- Review
- Open access
- Published:
Molecular mechanisms of miR-192 in cancer: a biomarker and therapeutic target
Cancer Cell International volume 25, Article number: 94 (2025)
Abstract
Cancer remains a major global health challenge due to its rising prevalence and high mortality rates. The field of microRNAs (miRNAs) has made significant progress in the understanding of tumorigenesis and has broadened our knowledge of their targeting, especially in cancer therapy. miRNAs, a class of small non-coding RNAs, participate in post-transcriptional gene regulation by translational inhibition or mRNA degradation. Among these, microRNA-192 (miR-192) is located on human chromosome 11q13.1, and is highly correlated with the occurrence and development of various human cancers. Dysregulation of miR-192 has been extensively studied in various pathological processes, including tumorigenesis, making it a valuable biomarker for cancer diagnosis and prognosis. The functional role of miR-192 varies across cancer types, acting as either a tumor suppressor or as an oncogene through the modulation of multiple gene expressions and downstream signaling pathways. However, the roles of miR-192 in cancer appear inconsistent across types, with current research often focused on specific genes or pathways, limiting insight into its broader impact on cellular signaling networks. Therefore, this review aims to provide a comprehensive overview of miR-192 research. The paper reviews differences in miR-192 expression in cancer and systematically summarizes the role of miR-192 in cancers. The review further explores the complex roles of miR-192 in various pathological processes, emphasizing its regulatory pathways, interaction networks, and association with tumor progression. This review also illustrates the clinical application of miR-192 as a diagnostic and prognostic biomarker for non-invasive cancer detection, as it is consistently present in both serum and exosomes. A comprehensive summary and analysis of the relationship between miR-192 and various cancers may provide valuable insights, potentially guiding novel approaches in clinical diagnosis, therapeutic strategies, and foundational cancer research.
Background
Cancer poses a significant global public health challenge due to its increasing prevalence and high mortality rate. There remains an urgent need to identify novel diagnostic markers and therapeutic targets to improve patient outcomes [1, 2]. The exploration of molecular targets, particularly in the field of non-coding RNAs, has highlighted miRNAs as promising candidates due to their extensive regulatory roles in various pathological processes, including tumorigenesis [3]. Essentially, miRNAs are small non-coding RNA molecules (around 22 nucleotides) that regulate gene expression at the post-transcriptional level in a wide range of biological contexts [4, 5].
Furthermore, miRNAs are distinguished by a “seed” sequence, typically 7–8 nucleotides long, which binds to the 3'-untranslated regions (3'-UTRs) of target mRNAs, influencing mRNA stability and translation. The human miR-192 sequence, specifically the 24-CUGACCUAUGAAUUGACAGCC-44 seed (https://www.mirbase.org/search.shtml), is located on the human chromosome 11q13.1 [6]. miRNA biogenesis is a multi-step process involving transcription of primary miRNAs (pri-miRNAs) by RNA polymerase II or III, processed by the Drosha-DiGeorge Syndrome Critical Region 8 (Drosha-DGCR8) complex into precursor miRNAs (pre-miRNAs), with further maturation in the cytoplasm by Dicer to form the mature miRNA duplex [7,8,9,10,11]. For miR-192, the guide strand (miR-192-5p, originating from the 5’ side of the pre-miRNA) binds to the argonaut protein (AGO) to form the miRNA-induced silencing complex (RISC). Meanwhile, the passenger strand (miR-192-3p, the complementary 3’ strand) is usually degraded. Both miR-192-3p and miR-192-5p, however, target genes implicated in oncogenic processes, affecting cell proliferation, migration, invasion, epithelial-mesenchymal transition (EMT), angiogenesis, and drug resistance [12]. Both miR-192-3p and miR-192-5p target specific genes involved in cancer, thereby influencing key cellular processes, including cell proliferation, migration, invasion, EMT, angiogenesis and drug resistance.
miR-192 is aberrantly expressed across multiple cancer types, where it exerts either oncogenic or oncostatic effects, making it a promising target in cancer therapy. This dual role indicates its complex and context-dependent impact on cancer biology. Despite the growing evidence of miR-192's involvement in cancer, its functional roles appear to vary across different cancer types, and the consistency of these roles remains uncertain. Current research focuses on isolated target genes or specific signaling pathways, which limits our understanding of miR-192’s broader influence within the intricate network of cellular signaling pathways involved in cancer progression.
In understanding miR-192’s role in cancer, this review consolidates current insights into the aberrant expression, multifaceted functions, and complex regulatory mechanisms of miR-192 across various cancers. By highlighting its upstream regulators, downstream targets, and associated signaling pathways, The review aims to provide a comprehensive understanding of miR-192’s role in cancer progression. Additionally, its potential clinical applications as a diagnostic and prognostic biomarker are highlighted.
Expression of miR-192 in various cancers
miR-192 exhibits dynamic expression patterns that vary across tissues. It is highly expressed in certain normal tissues, such as the liver, kidney, intestine, and epithelial tissues, while its expression may be minimal in others. Notably, miR-192 expression is often dysregulated in cancers, where it undergoes significant alterations. A summary of miR-192 expression patterns in different cancers is provided in Table 1.
Upregulation of miR-192 in cancers
Several investigations have found elevated levels of miR-192 in various cancers, including cholangiocarcinoma, cervical cancer, hypoxic head and neck squamous cell carcinoma (HNSCC), and nasopharyngeal carcinoma. Specifically, studies have observed increased levels of miR-192-5p in cholangiocarcinoma tissues [12, 13], cell lines [12], urine [14], and serum of cholangiocarcinoma patients [15]. Similarly, elevated miR-192-5p levels have been found in cervical cancer tissues [16], hypoxic small extracellular vesicles (sEVs) from HNSCC [17], and nasopharyngeal carcinoma tissues and cells [18].
These findings emphasize the overexpression of miR-192 across various cancer types, indicating its potential role in cancer progression.
