  88. Marwarha, G.; Røsand, Ø.; Scrimgeour, N.; Slagsvold, K.H.; Høydal, M.A. miR-210 Regulates Apoptotic Cell Death during Cellular Hypoxia and Reoxygenation in a Diametrically Opposite Manner. Biomedicines 2021, 10, 42. [Google Scholar] [CrossRef]
  89. Qi, Y.; Wang, X.; Kong, X.; Zhai, J.; Fang, Y.; Guan, X.; Wang, J. Expression signatures and roles of microRNAs in inflammatory breast cancer. Cancer Cell Int. 2019, 19, 23. [Google Scholar] [CrossRef]
  90. Michaille, J.-J.; Piurowski, V.; Rigot, B.; Kelani, H.; Fortman, E.C.; Tili, E. MiR-663, a MicroRNA Linked with Inflammation and Cancer That Is under the Influence of Resveratrol. Medicines 2018, 5, 74. [Google Scholar] [CrossRef] [Green Version]
  91. Carden, T.; Singh, B.; Mooga, V.; Bajpai, P.; Singh, K.K. Epigenetic modification of miR-663 controls mitochondria-to-nucleus retrograde signaling and tumor progression. J. Biol. Chem. 2017, 292, 20694–20706. [Google Scholar] [CrossRef] [PubMed]
  92. Wang, B.; Wang, K.; Yu, J.; Hao, X.-M.; Liu, Y.-L.; Xing, A.-Y. miR-638 Serves as a Biomarker of 5-Fluorouracil Sensitivity to Neoadjuvant Chemotherapy in Breast Cancer. J. Breast Cancer 2022, 25, 193–206. [Google Scholar] [CrossRef] [PubMed]
  93. Bandiera, S.; Rüberg, S.; Girard, M.; Cagnard, N.; Hanein, S.; Chrétien, D.; Munnich, A.; Lyonnet, S.; Henrion-Caude, A. Nuclear Outsourcing of RNA Interference Components to Human Mitochondria. PLoS ONE 2011, 6, e20746. [Google Scholar] [CrossRef] [PubMed]
  94. Li, W.; Yang, C.; Li, J.; Li, X.; Zhou, P. MicroRNA-217 aggravates breast cancer through activation of NF1-mediated HSF1/ATG7 axis and c-Jun/ATF3/MMP13 axis. Hum. Cell 2023, 36, 377–392. [Google Scholar] [CrossRef] [PubMed]
  95. Shi, Z.; Li, Y.; Qian, X.; Hu, Y.; Liu, J.; Zhang, S.; Zhang, J. MiR-340 Inhibits Triple-Negative Breast Cancer Progression by Reversing EZH2 Mediated miRNAs Dysregulated Expressions. J. Cancer 2017, 8, 3037–3048. [Google Scholar] [CrossRef] [PubMed]
  96. Zimta, A.-A.; Cenariu, D.; Irimie, A.; Magdo, L.; Nabavi, S.M.; Atanasov, A.G.; Berindan-Neagoe, I. The Role of Nrf2 Activity in Cancer Development and Progression. Cancers 2019, 11, 1755. [Google Scholar] [CrossRef]
  97. Yu, T.; Ma, P.; Wu, D.; Shu, Y.; Gao, W. Functions and mechanisms of microRNA-31 in human cancers. Biomed. Pharmacother. 2018, 108, 1162–1169. [Google Scholar] [CrossRef]
  98. Hashemi, Z.S.; Moghadam, M.F.; Farokhimanesh, S.; Rajabibazl, M.; Sadroddiny, E. Inhibition of breast cancer metastasis by co-transfection of miR-31/193b-mimics. Iran J Basic Med. Sci 2018, 21, 427–433. [Google Scholar] [CrossRef]
  99. Chen, L.; Zhu, X.; Han, B.; Ji, L.; Yao, L.; Wang, Z. High Expression of microRNA-223 Indicates a Good Prognosis in Triple-Negative Breast Cancer. Front. Oncol. 2021, 11, 630432. [Google Scholar] [CrossRef]
  100. Favero, A.; Segatto, I.; Perin, T.; Belletti, B. The many facets of miR -223 in cancer: Oncosuppressor, oncogenic driver, therapeutic target, and biomarker of response. Wiley Interdiscip. Rev. RNA 2021, 12, e1659. [Google Scholar] [CrossRef]
  101. Socinski, M.A.; Jotte, R.M.; Cappuzzo, F.; Orlandi, F.; Stroyakovskiy, D.; Nogami, N.; Rodríguez-Abreu, D.; Moro-Sibilot, D.; Thomas, C.A.; Barlesi, F.; et al. Atezolizumab for First-Line Treatment of Metastatic Nonsquamous NSCLC. N. Engl. J. Med. 2018, 378, 2288–2301. [Google Scholar] [CrossRef] [PubMed]
  102. Tang, R.; Xu, J.; Zhang, B.; Liu, J.; Liang, C.; Hua, J.; Meng, Q.; Yu, X.; Shi, S. Ferroptosis, necroptosis, and pyroptosis in anticancer immunity. J. Hematol. Oncol. 2020, 13, 110. [Google Scholar] [CrossRef] [PubMed]
  103. Wang, Y.; Gao, W.; Shi, X.; Ding, J.; Liu, W.; He, H.; Wang, K.; Shao, F. Chemotherapy drugs induce pyroptosis through caspase-3 cleavage of a gasdermin. Nature 2017, 547, 99–103. [Google Scholar] [CrossRef] [PubMed]
  104. Rupaimoole, R.; Slack, F.J. MicroRNA therapeutics: Towards a new era for the management of cancer and other diseases. Nat. Rev. Drug Discov. 2017, 16, 203–222. [Google Scholar] [CrossRef] [PubMed]
  105. Alyami, N.M. MicroRNAs Role in Breast Cancer: Theranostic Application in Saudi Arabia. Front. Oncol. 2021, 11, 717759. [Google Scholar] [CrossRef]
  106. Otmani, K.; Lewalle, P. Tumor Suppressor miRNA in Cancer Cells and the Tumor Microenvironment: Mechanism of Deregulation and Clinical Implications. Front. Oncol. 2021, 11, 708765. [Google Scholar] [CrossRef]
  107. Mehrgou, A.; Akouchekian, M. Therapeutic impacts of microRNAs in breast cancer by their roles in regulating processes involved in this disease. J. Res. Med. Sci. 2017, 22, 130. [Google Scholar] [CrossRef]
  108. Wu, J.; Sun, Z.; Sun, H.; Li, Y. MicroRNA-27a promotes tumorigenesis via targeting AKT in triple negative breast cancer. Mol. Med. Rep. 2018, 17, 562–570. [Google Scholar] [CrossRef]
  109. Wang, Y.; Zhang, Z.; Wang, J. MicroRNA-384 inhibits the progression of breast cancer by targeting ACVR1. Oncol. Rep. 2018, 39, 2563–2574. [Google Scholar] [CrossRef]
  110. Dong, G.; Liang, X.; Wang, D.; Gao, H.; Wang, L.; Wang, L.; Liu, J.; Du, Z. High expression of miR-21 in triple-negative breast cancers was correlated with a poor prognosis and promoted tumor cell in vitro proliferation. Med. Oncol. 2014, 31, 57. [Google Scholar] [CrossRef]
  111. Cheng, H.-Y.; Hsieh, C.-H.; Lin, P.-H.; Chen, Y.-T.; Hsu, D.S.-S.; Tai, S.-K.; Chu, P.-Y.; Yang, M.-H. Snail-regulated exosomal microRNA-21 suppresses NLRP3 inflammasome activity to enhance cisplatin resistance. J. Immunother. Cancer 2022, 10, e004832. [Google Scholar] [CrossRef]