Downregulation of miR-192 in cancers
The downregulation of miR-192 has been consistently reported across multiple cancer types, including breast cancer, glioma, medulloblastoma, colorectal cancer, bladder cancer, pediatric acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), papillary thyroid carcinoma (PTC), osteosarcoma, and epithelial ovarian cancer (EOC) (Table 1). Numerous studies have documented decreased miR-192 expression in breast cancer tissues and cells [19, 20], with even lower levels observed in doxorubicin (DOX)-resistant breast cancer cell lines [21]. Significant reductions in miR-192 levels have also been observed in tissues and cells of glioma [22, 23], medulloblastoma [24], colorectal cancer [25,26,27,28,29,30,31], bladder Cancer [32], PTC [33], osteosarcoma [34,35,36,37,38], nephroblastoma subtypes [39], and endometrial cancer tissues [40], as well as in exosomes derived from tumor-associated macrophages (TAMs) [41].
Furthermore, miR-192 downregulation has been reported in various biofluids, including the serum of colon cancer [42, 43] and AML [44, 45] patients, the urine of bladder cancer patients [46] and the peripheral blood mononuclear cells (PBMCs) from CLL cases [47]. Interestingly, one study reported no difference in miR-192-5p expression in both colon cancer tissues and serum samples [43], suggesting further investigation to elucidate its role in this context.
Variable expression of miR-192 in cancers
Certainly, the expression profile of miR-192 exhibits paradoxical patterns across different cancers, particularly in esophageal cancer, EOC, gastric cancer, hepatocellular carcinoma, pancreatic cancer, lung cancer, and prostate cancer.
miR-192 expression increases in the plasma of esophageal cancer patients [48] and in the tissues and cells of esophageal squamous cell carcinoma (ESCC) [49]. Additionally, miR-192 expression decreases post-therapy [50]. Notably, one study found that miR-192-5p expression rises following neoadjuvant chemotherapy and cisplatin treatment [48], suggesting its role in enhancing sensitivity to cisplatin, though it may also contribute to esophageal cancer progression.
In EOC, miR-192 has been shown to be highly expressed in ovarian mucinous carcinoma tissues [51] but reduced in plasma exosomes of individuals with EOC [52]. This indicates that cancer development involves diverse types and phases, each characterized by distinct molecular features that contribute to the variation in miR-192 expression. Even within the same cancer, distinct cell types can exhibit multiple miR-192 expression patterns. For instance, miR-192 expression is higher in the serum of gastric cancer patients with distant metastasis compared to those without metastasis [67]. Additionally, studies reported both upregulation [53,54,55,56,57,58] and downregulation [43, 59, 60] of miR-192 in gastric cancer tissues, cell lines, and serum from gastric cancer patients, which indicates significant variability in its expression.
A similar pattern of discrepancies is observed in hepatocellular carcinoma and pancreatic cancer. In hepatocellular carcinoma, miR-192 is generally downregulated in tissues [61,62,63,64] and cell lines [62, 65, 66], but upregulated in patients’ serum exosomes and specific cell populations resembling cancer stem cells (CSC) [67]. In pancreatic cancer, elevated expression of miR-192 has been observed in the serum of patients [68], as well as in serum exosomes, tissues, and pancreatic ductal adenocarcinoma (PDAC) cells [69, 70]. However, it is downregulated in PDAC tissues and gemcitabine-resistant cells [69]. These variations can be attributed to the diverse sources of circulating miRNAs, which include tumor cells and PBMCs. miRNAs are encapsulated within extracellular vesicles or bound to protein complexes, providing protection from degradation and explaining the discrepancies in miR-192 levels between tissues and circulation. Furthermore, the differing half-lives of miRNAs in blood versus tissues, influenced by factors such as RNase activity, contribute to these variations.
In lung cancer, miR-192 displays diminished expression in tissues [71], serum of patients with bone metastasis [72], and bronchial lavage fluid [73]. However, cellular studies present conflicting results, with some showing decreased expression [72, 73], while others report increased expression [74,75,76], particularly higher in cisplatin-resistant lung carcinoma cells [77, 78]. In prostate cancer, data from the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) show elevated miR-192 expression in tissues [79], whereas cell studies on cultured cells report downregulation [80, 81]. Notably, cell lines often deviate from the characteristics of the original tumor tissues due to accumulated mutations over time, which can lead to discrepancies in miRNA expression. Additionally, the tumor microenvironment and various cell types present in tumor tissues further influence miR-192 levels, which may not be fully represented by in vitro studies.
Therefore, miR-192 shows variable expression across different cancers, influenced by factors such as tumor stage, metastasis, drug resistance, and the tumor microenvironment. These expression differences highlight its potential as a biomarker for cancer diagnosis, prognosis, and treatment monitoring. However, further research is needed to fully elucidate its role and mechanisms in different cancer types.
The role of miR-192 in cancer progression
The dysfunction of miR-192 is implicated in several key processes of tumorigenesis, including cancer cell proliferation, apoptosis, migration, invasion, metastasis, EMT, and angiogenesis. miR-192 exerts distinct roles across various tumors by regulating multiple target genes. Its primary role in most cancers is to prevent tumor development, However, in gastric cancer, it predominantly exhibits oncogenic properties (Fig. 1). The tumor-suppressive effect of miR-192 on specific target genes, such as MYC, XIAP, and ZEB2, is consistent across various cancers (Fig. 1). However, miR-192 can exhibit dual effects when targeting the same gene in different cancers, such as BCL-2 and RB1 (Fig. 1). Therefore, the distinct functions of miR-192 can be further explored by targeting specific genes in distinct types of cancer (Fig. 1).
Gastric cancer
Most studies have found that miR-192 functions as an oncogene in gastric cancer. Specifically, miR-192 has been observed to enhance cell proliferation and migration by downregulating ALCAM [56], APC [55], and Rab11-FIP2 [82]. In addition, it promotes EMT and Treg cell differentiation by suppressing RB1 [53]. Its role in decreasing SET domain containing lysine methyltransferase 8 (SET8) expression prevents cellular senescence and apoptosis, thus promoting gastric cancer initiation and progression [54].
However, one specific study identifies miR-192-5p as a tumor suppressor, reversing cisplatin resistance in gastric cancer cells by targeting ERCC3 and ERCC4 [60].
Lung cancer
Lung cancer remains consistently ranked as the most diagnosed cancer and was the primary cause of cancer-related deaths by 2022 [1, 2]. miR-192 plays a complex role in lung cancer, acting both as a tumor suppressor and an oncogene. It induces cell cycle arrest by downregulating CCNB1 and MYC [83, 84]. It also inhibits proliferation and EMT while promoting apoptosis through RB1 downregulation in non-small-cell lung cancer (NSCLC) [71, 85], and has been found to target TRIM44 to curb cancer cell proliferation, migration, and invasion [54]. Moreover, miR-192-5p exerts pro-apoptotic effects by interacting with XIAP in NSCLC cells [56].
Additionally, miR-192 may contribute to lung cancer progression by promoting cisplatin resistance and inhibiting apoptosis through the downregulation of NKRF [77] or BIM [78]. Additionally, miR-192 regulates chemoresistance to combined gemcitabine and cisplatin chemotherapy by targeting BCL-2 in human adenocarcinoma lung cancer A549 cells [74].
Liver cancer
Primarily, miR-192 primarily functions as a tumor suppressor in liver cancer. Studies have shown that miR-192-5p triggers apoptosis and hampers cell growth, migration, and invasion in hepatocellular carcinoma by downregulating XIAP [86]. Additionally, miR-192 targets TRIM25 to inhibit proliferation and migration in hepatitis B virus-related hepatocellular carcinoma [61], and targets SLC39A6 to reduce tumor metastasis in hepatocellular carcinoma cells [62]. Upregulation of miR-192-5p has also been associated with the activation of autophagy and apoptosis via CYR61 in hepatocellular carcinoma [66].
Interestingly, the absence of miR-192-5p has been observed to promote stemness features in CSC-positive hepatocellular carcinoma cells by upregulating MYC, PFKFB3, GLUT1 [87], and PABPC4 [65]. Another study demonstrated that ZEB2, both a target of miR-192 and an EMT activator, increases in the absence of miR-192, thereby promoting EMT and invasion [88].
However, a few studies indicate the oncogenic role of miR-192. For instance, miR-192-5p has been implicated in targeting SEMA3A, potentially enhancing cell proliferation, angiogenesis, and metastasis [67].
Colorectal cancer
Colorectal cancer research has identified miR-192-5p as a tumor suppressor, playing a crucial role in inhibiting cancer progression. In particular, miR-192-5p modulates apoptosis by targeting EIF5A2 [30], RAB2A [26], and BMPR2 [25], while also inhibiting cell proliferation, migration, and invasion. Additionally, its interaction with SRPX2 has been shown to suppress colorectal cancer development by inhibiting proliferation and enhancing glycolysis [28]. It also promotes apoptosis by downregulating BCL-2 [29], and inhibits EMT by targeting ZEB2 [29] and NID1 [89]. In addition, it hinders angiogenesis by regulating VEGFA [29] in colorectal cancer. Another study demonstrated that miR-192 sensitizes HCT-116 cells to methotrexate (MTX) by regulating DHFR, suggesting a novel therapeutic approach to the treatment of patients with colon cancer [90].
Breast cancer
Breast cancer is the most commonly diagnosed cancer among women and is the leading cause of cancer-related deaths globally [2]. In breast cancer, miR-192 exhibits tumor-suppressive effects. Specifically, miR-192 inhibits proliferation and promotes apoptosis by targeting CAV1 [19]. Additionally, miR-192-5p sensitizes breast cancer cells to DOX and regulates cell growth by targeting PPIA, highlighting its potential as a therapeutic target [21]. A mechanistic study further demonstrated that miR-192 induces cell cycle arrest by downregulating RB1 [20]. In triple-negative breast cancer (TNBC), miR-192 also acts as a tumor suppressor by increasing apoptosis and inhibiting both proliferation and migration, with ARHGAP19 identified as a novel target gene of miR-192 [91].
Cervical cancer
Cervical cancer ranked fourth for both incidence and mortality among women by 2022 [2]. In cervical cancer, miR-192-5p exerts a tumor-suppressive function by inhibiting proliferation, migration, and invasion through direct targeting of TRPM7 [92]. Silencing miR-192-5p has the opposite effect, promoting these oncogenic processes by regulating ALX1 [40] and RB1 [93]. Furthermore, miR-192-5p derived from TAMs exosomes has been observed to suppress EMT and promote apoptosis by targeting IRAK1 in endometrial cancer [41].
Osteosarcoma
miR-192 plays a tumor-suppressive role in osteosarcoma by regulating cell proliferation, apoptosis, migration, and invasion. It achieves these effects by selectively targeting specific genes, including TCF7 [37], MMP11 [35], and USP1 [36]. Additionally, another study demonstrated that miR-192 inhibits osteosarcoma cell proliferation and induces apoptosis by interacting with XIAP [34]. Additionally, miR-192 has been shown to enhance the sensitivity of MG-63 cells to MTX [94] and to improve the sensitivity to cisplatin by targeting USP1 [36].
Others
In nasopharyngeal carcinoma, miR-192 promotes tumor progression through RB1 inhibition [18], while in HNSCC, it enhances malignancy via CAV1 downregulation [17]. Similarly, miR-192 contributes to neuroblastoma development by targeting Dicer1 [95].
Conversely, miR-192 serves a tumor-suppressive function in various cancers. In PTC, it reduces malignancy by regulating SH3RF3, affecting migration, invasion, and EMT [33]. In bladder cancer, miR-192 inhibits tumor growth [32, 96], by downregulating the transcription factor Yin Yang 1 linked to cancer cell proliferation [32], and is also associated with gemcitabine resistance in bladder cancer [97]. In multiple myeloma, miR-192 acts as a tumor suppressor by targeting IL-17Rs [98], and bioinformatic analyses suggest it connects with the bone marrow microenvironment by interacting with CDKN2A, influencing tumor progression [99]. In AML, miR-192 suppresses cancer progression by targeting CCNT2 [45], ULK1 [100], and ZBTB20 [101], with additional anti-tumor effects linked to modulation of the WNT signaling pathway [102].
In renal cancers, miR-192 acts as a tumor suppressor by targeting MDM2, ZEB2, and TYMS in renal cell carcinoma, thereby reducing migration and invasion [103]. In nephroblastoma, it inhibits tumor growth by downregulating ACVR2B [39], and it further limits angiogenesis in renal tumors through targeting ECR1 [104]. Additionally, miR-192 suppresses tumor growth in brain cancers, targeting RAB2A in glioblastoma multiforme [23], ZEB2 in glioma [22], and DHFR in medulloblastoma [24]. In retinoblastoma, miR-192 curtails tumor cell migration and invasion by regulating ITGA1 [105].