  112. Song, H.; Li, D.; Wu, T.; Xie, D.; Hua, K.; Hu, J.; Deng, X.; Ji, C.; Deng, Y.; Fang, L. MicroRNA-301b promotes cell proliferation and apoptosis resistance in triple-negative breast cancer by targeting CYLD. BMB Rep. 2018, 51, 602–607. [Google Scholar] [CrossRef] [PubMed]
  113. Stinson, S.; Lackner, M.R.; Adai, A.T.; Yu, N.; Kim, H.-J.; O’Brien, C.; Spoerke, J.; Jhunjhunwala, S.; Boyd, Z.; Januario, T.; et al. miR-221/222 Targeting of Trichorhinophalangeal 1 (TRPS1) Promotes Epithelial-to-Mesenchymal Transition in Breast Cancer. Sci. Signal. 2011, 4, 5. [Google Scholar] [CrossRef] [PubMed]
  114. Niedzielski, M.; Broncel, M.; Gorzelak-Pabiś, P.; Woźniak, E. New possible pharmacological targets for statins and ezetimibe. Biomed. Pharmacother. 2020, 129, 110388. [Google Scholar] [CrossRef] [PubMed]
  115. Garrido-Cano, I.; Pattanayak, B.; Adam-Artigues, A.; Lameirinhas, A.; Torres-Ruiz, S.; Tormo, E.; Cervera, R.; Eroles, P. MicroRNAs as a clue to overcome breast cancer treatment resistance. Cancer Metastasis Rev. 2022, 41, 77–105. [Google Scholar] [CrossRef]
  116. Vlacil, A.-K.; Vollmeister, E.; Bertrams, W.; Schoesser, F.; Oberoi, R.; Schuett, J.; Schuett, H.; Huehn, S.; Bedenbender, K.; Schmeck, B.T.; et al. Identification of microRNAs involved in NOD-dependent induction of pro-inflammatory genes in pulmonary endothelial cells. PLoS ONE 2020, 15, e0228764. [Google Scholar] [CrossRef]
  117. Tormo, E.; Ballester, S.; Adam-Artigues, A.; Burgués, O.; Alonso, E.; Bermejo, B.; Menéndez, S.; Zazo, S.; Madoz-Gúrpide, J.; Rovira, A.; et al. The miRNA-449 family mediates doxorubicin resistance in triple-negative breast cancer by regulating cell cycle factors. Sci. Rep. 2019, 29, 5316. [Google Scholar] [CrossRef]
  118. Qu, Y.; Dou, B.; Tan, H.; Feng, Y.; Wang, N.; Wang, D. Tumor microenvironment-driven non-cell-autonomous resistance to antineoplastic treatment. Mol. Cancer 2019, 18, 69. [Google Scholar] [CrossRef]
  119. Cataldo, A.; Romero-Cordoba, S.; Plantamura, I.; Cosentino, G.; Hidalgo-Miranda, A.; Tagliabue, E.; Iorio, M.V. MiR-302b as a Combinatorial Therapeutic Approach to Improve Cisplatin Chemotherapy Efficacy in Human Triple-Negative Breast Cancer. Cancers 2020, 12, 2261. [Google Scholar] [CrossRef]
  120. Ma, T.; Liu, X.; Cen, Z.; Xin, C.; Guo, M.; Zou, C.; Song, W.; Xie, R.; Wang, K.; Zhou, H.; et al. MicroRNA-302b negatively regulates IL-1β production in response to MSU crystals by targeting IRAK4 and EphA2. Arthritis Res. Ther. 2018, 20, 34. [Google Scholar] [CrossRef]
  121. Lee, S.J.; Jeong, J.-H.; Kang, S.H.; Kang, J.; Kim, E.A.; Lee, J.; Jung, J.H.; Park, H.Y.; Chae, Y.S. MicroRNA-137 Inhibits Cancer Progression by Targeting Del-1 in Triple-Negative Breast Cancer Cells. Int. J. Mol. Sci. 2019, 20, 6162. [Google Scholar] [CrossRef]
  122. Ying, X.; Sun, Y.; He, P. MicroRNA-137 inhibits BMP7 to enhance the epithelial-mesenchymal transition of breast cancer cells. Oncotarget 2017, 8, 18348–18358. [Google Scholar] [CrossRef] [PubMed]
  123. Zimta, A.-A.; Tigu, A.B.; Muntean, M.; Cenariu, D.; Slaby, O.; Berindan-Neagoe, I. Molecular Links between Central Obesity and Breast Cancer. Int. J. Mol. Sci. 2019, 20, 5364. [Google Scholar] [CrossRef] [PubMed]
  124. Xu, W.; Song, C.; Wang, X.; Li, Y.; Bai, X.; Liang, X.; Wu, J.; Liu, J. Downregulation of miR-155-5p enhances the anti-tumor effect of cetuximab on triple-negative breast cancer cells via inducing cell apoptosis and pyroptosis. Aging 2021, 13, 228–240. [Google Scholar] [CrossRef] [PubMed]
  125. Bar, I.; Theate, I.; Haussy, S.; Beniuga, G.; Carrasco, J.; Canon, J.-L.; Delrée, P.; Merhi, A. MiR-210 Is Overexpressed in Tumor-infiltrating Plasma Cells in Triple-negative Breast Cancer. J. Histochem. Cytochem. 2020, 68, 25–32. [Google Scholar] [CrossRef] [PubMed]
  126. Pottoo, F.H.; Iqubal, A.; Iqubal, M.K.; Salahuddin, M.; Rahman, J.U.; AlHajri, N.; Shehadeh, M. miRNAs in the Regulation of Cancer Immune Response: Effect of miRNAs on Cancer Immunotherapy. Cancers 2021, 13, 6145. [Google Scholar] [CrossRef]
  127. Piasecka, D.; Braun, M.; Kordek, R.; Sadej, R.; Romanska, H. MicroRNAs in regulation of triple-negative breast cancer progression. J. Cancer Res. Clin. Oncol. 2018, 144, 1401–1411. [Google Scholar] [CrossRef]
  128. Rippo, M.R.; Olivieri, F.; Monsurrò, V.; Prattichizzo, F.; Albertini, M.C.; Procopio, A.D. MitomiRs in human inflamm-aging: A hypothesis involving miR-181a, miR-34a and miR-146a. Exp. Gerontol. 2014, 56, 154–163. [Google Scholar] [CrossRef]
  129. Li, D.; Hu, J.; Song, H.; Xu, H.; Wu, C.; Zhao, B.; Xie, D.; Wu, T.; Zhao, J.; Fang, L. miR-143-3p targeting LIM domain kinase 1 suppresses the progression of triple-negative breast cancer cells. Am. J. Transl. Res. 2017, 9, 2276–2285. [Google Scholar]
  130. Meng, L.; Li, L.; Lu, S.; Li, K.; Su, Z.; Wang, Y.; Fan, X.; Li, X.; Zhao, G. The protective effect of dexmedetomidine on LPS-induced acute lung injury through the HMGB1-mediated TLR4/NF-κB and PI3K/Akt/mTOR pathways. Mol. Immunol. 2018, 94, 7–17. [Google Scholar] [CrossRef]
  131. Li, J.; Lai, Y.; Ma, J.; Liu, Y.; Bi, J.; Zhang, L.; Chen, L.; Yao, C.; Lv, W.; Chang, G.; et al. miR-17-5p suppresses cell proliferation and invasion by targeting ETV1 in triple-negative breast cancer. BMC Cancer 2017, 17, 745. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Wang, Y.; Xu, W.; Wang, Y.; Xu, X.; Lv, S.; Dong, X. miR-17-5p promotes migration and invasion in breast cancer cells by repressing netrin 4. Int. J. Clin. Exp. Pathol. 2019, 12, 1649–1657. [Google Scholar]