In ovarian cancer, miR-192 exhibits anti-angiogenic effects by targeting HOXB9 [104]. Bioinformatic studies also identify miR-192-5p as a key miRNA in high-grade primary ovarian tumors [106].
Notably, miR-192 displays a dual role in pancreatic, esophageal, and prostate cancers. In PDAC, miR-192 overexpression promotes tumor growth by repressing SIP1 [70], though other studies suggest it may function as a tumor suppressor by inhibiting EMT via ZEB2 downregulation [69]. In esophageal cancer, miR-192 enhances cisplatin sensitivity by targeting ERCC3/4 [48] and increases tumor cell susceptibility to cytotoxic T lymphocytes by targeting BCL-2 [107]. However, miR-192 promotes proliferation and inhibits apoptosis by suppressing BIM [49], indicating a complex role in esophageal cancer progression and treatment response. In prostate cancer, miR-192 reduces cell proliferation by targeting NOB1 [80], though contrasting findings indicate it may also promote cell cycle progression and proliferation [79].
Mechanism of action of miR-192 as a tumor suppressor
Primarily, miR-192 acts as a tumor suppressor in cancer by regulating specific genes and pathways (Fig. 2). miR-192 is regulated by upstream factors, including non-coding RNAs, pharmaceutical treatments, and other factors.
The Anti-tumor Mechanisms of miR-192 in Cancer. This figure provides a comprehensive summary of the regulatory pathways through which miR-192 exerts tumor-suppressive effects in various cancers, including its upstream regulators, downstream targets, and associated signaling pathways, as well as potential therapeutic implications
Regulation of miR-192 by upstream factors
Circular RNAs (circRNAs) and long non-coding RNAs (lncRNAs) regulate cancer progression by acting as competitive endogenous RNAs (ceRNAs). They sequester miR-192, thereby protecting mRNAs from miRNA-induced degradation [108, 109]. For instance, circKIF5B [86] and circ_0000282 [34] promote osteosarcoma progression by specifically modulating the miR-192/XIAP axis. Similarly, circ_0000189 and circ_0001602 facilitate glioma and AML progression via the miR-192-5p/ZEB2 [22] and miR-192-5p/ZBTB20 axes, respectively [101].The IncRNAs such as SOX2-OT [23], XIST [61], and FTX [30] enhance the production of RAB2A, TRIM25, and EIF5A2 by sponging miR-192, thereby contributing to glioblastoma multiforme, hepatocellular carcinoma, and colorectal cancer progression, respectively.
Several pharmacological agents target miR-192 to regulate cancer progression, offering promising therapeutic strategies. For example, thymoquinone preserves miR-192 levels, inhibiting EMT by increasing E-cadherin expression and inducing apoptosis through the BAX/BCL-2 ratio [64]. BK002, a compound derived from Achyranthes japonica Nakai (AJN) and Melandrium firmum Rohrbach (MFR), both of which have been traditionally used as herbal medicines in China and Korea, induces the expression of miR-192-5p and the C/EBP homologous protein (CHOP). This in turn triggers apoptosis by downregulating BCL-2 and upregulating caspase activity [81]. Simvastatin activates miR-192 to inhibit colon cancer progression by targeting RAB2A [26]. Additionally, curcumin-induced upregulation of miR-192 in NSCLC exhibits anti-proliferative effects by targeting MYC and induces apoptosis by targeting XIAP [75, 84, 110]. Additionally, solamargine that is a natural compound found in Solanum nigrum L., exhibits multifaceted antitumor mechanisms. It elevates miR-192-5p expression through leukemia inhibitory factor (LIF) regulation, effectively inducing autophagy and apoptosis in hepatocellular carcinoma by targeting CYR61 [66]. Matrine, a quinolizidine alkaloid extracted from the traditional Chinese medicinal plant Sophora flavescens, exhibits diverse biological activities, including anticancer, anti-inflammatory, and antiviral effects. In thyroid cancer cells, matrine inhibits migration and invasion by regulating the miR-192-5p/SH3RF3 signaling axis [33].
Other factors also regulate miR-192 expression across various cancers. Nicotine downregulates miR-192 in NSCLC, enhancing proliferation and EMT through RB1 upregulation [85]. Rac GTPase-activating protein 1 (RacGAP1) inhibits miR-192-5p expression by suppressing p53, thereby promoting the carcinogenesis of cervical cancer through RB1 overexpression [93]. The intestinal microflora (MIM) in colon cancer increases miR-192-5p levels, inhibiting progression through BMPR2 suppression [25]. Furthermore, a regulatory feedback loop involving IL-17, miR-192, and IL-17Rs regulates proliferation and EMT in multiple myeloma [98]. tRF-3024b, a specific tRNA-derived fragment, hijacks miR-192-5p in ESCC, enhancing BCL-2 levels and contributing to cytotoxic T lymphocyte resistance [107]. Additionally, a study proposed a model in which p53 typically binds to miR-192, but when p53 binding is lost (due to P53 loss or mutation), Kruppel-like factor 5 (KLF5) binds to miR-192 instead [88]. This interaction leads to the transactivation of miR-192, effectively substituting for p53 function and inhibiting EMT in liver cancer cells. However, the loss of KLF5 results in the inactivation of miR-192 transcription. The expression of ZEB2, a downstream target of miR-192 and a driver of EMT, regulates EMT and invasion in cancers.
Pathways
PI3K/AKT signaling pathway
The PI3K/AKT signaling pathway is a crucial cellular signaling cascade involved in processes such as cell survival, proliferation, growth, and metabolism. Dysregulation of this pathway is associated with the development of several diseases, particularly cancer. Curcumin has shown the ability to inhibit cell proliferation and induce apoptosis in NSCLC cells. These effects are attributed to the upregulation of miR-192-5p, which suppresses the PI3K/AKT signaling pathway [75]. Similarly, BK002 has been shown to hinder the PI3K/AKT signaling pathway by upregulating miR-192, thereby promoting apoptosis in castration-resistant prostate cancer [81]. Furthermore, solamargine has been found to enhance cell apoptosis and autophagy by reducing the phosphorylation of AKT (pAKT) which is a key component of the PI3K/Akt pathway [66].
NF-κB and MAPK signaling pathways
The Nuclear Factor-kappa B (NF-κB) signaling pathway is a pivotal cellular pathway that regulates the transcription of genes involved in immune responses, inflammation, cell survival, and proliferation. In normal cells, NF-κB remains inactive in the cytoplasm by binding to IκB [111]. Upon IκB degradation, NF-κB translocates into the nucleus to activate target genes and execute its biological functions [111]. This pathway is implicated in various pathological processes in cancer. Thymoquinone has been shown to protect liver tissues by preserving miR-192 and interrupting the NF-κB signaling [64]. Additionally, the overexpression of miR-192-5p in TAMs-derived exosomes exhibits a suppressive effect on EMT by inhibiting the NF-κB signaling pathway in esophageal cancer [41].
The Mitogen-Activated Protein Kinase (MAPK) pathway is a fundamental signaling cascade that mediates the transmission of extracellular signals to the nucleus. This pathway regulates a spectrum of cellular processes, including cell growth, proliferation, differentiation, survival, and response to stress. This pathway plays a pivotal role in coordinating cellular responses to various stimuli, including growth factors, cytokines, and environmental stressors. Furthermore, Vacuolar Protein Sorting 33B (VPS33B) has been shown to increase the expression of miR-192-3p through the inactivation of Ras/ERK signaling. This leads to the inhibition of CCNB1 and subsequent blockade of cell proliferation [83].
TGF-β/SMAD and WNT/β-catenin signaling pathways
The Transforming Growth Factor-beta (TGF-β) signaling pathway is a pivotal and multifunctional cascade that intricately regulates diverse cellular processes, including cell proliferation, differentiation, apoptosis, and immune responses. It plays a crucial role in embryonic development, tissue homeostasis, and maintaining immune system balance. Dysregulation of the TGF-β pathway has been linked to various diseases, including cancer and fibrosis. Thymoquinone protects liver tissues by maintaining the levels of miR-192 and E-cadherin while also inhibiting TGF-β signaling [64]. Additionally, ACVR2B, which encodes a protein that functions as a member of the TGF-β signaling pathway, has been identified as a target gene of miR-192 [39]. The downregulating miR-192 may contribute to nephroblastoma development by targeting ACVR2B [39]. These findings emphasize the intricate role of the TGF-β signaling pathway in physiological processes and its involvement in pathological conditions.
The Wnt/β-catenin signaling pathway, also known as the classical Wnt pathway, is a critical and evolutionarily conserved cellular signaling cascade with essential roles in embryonic development, tissue homeostasis, and disease. Dysregulation of this pathway is implicated in multiple cancers. The inhibitory effects of curcumin on NSCLC cells may be associated with the upregulation of miR‑192‑5p. This occurs through the targeting of MYC and the deactivating of Wnt/β-catenin signaling pathway [84]. Delivery of miR-192 via hydrogel-based methods inhibits hepatocellular carcinoma progression by specifically targeting WNT10B, encoding Wnt Family Member 10B protein (WNT10B) which plays a crucial role in the GSK3β/Wnt/β-catenin pathway [112]. This finding highlights the complex role of the Wnt/β-catenin pathway in cellular processes and its potential modulation for therapeutic purposes, particularly involving miRNAs such as miR‑192‑5p.
These intricate regulatory mechanisms underscore the multifaceted role of miR-192 in cancer pathogenesis and underscore its potential as a therapeutic target in diverse malignancies.
Mechanism of action of miR-192 as an oncogene
While miR-192 acts as a tumor suppressor in many cancers, its role as an oncogene has also been observed across different malignancies. The regulation of miR-192 expression and activity in cancer is highly complex, involving multiple factors and signaling pathways (Fig. 3).
Regulation of miR-192 by upstream factors
The combination of bile and acid induces the deregulation of miR-192, a process facilitated by NF-κB activation, is associated with molecular and early histopathological alterations linked to neoplastic transformation [113]. Similarly, the NF-κB inhibitor BAY 11–7072 effectively prevents the acidic bile-induced upregulation of miR-192 in normal human hypopharyngeal cells [114].
Pathways
NF-κB and WNT/β-catenin signaling pathways
miR-192 can activate NF-κB either in various cancers by either directly or indirectly influencing specific target genes. In gastric cancer, miR-192 activates NF-κB by repressing its target RB1, hence promoting tumorigenesis through the induction of EMT [53]. Similarly, miR-192 downregulates the target gene NKRF to directly activate NF-κB, which contributes to tumorigenesis by inhibiting cell apoptosis, enhancing cell cycle progression, and promoting chemo-resistance in lung cancer [77]. Additionally, acidic bile induces upregulation of miR-192 by indirectly activating NF-κB, thereby promoting tumorigenesis in laryngeal cancer [113]. Conversely, the NF-κB pathway inhibitor BAY 11–7072 reduces the expression of miR-192 induced by acidic bile [114].
The WNT signaling pathway is activated by tumor suppressor APC [55] and SMG-1 [115], which have been identified as target genes of miR-192. This activation contributes to the occurrence and development of gastric cancer.
MAPK and PI3K-Akt signaling pathways
miR-192-5p has been shown to promote proliferation and suppress apoptosis in cholangiocarcinoma cells by activating the MEK/ERK pathway [12]. Additionally, miR-192 can activate the PI3K/Akt pathway, contributing to the progression of nasopharyngeal carcinoma [18].
Clinical application of miR-192 in human cancers
In cancer research, miR-192 has emerged as a central focus due to its potential as a diagnostic or prognostic biomarker across various cancer types. Its detectability in various biological fluids and tissues, and resistance to degradation by endogenous ribonuclease, make miR-192 an economical, sensitive, and non-invasive biomarker method. This overview explores the diverse value of miR-192 in cancer diagnosis and prognosis (Table 2).
Diagnostic biomarker
Firstly, miR-192 exhibits significant diagnostic potential across multiple cancer types, with its expression in blood (including serum, serum exosomes and PBMCs), tissues, and urinary sediment emerging as a valuable diagnostic marker.