  133. Zhou, W.; Song, F.; Wu, Q.; Liu, R.; Wang, L.; Liu, C.; Peng, Y.; Mao, S.; Feng, J.; Chen, C. miR-217 inhibits triple-negative breast cancer cell growth, migration, and invasion through targeting KLF5. PLoS ONE 2017, 12, e0176395. [Google Scholar] [CrossRef] [PubMed]
  134. Chen, L.-L.; Zhang, Z.-J.; Yi, Z.-B.; Li, J.-J. MicroRNA-211-5p suppresses tumour cell proliferation, invasion, migration and metastasis in triple-negative breast cancer by directly targeting SETBP1. Br. J. Cancer 2017, 117, 78–88. [Google Scholar] [CrossRef] [PubMed]
  135. Yarahmadi, S.; Abdolvahabi, Z.; Hesari, Z.; Tavakoli-Yaraki, M.; Yousefi, Z.; Seiri, P.; Hosseinkhani, S.; Nourbakhsh, M. Inhibition of sirtuin 1 deacetylase by miR-211-5p provides a mechanism for the induction of cell death in breast cancer cells. Gene 2019, 711, 143939. [Google Scholar] [CrossRef]
  136. Tang, H.; Liu, P.; Yang, L.; Xie, X.; Ye, F.; Wu, M.; Liu, X.; Chen, B.; Zhang, L.; Xie, X. miR-185 Suppresses Tumor Proliferation by Directly Targeting E2F6 and DNMT1 and Indirectly Upregulating BRCA1 in Triple-Negative Breast Cancer. Mol. Cancer Ther. 2014, 13, 3185–3197. [Google Scholar] [CrossRef]
  137. Toda, H.; Kurozumi, S.; Kijima, Y.; Idichi, T.; Shinden, Y.; Yamada, Y.; Arai, T.; Maemura, K.; Fujii, T.; Horiguchi, J.; et al. Molecular pathogenesis of triple-negative breast cancer based on microRNA expression signatures: Antitumor miR-204-5p targets AP1S3. J. Hum. Genet. 2018, 63, 1197–1210. [Google Scholar] [CrossRef]
  138. Xiao, M.; Lou, C.; Xiao, H.; Yang, Y.; Cai, X.; Li, C.; Jia, S.; Huang, Y. MiR-128 regulation of glucose metabolism and cell proliferation in triple-negative breast cancer. Br. J. Cancer 2018, 105, 75–85. [Google Scholar] [CrossRef]
  139. Wang, Q.; Liang, D.; Shen, P.; Yu, Y.; Yan, Y.; You, W. Hsa_circ_0092276 promotes doxorubicin resistance in breast cancer cells by regulating autophagy via miR-348/ATG7 axis. Transl. Oncol. 2021, 14, 101045. [Google Scholar] [CrossRef]
  140. Rogers, T.J.; Christenson, J.L.; Greene, L.I.; O’Neill, K.I.; Williams, M.M.; Gordon, M.A.; Nemkov, T.; D’Alessandro, A.; Degala, G.D.; Shin, J.; et al. Reversal of Triple-Negative Breast Cancer EMT by miR-200c Decreases Tryptophan Catabolism and a Program of Immunosuppression. Mol. Cancer Res. 2019, 17, 30–41. [Google Scholar] [CrossRef]
  141. Carlomosti, F.; D’Agostino, M.; Beji, S.; Torcinaro, A.; Rizzi, R.; Zaccagnini, G.; Maimone, B.; Di Stefano, V.; De Santa, F.; Cordisco, S.; et al. Oxidative Stress-Induced miR-200c Disrupts the Regulatory Loop Among SIRT1, FOXO1, and eNOS. Antioxid. Redox Signal. 2017, 27, 328–344. [Google Scholar] [CrossRef] [PubMed]
  142. Fontana, A.; Barbano, R.; Dama, E.; Pasculli, B.; Rendina, M.; Morritti, M.G.; Melocchi, V.; Castelvetere, M.; Valori, V.M.; Ravaioli, S.; et al. Combined analysis of miR-200 family and its significance for breast cancer. Sci. Rep. 2021, 11, 2980. [Google Scholar] [CrossRef] [PubMed]
  143. Kim, M.-K.; Moon, Y.-A.; Kil Song, C.; Baskaran, R.; Bae, S.; Yang, S.-G. Tumor-suppressing miR-141 gene complex-loaded tissue-adhesive glue for the locoregional treatment of hepatocellular carcinoma. Theranostics 2018, 8, 3891–3901. [Google Scholar] [CrossRef] [PubMed]
  144. Shen, S.-J.; Song, Y.; Ren, X.-Y.; Xu, Y.-L.; Zhou, Y.-D.; Liang, Z.-Y.; Sun, Q. MicroRNA-27b-3p Promotes Tumor Progression and Metastasis by Inhibiting Peroxisome Proliferator-Activated Receptor Gamma in Triple-Negative Breast Cancer. Front. Oncol. 2020, 10, 1371. [Google Scholar] [CrossRef]
  145. Lamberti, M.J.; Nigro, A.; Casolaro, V.; Rumie Vittar, N.B.R.; Dal Col, J.D. Damage-Associated Molecular Patterns Modulation by microRNA: Relevance on Immunogenic Cell Death and Cancer Treatment Outcome. Cancers 2021, 13, 2566. [Google Scholar] [CrossRef]
  146. Chen, M.-Y.; Ye, X.-J.; He, X.-H.; Ouyang, D.-Y. The Signaling Pathways Regulating NLRP3 Inflammasome Activation. Inflammation 2021, 44, 1229–1245. [Google Scholar] [CrossRef]
  147. Zheng, D.; Liwinski, T.; Elinav, E. Inflammasome activation and regulation: Toward a better understanding of complex mechanisms. Cell Discov. 2020, 6, 36. [Google Scholar] [CrossRef]
  148. Wang, M.; Zhang, Y.; Chang, W.; Zhang, L.; Syrigos, K.N.; Li, P. Noncoding RNA-mediated regulation of pyroptotic cell death in cancer. Front. Oncol. 2022, 12, 1015587. [Google Scholar] [CrossRef]
  149. Wang, J.G.; Jian, W.J.; Li, Y.; Zhang, J. Nobiletin promotes the pyroptosis of breast cancer via regulation of miR -200b/JAZF1 axis. Kaohsiung J. Med. Sci. 2021, 37, 572–582. [Google Scholar] [CrossRef]
  150. Glinsky, G.V. SNP-guided microRNA maps (MirMaps) of 16 common human disorders identify a clinically accessible therapy reversing transcriptional aberrations of nuclear import and inflammasome pathways. Cell Cycle 2008, 7, 3564–3576. [Google Scholar] [CrossRef]
  151. Tang, H.-C.; Lai, Y.-Y.; Zheng, J.; Jiang, H.-Y.; Xu, G. miR-223-3p Inhibits Antigen Endocytosis and Presentation and Promotes the Tolerogenic Potential of Dendritic Cells through Targeting Mannose Receptor Signaling and Rhob. J. Immunol. Res. 2020, 1379458. [Google Scholar] [CrossRef] [PubMed]
  152. Xu, J.; Li, X.; Zhang, P.; Luo, J.; Mou, E.; Liu, S. miR-143-5p suppresses breast cancer progression by targeting the HIF-1α-related GLUT1 pathway. Oncol. Lett. 2022, 23, 147. [Google Scholar] [CrossRef] [PubMed]