In pancreatic cancer (including PDAC) [68,69,70, 116,117,118] and gastric cancer [43, 57,58,59], miR-192 serves as a reliable biomarker in serum, serum exosomes and tissue samples. Similarly, its presence in serum and tissue from colorectal cancer patients enhances its diagnostic utility [31, 42]. Also, miR-192 levels in hepatocellular carcinoma [119], EOC [52], and HNSCC [17], particularly in serum and serum exosomes, underline its broader clinical applicability. Additionally, circulating miR-192 in serum has shown promise as a marker for the early detection of cholangiocarcinoma [13], multiple myeloma [120] and pediatric AML [44]. Furthermore, mR-192 expression in PBMCs also holds potential for the early diagnosis of CLL [47].
miR-192 expression in tissues has shown diagnostic value in cervical cancer, prostate cancer, and ovarian carcinoma. Notably, miR-192 levels were observed to be six times higher in mucinous tumors compared to other histotypes of ovarian carcinoma [51]. This distinct pattern underlines the potential of miR-192 as a diagnostic biomarker, particularly for mucinous tumors within the spectrum of ovarian carcinoma.
Furthermore, miR-192 levels in urine have emerged as diagnostic biomarkers for bladder cancer [46, 121] and liver fluke-associated cholangiocarcinoma [14]. A decrease of miR-192 expression in the urinary sediment from bladder cancer patients may indicate tumor progression and combining miR-192 expression in urinary sediment with B-ultrasound demonstrates high sensitivity in diagnosing bladder cancer [46].
Prognostic biomarker
Also, miR-192 has demonstrated significant prognostic value across various cancer types, with its expression correlating with disease progression and clinical outcomes. In cholangiocarcinoma, postoperative declines in circulating miR-192 are associated with favorable outcomes, including reduced lymph node metastasis and improved overall survival [13, 15]. Similarly, in NSCLL, miR-192 expression levels in serum and bronchial wash samples are significantly associated with TNM stages, distant metastases, pathological grade, and overall survival, establishing its role as a valuable prognostic marker [73]. In colon cancer, miR-192 levels in serum and tissue samples from colon cancer patients have been linked to poor differentiation, lymphatic metastasis, vascular invasion, and advanced TNM stages, further underscoring the prognostic value of miR-192 [27, 31, 42, 43]. Reduced miR-192 expression in serum from pediatric AML patients has been associated with poor prognosis [44], while miR-192 levels in PBMCs hold potential as a prognostic biomarker for CLL [47]. Exosomal miR-192 also exhibits prognostic significance in hepatocellular carcinoma [119], EOC [52] and gastric cancer [57], where its overexpression correlates with poor overall survival. Likewise, miR-192 expression patterns in colorectal cancer, osteosarcoma, HNSCC, and esophageal cancer tissues correlate with clinical outcomes, offering further prognostic insights [38, 50, 122].
Conclusions
A substantial body of research has explored the context-dependent roles of miR-192 in various cancers, underscoring its promising clinical potential. The distinct expression patterns of miR-192 across cancer types highlight its value as a diagnostic and prognostic biomarker. Notably, aberrant miR-192 levels detected in whole blood, plasma, and other patient specimens suggest its potential utility in non-invasive cancer diagnosis, disease monitoring, and prognostic assessment. Also, miR-192 plays a critical role in the molecular networks underlying tumorigenesis by modulating key functional pathways. These findings highlight miR-192 as potential therapeutic targets, offering new avenues for cancer treatment and intervention. Recent advancements in delivery technologies, including nanoparticles, liposomes, and viral vectors, have improved the feasibility of miRNA-based therapies by enabling precise delivery to tumor sites or systemic administration. Non-invasive delivery strategies are also under development to enhance miRNA transport across cellular barriers, improving targeting specificity and therapeutic efficiency.
This review has comprehensively discussed the aberrant expression, diverse functions, and clinical applications of miR-192 in cancer, with an emphasis on its regulatory mechanisms and networks. However, many of the mechanisms and pathways highlighted are based on preliminary findings, necessitating further validation through larger-scale studies to confirm their clinical relevance and therapeutic applicability.
Availability of data and materials
No datasets were generated or analysed during the current study.
Abbreviations
- miR-192:
-
microRNA-192
- miRNAs:
-
microRNAs
- EMT:
-
Epithelial-mesenchymal transition
- HNSCC:
-
Hypoxic head and neck squamous cell carcinoma
- sEVs:
-
Small extracellular vesicles
- AML:
-
Acute myeloid leukemia
- CLL:
-
Chronic lymphocytic leukemia
- PTC:
-
Papillary thyroid carcinoma
- EOC:
-
Epithelial ovarian cancer
- DOX:
-
Doxorubicin
- TNBC:
-
Triple-negative breast cancer
- PBMCs:
-
Peripheral blood mononuclear cells
- TAMs:
-
Tumor-associated macrophages
- ESCC:
-
Esophageal squamous cell carcinoma
- CSC:
-
Cancer stem cell
- PDAC:
-
Pancreatic ductal adenocarcinoma
- NSCLC:
-
Non-small-cell lung cancer
- MTX:
-
Methotrexate
- circRNAs:
-
Circular RNAs
- lncRNAs:
-
Long non-coding RNAs
- NF-κB:
-
Nuclear Factor-kappa B
- MAPK:
-
Mitogen-Activated Protein Kinase
- TGF-β:
-
Transforming Growth Factor-beta
References
Sung H, Ferlay J, Siegel R, 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 A Cancer J Clin. 2021;71(3):209–49.
Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, Jemal A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA A Cancer J Clin. 2024;74(3):229–63.
Condrat CE, Thompson DC, Barbu MG, Bugnar OL, Boboc A, Cretoiu D, Suciu N, Cretoiu SM, Voinea SC. miRNAs as biomarkers in disease: latest findings regarding their role in diagnosis and prognosis. Cells. 2020;9(2):276.
Ashrafizadeh M, Zarrabi A, Hushmandi K, Hashemi F, Moghadam ER, Owrang M, Hashemi F, Makvandi P, Goharrizi MASB, Najafi M, et al. Lung cancer cells and their sensitivity/resistance to cisplatin chemotherapy: role of microRNAs and upstream mediators. Cell Signal. 2021;78: 109871.