  153. Indrieri, A.; Carrella, S.; Carotenuto, P.; Banfi, S.; Franco, B. The Pervasive Role of the miR-181 Family in Development, Neurodegeneration, and Cancer. Int. J. Mol. Sci. 2020, 21, 2092. [Google Scholar] [CrossRef]
  154. Heneghan, H.M.; Miller, N.; Lowery, A.; Sweeney, K.J.; Kerin, M.J. MicroRNAs as Novel Biomarkers for Breast Cancer. J. Oncol. 2010, 1950201. [Google Scholar] [CrossRef]
  155. Keklikoglou, I.; Koerner, C.; Schmidt, C.; Zhang, J.D.; Heckmann, D.; Shavinskaya, A.; Allgayer, H.; Guckel, B.; Fehm, T.F.; Schneeweiss, A.; et al. MicroRNA-520/373 family functions as a tumor suppressor in estrogen receptor negative breast cancer by targeting NF-κB and TGF-β signaling pathways. Oncogene 2012, 31, 4150–4163. [Google Scholar] [CrossRef]
  156. Ding, Q.; Shen, L.; Nie, X.; Lu, B.; Pan, X.; Su, Z.; Yan, A.; Yan, R.; Zhou, Y.; Li, L.; et al. MiR-223-3p overexpression inhibits cell proliferation and migration by regulating inflammation-associated cytokines in glioblastomas. Pathol. Res. Pr. 2018, 214, 1330–1339. [Google Scholar] [CrossRef]
  157. Farooqi, A.A.; Attar, R.; Tanriover, G.; Sabitaliyevich, U.Y.; Zhailganov, A.; Rabandiyarov, M. Regulation of NLRP3 by non-coding RNAs in different cancers: Interplay between non-coding RNAs and NLRP3 in carcinogenesis and metastasis. Cell. Mol. Biol. 2020, 66, 47–51. [Google Scholar] [CrossRef] [PubMed]
  158. Wang, Y.; Han, Z.; Fan, Y.; Zhang, J.; Chen, K.; Gao, L.; Zeng, H.; Cao, J.; Wang, C. MicroRNA-9 Inhibits NLRP3 Inflammasome Activation in Human Atherosclerosis Inflammation Cell Models through the JAK1/STAT Signaling Pathway. Cell. Physiol. Biochem. 2017, 41, 1555–1571. [Google Scholar] [CrossRef] [PubMed]
  159. 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]
  160. Grimaldi, A.M.; Salvatore, M.; Incoronato, M. miRNA-Based Therapeutics in Breast Cancer: A Systematic Review. Front. Oncol. 2021, 11, 668464. [Google Scholar] [CrossRef]
  161. Hussen, B.M.; Abdullah, S.T.; Rasul, M.F.; Salihi, A.; Ghafouri-Fard, S.; Hidayat, H.J.; Taheri, M. MicroRNAs: Important Players in Breast Cancer Angiogenesis and Therapeutic Targets. Front. Mol. Biosci. 2021, 8, 764025. [Google Scholar] [CrossRef] [PubMed]
  162. Cui, S.; Wang, R.; Chen, L. Micro RNA-145: A potent tumour suppressor that regulates multiple cellular pathways. J. Cell. Mol. Med. 2014, 18, 1913–1926. [Google Scholar] [CrossRef] [PubMed]
  163. Li, Y.; Li, X. miR-1290 modulates the radioresistance of triple-negative breast cancer by targeting NLRP3-mediated pyroptosis. Clin. Transl. Oncol. 2022, 24, 1764–1775. [Google Scholar] [CrossRef] [PubMed]
  164. Xue, Z.; Xi, Q.; Liu, H.; Guo, X.; Zhang, J.; Zhang, Z.; Li, Y.; Yang, G.; Zhou, D.; Yang, H.; et al. miR-21 promotes NLRP3 inflammasome activation to mediate pyroptosis and endotoxic shock. Cell Death Dis. 2019, 10, 461. [Google Scholar] [CrossRef]
  165. Kotiyal, S.; Bhattacharya, S. Biochemical and biophysical research communications breast cancer stem cells, EMT and therapeutic targets. Biochem. Biophys. Res. Commun. 2014, 453, 112–116. [Google Scholar] [CrossRef]
  166. Chabot, S.; Teissié, J.; Golzio, M. Targeted electro-delivery of oligonucleotides for RNA interference: siRNA and antimiR. Adv. Drug Deliv. Rev. 2015, 81, 161–168. [Google Scholar] [CrossRef]
  167. Conde, J.; Oliva, N.; Atilano, M.; Song, H.S.; Artzi, N. Self-assembled RNA-triple-helix hydrogel scaffold for microRNA modulation in the tumour microenvironment. Nat. Mater. 2016, 15, 353–363. [Google Scholar] [CrossRef]
  168. Zhang, S.; Lin, S.; Tang, Q.; Yan, Z. Knockdown of miR-205-5p alleviates the inflammatory response in allergic rhinitis by targeting B-cell lymphoma 6. Mol. Med. Rep. 2021, 24, 181. [Google Scholar] [CrossRef]
  169. Zhang, G.-F.; Wu, J.-C.; Wang, H.-Y.; Jiang, W.-D.; Qiu, L. Overexpression of microRNA-205-5p exerts suppressive effects on stem cell drug resistance in gallbladder cancer by down-regulating PRKCE. Biosci. Rep. 2020, 40, BSR20194509. [Google Scholar] [CrossRef]
  170. Lee, P.-S.; Chiou, Y.-S.; Ho, C.-T.; Pan, M.-H. Chemoprevention by resveratrol and pterostilbene: Targeting on epigenetic regulation. Biofactors 2017, 44, 26–35. [Google Scholar] [CrossRef]
  171. Song, J.; Ouyang, Y.; Che, J.; Li, X.; Zhao, Y.; Yang, K.; Zhao, X.; Chen, Y.; Fan, C.; Yuan, W. Potential Value of miR-221/222 as Diagnostic, Prognostic, and Therapeutic Biomarkers for Diseases. Front. Immunol. 2017, 8, 56. [Google Scholar] [CrossRef] [PubMed]
  172. Ebert, M.S.; Sharp, P.A. MicroRNA sponges: Progress and possibilities. RNA 2010, 16, 2043–2050. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Tay, F.C.; Lim, J.K.; Zhu, H.; Hin, L.C.; Wang, S. Using artificial microRNA sponges to achieve microRNA loss-of-function in cancer cells. Adv. Drug Deliv. Rev. 2014, 81, 117–127. [Google Scholar] [CrossRef] [PubMed]
  174. Zhang, K.; Liu, J.; Song, Q.; Yang, X.; Wang, D.; Liu, W.; Shi, J.; Zhang, Z. DNA Nanosponge for Adsorption and Clearance of Intracellular miR-21 and Enhanced Antitumor Chemotherapy. ACS Appl. Mater. Interfaces 2019, 11, 46604–46613. [Google Scholar] [CrossRef]
  175. Mandal, C.C.; Ghosh-Choudhury, T.; Dey, N.; Choudhury, G.G.; Ghosh-Choudhury, N. miR-21 is targeted by omega-3 polyunsaturated fatty acid to regulate breast tumor CSF-1 expression. Carcinogenesis 2012, 33, 1897–1908. [Google Scholar] [CrossRef]
  176. Singh, S.K.; Singh, S.; Lillard, J.W., Jr.; Singh, R. Drug delivery approaches for breast cancer. Int. J. Nanomed. 2017, 12, 6205–6218. [Google Scholar] [CrossRef]
  177. Adinew, G.; Taka, E.; Mendonca, P.; Messeha, S.; Soliman, K. The Anticancer Effects of Flavonoids through miRNAs Modulations in Triple-Negative Breast Cancer. Nutrients 2021, 13, 1212. [Google Scholar] [CrossRef]
  178. Cassidy, A.; Minihane, A.M. The role of metabolism (and the microbiome) in defining the clinical efficacy of dietary flavonoids. Am. J. Clin. Nutr. 2016, 105, 10–22. [Google Scholar] [CrossRef]
  179. Wang, M.; Chen, X.; Yu, F.; Zhang, L.; Zhang, Y.; Chang, W. The Targeting of Noncoding RNAs by Quercetin in Cancer Prevention and Therapy. Oxidative Med. Cell. Longev. 2022, 2022, 4330681. [Google Scholar] [CrossRef]
  180. La Rosa, F.; Mancuso, R.; Agostini, S.; Piancone, F.; Marventano, I.; Saresella, M.; Hernis, A.; Fenoglio, C.; Galimberti, D.; Scarpini, E.; et al. Pharmacological and Epigenetic Regulators of NLRP3 Inflammasome Activation in Alzheimer’s Disease. Pharmaceuticals 2021, 14, 1187. [Google Scholar] [CrossRef]
  181. Xie, Q.; Wei, M.; Zhang, B.; Kang, X.; Liu, D.; Zheng, W.; Pan, X.; Quan, Y.; Liao, D.; Shen, J. MicroRNA-33 regulates the NLRP3 inflammasome signaling pathway in macrophages. Mol. Med. Rep. 2017, 17, 3318–3327. [Google Scholar] [CrossRef] [PubMed]
  182. Hu, Y.; Wu, D.; Feng, X.; Shi, Z. Research on the effect of interfering with miRNA-155 on triple-negative breast cancer cells. Genes Genom. 2022, 44, 1117–1124. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The mechanism of the NLRP3 inflammasome assembly. NLRP3 activation require two signals: priming and activation. The priming signal is induced by the binding of DAMPs or PAMPs and stimulates the up-regulation of inflammasome components through the NF-kB inflammatory signaling pathway. Together, these two signals determine the assembly of the NLRP3 inflammasome and the activation of CASP1, which supports the maturation and release of the pro-inflammatory cytokines (pro-IL-1β and pro-IL-18), and catalyzes the cleavage of GSDMD and pore formation into the membrane, leading to pyroptosis (created with BioRender.com).
Figure 1. The mechanism of the NLRP3 inflammasome assembly. NLRP3 activation require two signals: priming and activation. The priming signal is induced by the binding of DAMPs or PAMPs and stimulates the up-regulation of inflammasome components through the NF-kB inflammatory signaling pathway. Together, these two signals determine the assembly of the NLRP3 inflammasome and the activation of CASP1, which supports the maturation and release of the pro-inflammatory cytokines (pro-IL-1β and pro-IL-18), and catalyzes the cleavage of GSDMD and pore formation into the membrane, leading to pyroptosis (created with BioRender.com).
Ijms 24 03245 g001
Figure 2. MiRNAs biogenesis and roles in TNBC progression. Genes are transcribed by RNA polymerase II/III into a pri-miRNA molecule, which is cleaved by DGRC8 and Drosha generating the pre-miRNA. The pre-miRNA molecule is exported into the cytoplasm via exportin-5, where Dicer and TRBP cleaves the pre-miRNA, resulting in the mature miRNA. The functional mature miRNA molecule is associated with AGO2 onto the RISC, leading to gene silencing via mRNA cleavage, translational repression and degradation. Additionally, the up-regulation of oncogenic miRNAs determines the suppression of target genes, while the down-regulation of tumor suppressor miRNAs stimulates the overexpression of target genes. MiRNAs assist essential biological processes such as cell growth, proliferation, migration and invasion, hypoxia, inflammation, apoptosis, pyroptosis and metastasis in TNBC (created with BioRender.com).
Figure 2. MiRNAs biogenesis and roles in TNBC progression. Genes are transcribed by RNA polymerase II/III into a pri-miRNA molecule, which is cleaved by DGRC8 and Drosha generating the pre-miRNA. The pre-miRNA molecule is exported into the cytoplasm via exportin-5, where Dicer and TRBP cleaves the pre-miRNA, resulting in the mature miRNA. The functional mature miRNA molecule is associated with AGO2 onto the RISC, leading to gene silencing via mRNA cleavage, translational repression and degradation. Additionally, the up-regulation of oncogenic miRNAs determines the suppression of target genes, while the down-regulation of tumor suppressor miRNAs stimulates the overexpression of target genes. MiRNAs assist essential biological processes such as cell growth, proliferation, migration and invasion, hypoxia, inflammation, apoptosis, pyroptosis and metastasis in TNBC (created with BioRender.com).
Ijms 24 03245 g002
Table 1. Molecular comparison between three proposed classifications of TNBC.
Table 1. Molecular comparison between three proposed classifications of TNBC.
TNBC ClassificationMethod of AnalysesNumber of PatientsSubtypesAbnormal MechanismsRelevant MarkersTherapeutic StrategiesRefs.
The Vanderbilt SubtypeK-means clustering586Basal-like 1Cell cycle
Cell proliferation
DNA damage response
MYC, PIK3CA, CDK6, AKT2, KRAS, FGFR1, IGF1R, CCNE1, CDKN2A/B, BRCA2, PTEN, MDM2, RB1, TP53, KI67PARP inhibitors
HDAC/DNMT inhibitors
Natural-killer therapy
Cisplatin,
[20,24,25,26]
Basal-like 2EGFR, MET, NGF, Wnt/β-catenin, TP63,
IGF1R signaling pathway
Glycolysis
Gluconeogenesis
TP53, TP63, EGFR, MET, BRCA1, RB1, PTEN, CDKN2A, UTXmTOR inhibitors
Growth factor inhibitors (lapatinib, gefitinib, cetuximab, etc.)