Paskeh MDA, Mirzaei S, Orouei S, Zabolian A, Saleki H, Azami N, Hushmandi K, Baradaran B, Hashmi M, Aref AR, et al. Revealing the role of miRNA-489 as a new onco-suppressor factor in different cancers based on pre-clinical and clinical evidence. Int J Biol Macromol. 2021;191:727–37.
Yu M, Du H, Zhang C, Shi Y. miR-192 family in breast cancer: regulatory mechanisms and diagnostic value. Biomed Pharmacother. 2024;175: 116620.
Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116(2):281–97.
Achkar NP, Cambiagno DA, Manavella PA. miRNA biogenesis: a dynamic pathway. Trends Plant Sci. 2016;21(12):1034–44.
Winter J, Jung S, Keller S, Gregory RI, Diederichs S. Many roads to maturity: microRNA biogenesis pathways and their regulation. Nat Cell Biol. 2009;11(3):228–34.
Fabian MR, Sonenberg N, Filipowicz W. Regulation of mRNA translation and stability by microRNAs. Annu Rev Biochem. 2010;79:351–79.
Yi R, Qin Y, Macara IG, Cullen BR. Exportin-5 mediates the nuclear export of pre-microRNAs and short hairpin RNAs. Genes Dev. 2003;17(24):3011–6.
Tang C, Yuan P, Wang J, Zhang Y, Chang X, Jin D, Lei P, Lu Z, Chen B. MiR-192–5p regulates the proliferation and apoptosis of cholangiocarcinoma cells by activating MEK/ERK pathway. 3 Biotech. 2021;11(2):99.
Loosen SH, Lurje G, Wiltberger G, Vucur M, Koch A, Kather JN, Paffenholz P, Tacke F, Ulmer FT, Trautwein C, et al. Serum levels of miR-29, miR-122, miR-155 and miR-192 are elevated in patients with cholangiocarcinoma. PLoS ONE. 2019;14(1): e0210944.
Silakit R, Loilome W, Yongvanit P, Thongchot S, Sithithaworn P, Boonmars T, Koonmee S, Titapun A, Khuntikeo N, Chamadol N, et al. Urinary microRNA-192 and microRNA-21 as potential indicators for liver fluke-associated cholangiocarcinoma risk group. Parasitol Int. 2017;66(4):479–85.
Silakit R, Loilome W, Yongvanit P, Chusorn P, Techasen A, Boonmars T, Khuntikeo N, Chamadol N, Pairojkul C, Namwat N. Circulating miR-192 in liver fluke-associated cholangiocarcinoma patients: a prospective prognostic indicator. J Hepatobiliary Pancreat Sci. 2014;21(12):864–72.
Xu J, Zou J, Wu L, Lu W. Transcriptome analysis uncovers the diagnostic value of miR-192-5p/HNF1A-AS1/VIL1 panel in cervical adenocarcinoma. Sci Rep. 2020;10(1):16584.
Zhu G, Cao B, Liang X, Li L, Hao Y, Meng W, He C, Wang L, Li L. Small extracellular vesicles containing miR-192/215 mediate hypoxia-induced cancer-associated fibroblast development in head and neck squamous cell carcinoma. Cancer Lett. 2021;506:11–22.
Huang Q, Hou S, Zhu X, Liu S. MicroRNA-192 promotes the development of nasopharyngeal carcinoma through targeting RB1 and activating PI3K/AKT pathway. World J Surg Oncol. 2020;18(1):29.
Chen P, Feng Y, Zhang H, Shi X, Li B, Ju W, Yu X, Zhang N, Luo X. MicroRNA-192 inhibits cell proliferation and induces apoptosis in human breast cancer by targeting caveolin 1. Oncol Rep. 2019;42(5):1667–76.
Hu F, Meng X, Tong Q, Liang L, Xiang R, Zhu T, Yang S. BMP-6 inhibits cell proliferation by targeting microRNA-192 in breast cancer. Biochim Biophys Acta. 2013;1832(12):2379–90.
Zhang Y, He Y, Lu LL, Zhou ZY, Wan NB, Li GP, He X, Deng HW. miRNA-192-5p impacts the sensitivity of breast cancer cells to doxorubicin via targeting peptidylprolyl isomerase A. Kaohsiung J Med Sci. 2019;35(1):17–23.
Yang J, Hou G, Chen H, Chen W, Ge J. Circ_0000189 promotes the malignancy of glioma cells via regulating miR-192-5p-ZEB2 Axis. Oxid Med Cell Longev. 2022;2022:2521951.
Wang H, Hu Q, Tong Y, Li S, Chen M, Wang B, Li H. LncRNA SOX2-OT regulates miR-192-5p/RAB2A axis and ERK pathway to promote glioblastoma cell growth. Cell Cycle. 2021;20(19):2010–20.
Yang SY, Choi SA, Lee JY, Park AK, Wang KC, Phi JH, Koh EJ, Park WY, Park SH, Hwang DW, et al. miR-192 suppresses leptomeningeal dissemination of medulloblastoma by modulating cell proliferation and anchoring through the regulation of DHFR, integrins, and CD47. Oncotarget. 2015;6(41):43712–30.
Huang YL, Li XH, Ma H, Yue HY, Hu XY. Metabolites of intestinal microflora upregulate miR-192-5p to suppress proliferation of colon cancer cells via RhoA-ROCK-LIMK2 pathway. Eur Rev Med Pharmacol Sci. 2020;24(4):1794–806.
Zheng XF, Liu KX, Wang XM, Zhang R, Li X. MicroRNA-192 acts as a tumor suppressor in colon cancer and simvastatin activates miR-192 to inhibit cancer cell growth. Mol Med Rep. 2019;19(3):1753–60.
Li P, Ou Q, Braciak TA, Chen G, Oduncu FS. MicroRNA-192-5p is a predictive biomarker of survival for Stage IIIB colon cancer patients. Jpn J Clin Oncol. 2018;48(7):619–24.
Zhao J, Xu J, Zhang R. SRPX2 regulates colon cancer cell metabolism by miR-192/215 via PI3K-Akt. Am J Transl Res. 2018;10(2):483–90.
Geng L, Chaudhuri A, Talmon G, Wisecarver JL, Are C, Brattain M, Wang J. MicroRNA-192 suppresses liver metastasis of colon cancer. Oncogene. 2014;33(46):5332–40.