[20,24,27]
ImmunomodulatoryTh1/2, IL-7, IL-12 signaling pathwayTP53, CTNNA1, DDX18, HUWE1, NFKBIA, APC, BRAF, MAP K4, RB1, CTLA4, PDL1PD1/PDL1/CTLA4 inhibitors
Cisplatin
PARP inhibitors
[20,24]
Mesenchymal-likeCell motility
Cell proliferation
Cell differentiation
Wnt, TGFβ, Notch signaling pathway
Epithelial-mesenchymal transition
PTEN, RB1, TP53, PIK3CA, VEGFR2, PI3KCAmTOR inhibitors
Drugs targeting epithelial–mesenchymal transition
Abl/Src inhibitor
Dasatinib
[20,24,28]
Mesenchymal stem-likeCell motility
Cell differentiation
Growth factor signaling
Epithelial–mesenchymal transition
Low proliferation
ABCA8, PROCR, ENG, ALDHA1, PER1, ABCB1, TERT2IP, BCL2, BMP2, THY, HOXA5, HOXA10, MEIS1, MEIS2, MEOX1, MEOX2, MSX1, BMP2, ENG, ITGAV, KDR, NGFR, NT5E, PDGFR, THY1, VCAM1, VEGFR2mTOR/MEK/PI3K inhibitors,
Src antagonists Antiangiogenic drugs
Abl/Src inhibitor
Dasatinib
[20,24]
Luminal androgen receptorSteroid synthesis, porphyrin metabolism,
Androgen/estrogen metabolism
DHCR24, CD166, FASN, FKBP5, APOD, PIP, SPDEF, CLDN8Anti-AR therapy
PI3K/CDK4/6 inhibitors
[20,24,26,29]
The Baylor SubtypeNon-negative matrix factorization198Luminal androgen receptorSteroid hormone biosynthesis
Porphyrin and chlorophyll metabolism
PPAR signaling pathway
Androgen and estrogen metabolism
Hormonale-mediated signaling
TP53, PI3KCA, AKT1, ERBB2, ERBB4, CDK4/6, AR, MUC1, ER, CDH1, KRT7, KRT8, KRT18, KRT19, XBP1, FOXA1Anti-AR/MUC1 therapy[21,30,31,32,33,34]
MesenchymalCell motility
Epithelial–mesenchymal transition
Focal adhesion
TGF-β signaling pathway
Adipocytokine signaling pathway
PIK3CA, PTEN, STAT3, IGF1, prostaglandin, TGF-β, Wnt, β-catenin, PDGFRα, c-Kit, ABC transporterTKI/RAS/mTOR inhibitor
Growth factor inhibitors
[21,30,31,32,33]
Basal-like immunosuppressedMitotic cell cycle
Mitotic prometaphase
M phase of mitotic cell cycle
DNA replication
DNA repair
Immune response
Innate immune response
VTCN1, TP53, CENPF,
BUB1, PRC1, VTCN1, MS4A6A, MTBP, FGFR2, BARD1, RNASE6
VTCN1 inhibition[21,31]
Basal-like immune-activatedCytokine–cytokine receptor interaction
T cell receptor signaling pathway
B cell receptor signaling pathway
Chemokine signaling pathway
NF-kB signaling pathway
CCR2, CXCL13, CXCL11, CD1C, CXCL10, CCL5, STATDrugs targeting stat signal transduction molecules and cytokines[21,31,35]
The French SubtypeFuzzy clustering194Cluster 1Luminal androgen receptor enrichedAR, Hsp90, PI3K, FGFR4, TTN, TNR, PKHD1L1, SPTA1, NCKAP5, COL15A1, ANKRD11, MYLKAnti-AR therapy[36,37,38]
Cluster 2Basal-like with low immune response
High M2-like macrophages
High pro-tumorigenic
Low anti-tumor immune response
CCL2, CCL5, CCL18, CCL10, CXCL22, IL4, IL8, IL10, IL13, TGFβ1, CD206, CD204, VEGF, Aginase1, PIK3CA, NF1, AKT1, FBN3, ABCC1, DNHD1M2 inhibition
Repolarization of M2 into M1 macrophages
[20,38,39,40,41,42,43,44,45]
Cluster 3Basal-enriched
High immune response
Low M2-like macrophages
Low pro-tumorigenic
High anti-tumor immune response
IL-1β, IL-6, IL-12, IL-23,CXCL9, TNF-α, CCL2, IFNγ, GSF10, DNAH1, CDH23, AHNAK2, GTF3C1Repolarization of M2 into M1 macrophages[38,41,45]
Abbreviations: ATP-binding cassette (ABC), ATP-binding cassette subfamily a member 8 (ABCA8), ATP-binding cassette subfamily b member 1 (ABCB1), ATP-binding cassette subfamily c member 1 (ABCC1), tyrosine-protein kinase ABL1 (Abl), AHNAK nucleoprotein 2 (AHNAK2), protein kinase B (AKT), aldehyde dehydrogenase 1 family, member A1 (ALDHA1), ankyrin repeat domain 11 (ANKRD11), antigen-presenting cell (APC), apolipoprotein D (APOD), androgen receptor (AR), BRCA1-associated RING domain protein 1 (BARD1), B-cell lymphoma 2 (BCL2), bone morphogenetic protein 2 (BMP2), serine/threonine-protein kinase B-Raf (BRAF), breast cancer type susceptibility (BRCA), mitotic checkpoint serine/threonine-protein kinase (BUB1), chemokine (C-C motif) ligand (CCL), cyclin E1 (CCNE1), C-C chemokine receptor type 2 (CCR2), cluster of differentiation (CD), cadherin (CDH), cyclin-dependent kinases (CDK), cyclin-dependent kinase inhibitor (CDKN), centromere protein F (CENPF), claudin 8 (CLDN8), collagen type XV alpha 1 chain (COL15A1), cytotoxic T-lymphocyte associated protein 4 (CTLA4), catenin alpha 1 (CTNNA1), C-X-C motif chemokine ligand (CXCL), DEAD-box helicase 18 (DDX18), 24-dehydrocholesterol reductase (DHCR24), dynein axonemal heavy chain 1 (DNAH1), DNA methyltransferase 1 (DNMT), epidermal growth factor receptor (EGFR), endoglin (ENG), epidermal growth factor (ERBB), fatty acid synthase (FASN), fibrillin 3 (FBN3), fibroblast growth factor receptor (FGFR), FK506 binding protein 5 (FKBP5), forkhead box A1 (FOXA1), growth differentiation factor 10 (GSF10), general transcription factor IIIC subunit 1 (GTF3C1), histone deacetylase (HDAC), homeobox A cluster (HOXA), heat shock protein 90 (Hsp90), HECT, UBA and WWE domain containing 1, E3 ubiquitin protein ligase (HUWE1), interferon (IFN), insulin-like growth factor 1 (IGF1), insulin-like growth factor 1 receptor (IGF1R), interleukin (IL), integrin subunit alpha V (ITGAV), kinase insert domain receptor (KDR), marker of proliferation Ki-67 (KI67), Kirsten rat sarcoma virus (KRAS), keratin (KRT), mitogen-activated protein kinase 4 (MAPK4), mouse double minute 2 homolog (MDM2), Meis homeobox 1 (MEIS), mitogen-activated protein kinase kinase 1 (MEK), mesenchyme homeobox 1 (MEOX), tyrosine-protein kinase Met (MET), membrane spanning 4-domains A6A (MS4A6A), msh homeobox 1 (MSX1), MDM2 binding protein (MTBP), mechanistic target of rapamycin kinase (mTOR), mucin 1 (MUC1), myosin light chain kinase (MYLK), NCK associated protein 5 (NCKAP5), neurofibromatosis type 1 (NF1), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB), NFKB inhibitor alpha (NFKBIA), nerve growth factor (NGF), nerve growth factor receptor (NGFR), ecto-5′-nucleotidase (NT5E), poly (ADP-ribose) polymerase (PARP), programmed cell death protein 1 (PD1), platelet-derived growth factor receptor (PDGFR), programmed death ligand 1 (PDL1), period circadian regulator 1 (PER1), phosphoinositide 3-kinase (PI3K), phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), phosphatidylinositol phosphate (PIP), polycystic kidney and hepatic disease 1-like protein 1 (PKHD1L1), polycomb repressive complex 1 (PRC1), protein C receptor (PROCR), phosphatase and tensin homolog (PTEN), retinoblastoma susceptibility gene (RB1), ribonuclease a family member K6 (RNASE6), SAM pointed domain-containing Ets (SPDEF), spectrin alpha, erythrocytic 1 (SPTA1), steroid receptor coactivator (Src), signal transducer and activator of transcription (STAT), telomerase reverse transcriptase (TERT), transforming growth factor (TGF), tumor necrosis factor (TNF), tenascin-R (TNR), transient tachypnea of the newborn (TTN), ubiquitously transcribed tetratricopeptide repeat X chromosome (UTX), vascular cell adhesion molecule 1 (VCAM1), vascular endothelial growth factor (VEGF), vascular endothelial growth factor receptor 2 (VEGFR2), V-set domain-containing T-cell activation inhibitor 1 (VTCN1), wingless/integrated (Wnt), X-box binding protein 1 (XBP1).
Table 2. SOC for TNBC.
Table 2. SOC for TNBC.
ApproachClass of AgentsExamples of TherapyMechanism of ActionRefs.
NeoadjuvantAnthracycline + TaxaneDoxorubicin + Cyclophosphamide + Paclitaxel
Epirubicin + Cyclophosphamide + Nab-paclitaxel
Inhibition of DNA and RNA synthesis
Inhibition of topoisomerase II enzyme
Generation of reactive oxygen species (ROS)
Stabilization of microtubules
[49,51]
Fluoropyrimidine + TaxaneCapecitabine + Docetaxel
Fluoropyrimidine + EpothiloneCapecitabine + Ixabepilone
AdjuvantAnthracycline + TaxaneDoxorubicin + Cyclophosphamide + Docetaxel[49,52]
Table 3. Novel strategies for TNBC treatment.
Table 3. Novel strategies for TNBC treatment.
Class of AgentsExamples of TherapyMechanism of ActionRefs.
PD-1 and PD-L1 inhibitorsPembrolizumab + Paclitaxel
Doxorubicin + Cyclophosphamide
Pembrolizumab + Paclitaxel + Carboplatin
Durvalumab + Nab-paclitaxel
Atezolizumab + Nab-paclitaxel
Atezolizumab + Nab-paclitaxel + Carboplatin
Reactivation of the anti-tumor immune response
PD-1/PD-L1 complex formation inhibition
[55]
Platinum-based therapyCarboplatin + Eribulin
Gemcitabine + Carboplatin + Iniparib
Carboplatin + Bevacizumab
Cisplatin + Paclitaxel + Everolimus
Paclitaxel + Carboplatin
Double-strand DNA break
Apoptosis initiation
[56,57,58]
Cell cycle inhibitorsTrilaciclib, etoposide, abemaciclib, prexasertibActivate the spindle assembly/mitotic checkpoint
Prolonged mitotic arrest
Cell death initiation
[59]
Angiogenesis inhibitorsCisplatin + Bevacizumab
Anlotinib, apatinib, afatinib, lenvatinib, erlotinib, famitinib, pyrotinib
Blocking new blood vessel formation
Tumor growth inhibition
VEGF signaling pathway disruption
[60]
PI3K/AKT/mTOR inhibitorsRapamycin, ipatasertip, buparlisib, pictilisib, alpelisibCancer cells migration and invasion inhibition
Apoptosis initiation
[61,62]
PARP inhibitorsOlaparib + Carboplatin + Paclitaxel
Veliparib + Carboplatin
Cisplatin + Rucaparib
Veliparib, niraparib, talazoparib
Double-strand DNA break
Cell death initiation
Base excision repair
Relax/condense chromatin bind nucleosom PARylate H1/H2B
[63]
EGFR inhibitorsBintrafusp Alfa, dasatinib, geftinib, sorafenib, nimotuzumab, panitumumab, erlotinib, osimertinibCell death initiation
Inhibition of cancer cell proliferation
Blocking dimerization of receptors, auto-phosphorylation and downstream signaling
Inducing receptor internalization, degradation and stable downregulation
[64]
Androgen receptor (AR) antagonistsBicalutamide, enzalutamide, abiraterone, palbociclibDecrease in cancer cell viability
G1 phase arrest
Apoptosis induction
[30,65]
Antibody drug conjugatesSacituzumab govitecan,
Ladiratuzumab vedotin,
Trastuzumab deruxtecan
Cell growth and migration inhibition
Binding to the topoisomerase in DNA replication inhibition
S-phase-specific cell death initiation
DNA damage
[66]
Table 4. Examples of miRNAs involved in TNBC and their specific targets.
Table 4. Examples of miRNAs involved in TNBC and their specific targets.
miRNARegulationTargetsMain Biological MechanismsInflammasome ModulationRefs.
Oncogenic miRNAs
miR-135bUp-regulatedAPC, TGF-βPromotes proliferation, invasion, migration and metastasisAlters expression of inflammatory mediators (IL-1R1)
Suppresses CASP1 expression following IL-1α stimulation
[86,87]
miR-21Up-regulatedPTENPromotes TNBC cell proliferation
Inhibits apoptosis
Modulates NLRP3 phosphorylation. Inhibits the assembly of NLRP3 inflammasomes[110,111]
miR-301bUp-regulatedCYLDPromotes cell proliferation
Resistance to apoptosis
Targets the TLR4/NF-kBsignaling pathway[89,112]
miR-221Up-regulatedFOSL1, MEK, ZEB2Decreases the expression of epithelial-specific genes
Increases the expression of mesenchymal-specific genes
Suppresses oxidative
Suppression of NLRP3/ASC/CASP1 signaling pathway[113,114]
miR-298Up-regulatedP-gpDrug efflux
Promotes VEGF signaling
Regulates NF-kBsignaling pathway
Regulates the secretion of pro-inflammatory cytokines [115,116]
miR-449Up-regulatedE2F1, E2F3, CDK2Cell-cycle regulationRegulation of NLRP3
inflammasome activation
[117,118]
miR-302bUp-regulatedE2F1, ATMCell-cycle progression
DNA damage repair
Inhibits IL-1β secretion and maturation
Regulates the TLR/NF-kB signaling pathway
[119,120]
miR-663aUp-regulatedHSPG2Anti-apoptotic activityRegulates the pro-inflammatory IL-1β expression[90,91]
miR-638Up-regulatedBRCA1DNA damage repairInvolved in ATP synthesis-coupled electron transport[92,121]
miR-137Up-regulatedFSTL1Wnt/β-catenin signaling
Cellular stemness
Reduces the oxidative stress and inflammation via MAPK signaling pathway
Modulates TLR4 expression levels
[121,122]
miR-140Up-regulatedWnt/β-cateninCellular stemnessMediates inflammatory cytokines production
Modulates TLR4 signaling pathway
[105,123]
miR-155Up-regulatedCD44, CD90, ABCG2Cellular stemness
Drug efflux
Inhibition of TLR4/MyD88/NF-kB signaling[14,124]
miR-210Up-regulatedHIF1αPromotes hypoxiaModulates the necroptosis and pyroptosis in hypoxic conditions[88,125]
miR-9Up-regulatedELAVL1, JAK1Inhibition of EMTSuppression of the NLRP3 inflammasome
Inhibition of CASP1 expression and secretion of pro-inflammatory mediators
[11,126,127]
miR-181aUp-regulatedATG5, ATG22BAutophagy
Cellular stemness
Down-regulates the Bcl-2 expression
Modulates the expression of IL-1
[11,128]
miR-373Up-regulatedCD44Invasion, intravasation, migration and metastasisModulates the activation of inflammasome via caspase-8[84,85]
Tumor-Suppressor miRNAs
miR-143-3pDown-regulatedLIMK1Inhibits cell growth, proliferation, migration and invasionRegulates the MyD88/NF-kB signaling pathway
Decreases the level of inflammatory mediators
[129,130]
miR-17-5pUp or down regulatedETV1, PDCD4, PTEN, DR4Inhibits cell proliferation, invasion and apoptosisInhibits TXNIP/NLRP3 inflammasome pathway
Inhibits pyroptosis
[131,132]
miR-217Down-regulatedKLF5Inhibits cell growth and migrationModulates oxidative stress and pro-inflammatory cytokines release[94,133]
miR-211-5pDown-regulatedSETBP1Cell proliferation, migration and metatasisInhibits pyroptosis
Inhibits the expression of CASP1 and caspase-4
[134,135]
miR-185Down-regulatedE2F6, DNMT1Inhibits cell proliferationModulates the expression of NLRP family genes[76,136]
miR-204-5pDown-regulatedAPL1S3Promotes cancer cell aggressivenessDecreases the CASP1 and ASC expression levels
Inhibits the activation of NLRP3 inflammasome
Suppresses the inflammatory responses
[137,138]
miR-128Down-regulatedINSR, IRS1Inhibits cell proliferationConstrains the inflammasome assembly
Modulates the pyroptotic cell death
[138,139]
miR-340Down-regulatedSOX2, p16, p27Cell cycleInhibits pyroptosis and inflammation
Mediates the NEK7/NLRP3 signaling pathway
[95,96]
miR-384Down-regulatedACVR1Inhibits cell proliferation and migration Regulates inflammation[109,140]
miR-200cDown-regulatedXIAPInhibits cell proliferation promotes apoptosisDecreases the NLRP3, CASP1, and GSDMD expression levels
Inhibits NLRP3 inflammasome activation via NEK7 targeting
Inhibits ATP and LPS-induced pyroptosis
[141,142,143]
miR-31Down-regulatedBCL2, PKCεInduces apoptosis
Inhibits NF-kB signaling
Modulates inflammation and oxidative stress
Inhibits caspase-1 activity
Decreases NLRP3 and ASC expression levels
Inhibits IL-1β and IL-18 releases
[97,98]
miR-200aDown-regulatedPKCα, UBASH3B, XIAPSuppresses proliferation, migration, invasion, and metastasis
Promotes apoptosis
Regulates NOD2 expression[116,144]
miR-141Down-regulatedP27, CDK6, STAT5Maintenance of epithelial phenotypeModulates the expression of NLRP family genes[76,145]
miR-27b-3pDown-regulatedCBLB, GRB2Regulation of PI3K/AKT and MAPK/ERK signalingAttenuates NLRP3, CASP1, GSDMD, IL-1β and IL-18 and expression
Anti-pyroptosic effects
[146,147]
miR-223Down-regulatedHAX-1Stimulates apoptosis
Blocks the growth and the immunosuppressive ability of TNBC
Inhibits the activity of the NLRP3 inflammasome by binding the 3′-UTR of NLRP3 mRNA[99,100]
Abbreviations: ATP-binding cassette super-family G member 2 (ABCG2), activin A receptor type I (ACVR1), autophagy related (ATG), ataxia telangiectasia mutated (ATM), casitas B-lineage lymphoma proto-oncogene-B (CBLB), cylindromatosis (CYLD), E2F Transcription Factor (E2F), embryonic lethal abnormal vision-like protein 1 (ELAVL1), Ets translocation variant 1 (ETV1), programmed cell death 4 (PDCD4), death receptor 4 (DR4), Fos-related antigen 1 (FOSL1), follistatin-related protein 1 (FSTL1), adaptor protein growth factor receptor-bound protein 2 (GRB2), hematopoietic lineage cell-specific protein-associated protein X-1 (HAX-1), hypoxia-inducible factor 1-alpha (HIF1α), heparan sulfate proteoglycan 2 (HSPG2), insulin receptor (INSR), insulin receptor substrate 1 (IRS1), Janus kinase 1 (JAK1), Krüppel-like factor 5 (KLF5), LIM kinase-1 (LIMK1), mitogen-activated protein kinase kinase (MEK), P-glycoprotein 1 (P-gp), protein kinase C (PKC), SET binding protein 1 (SETBP1), sex determining region Y-box 2 (SOX2), ubiquitin-associated and SH3 domain-containing protein B (UBASH3B), x-linked inhibitor of apoptosis protein (XIAP), zinc finger E-box-binding homeobox 2 (ZEB2).
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

, L.-R.B.; Dinescu, S.; Costache, M. Regulation of Inflammasome by microRNAs in Triple-Negative Breast Cancer: New Opportunities for Therapy. Int. J. Mol. Sci. 2023, 24, 3245. https://doi.org/10.3390/ijms24043245

AMA Style

L-RB, Dinescu S, Costache M. Regulation of Inflammasome by microRNAs in Triple-Negative Breast Cancer: New Opportunities for Therapy. International Journal of Molecular Sciences. 2023; 24(4):3245. https://doi.org/10.3390/ijms24043245

Chicago/Turabian Style

(Stămat), Liliana-Roxana Balahura, Sorina Dinescu, and Marieta Costache. 2023. "Regulation of Inflammasome by microRNAs in Triple-Negative Breast Cancer: New Opportunities for Therapy" International Journal of Molecular Sciences 24, no. 4: 3245. https://doi.org/10.3390/ijms24043245

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