Zhao K, Ye Z, Li Y, Li C, Yang X, Chen Q, Xing C. LncRNA FTX contributes to the progression of colorectal cancer through regulating miR-192-5p/EIF5A2 Axis. Onco Targets Ther. 2020;13:2677–88.
Chiang Y, Song Y, Wang Z, Liu Z, Gao P, Liang J, Zhu J, Xing C, Xu H. microRNA-192, -194 and -215 are frequently downregulated in colorectal cancer. Exp Ther Med. 2012;3(3):560–6.
Ji D, Jiang L, Li Y. MiR-192-5p suppresses the growth of bladder cancer cells via targeting Yin Yang 1. Hum Cell. 2018;31(3):210–9.
Fu S, Ma C, Tang X, Ma X, Jing G, Zhao N, Ran J. MiR-192-5p inhibits proliferation, migration, and invasion in papillary thyroid carcinoma cells by regulation of SH3RF3. 2021. Biosci Reports. https://doiorg.publicaciones.saludcastillayleon.es/10.1042/BSR20210342.
Li H, He L, Tuo Y, Huang Y, Qian B. Circular RNA hsa_circ_0000282 contributes to osteosarcoma cell proliferation by regulating miR-192/XIAP axis. BMC Cancer. 2020;20(1):1026.
Shang G, Mi Y, Mei Y, Wang G, Wang Y, Li X, Wang Y, Li Y, Zhao G. MicroRNA-192 inhibits the proliferation, migration and invasion of osteosarcoma cells and promotes apoptosis by targeting matrix metalloproteinase-11. Oncol Lett. 2018;15(5):7265–72.
Zhou S, Xiong M, Dai G, Yu L, Zhang Z, Chen J, Guo W. MicroRNA-192-5p suppresses the initiation and progression of osteosarcoma by targeting USP1. Oncol Lett. 2018;15(5):6947–56.
Wang Y, Zhang S, Xu Y, Zhang Y, Guan H, Li X, Li Y, Wang Y. Upregulation of miR-192 inhibits cell growth and invasion and induces cell apoptosis by targeting TCF7 in human osteosarcoma the journal of the international society for oncodevelopmental biology and medicine. Tumour Biol. 2016;37(11):15211–20.
Wang Y, Jia LS, Yuan W, Wu Z, Wang HB, Xu T, Sun JC, Cheng KF, Shi JG. Low miR-34a and miR-192 are associated with unfavorable prognosis in patients suffering from osteosarcoma. Am J Transl Res. 2015;7(1):111–9.
Senanayake U, Das S, Vesely P, Alzoughbi W, Fröhlich LF, Chowdhury P, Leuschner I, Hoefler G, Guertl B. miR-192, miR-194, miR-215, miR-200c and miR-141 are downregulated and their common target ACVR2B is strongly expressed in renal childhood neoplasms. Carcinogenesis. 2012;33(5):1014–21.
Ni J, Tian W, Liang S, Wang H, Ren Y. Promoter methylation-mediated silencing of the MiR-192-5p promotes endometrial cancer progression by targeting ALX1. Int J Med Sci. 2021;18(12):2510–20.
Wang Y, Ma H, Li Y, Su R. MiR-192-5p-modified tumor-associated macrophages-derived exosome suppressed endometrial cancer progression through targeting IRAK1/NF-κB signaling. Reprod Sci. 2022;29(2):436–47.
Chen X, Su X, Lin M, Fu B, Zhou C, Ling C, Qian Z, Yao Y. Expression of miR-192-5p in colon cancer serum and its relationship with clinicopathologic features. Am J Transl Res. 2021;13(8):9371–6.
Tavakolian S, Goudarzi H, Faghihloo E. Evaluating the expression level of miR-9-5p and miR-192-5p in gastrointestinal cancer: introducing novel screening biomarkers for patients. BMC Res Notes. 2020;13(1):226.
Tian C, Zhang L, Li X, Zhang Y, Li J, Chen L. Low miR-192 expression predicts poor prognosis in pediatric acute myeloid leukemia. Cancer Biomark. 2018;22(2):209–15.
Ke S, Li RC, Lu J, Meng FK, Feng YK, Fang MH. MicroRNA-192 regulates cell proliferation and cell cycle transition in acute myeloid leukemia via interaction with CCNT2. Int J Hematol. 2017;106(2):258–65.
Jiang F, Li C, Han J, Wang L. Diagnostic value of combination of MicroRNA-192 in urinary sediment and B-ultrasound for bladder cancer. Technol Cancer Res Treat. 2020;19:1533033819894573.
Fathullahzadeh S, Mirzaei H, Honardoost MA, Sahebkar A, Salehi M. Circulating microRNA-192 as a diagnostic biomarker in human chronic lymphocytic leukemia. Cancer Gene Ther. 2016;23(10):327–32.
Furuke H, Konishi H, Arita T, Kataoka S, Shibamoto J, Takabatake K, Takaki W, Shimizu H, Yamamoto Y, Komatsu S, et al. Plasma microRNA-192–5p can predict the response to neoadjuvant chemotherapy and prognosis in esophageal cancer. Cancer Sci. 2022. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/cas.15703.
Li S, Li F, Niu R, Zhang H, Cui A, An W, Wang X. Mir-192 suppresses apoptosis and promotes proliferation in esophageal aquamous cell caicinoma by targeting Bim. Int J Clin Exp Pathol. 2015;8(7):8048–56.
Odenthal M, Bollschweiler E, Grimminger PP, Schröder W, Brabender J, Drebber U, Hölscher AH, Metzger R, Vallböhmer D. MicroRNA profiling in locally advanced esophageal cancer indicates a high potential of miR-192 in prediction of multimodality therapy response. Int J Cancer. 2013;133(10):2454–63.
Agostini A, Brunetti M, Davidson B, Tropé CG, Eriksson AGZ, Heim S, Panagopoulos I, Micci F. The microRNA miR-192/215 family is upregulated in mucinous ovarian carcinomas. Sci Rep. 2018;8(1):11069.
Chen L, Wang K, Li L, Zheng B, Zhang Q, Zhang F, Chen J, Wang S. Plasma exosomal miR-1260a, miR-7977 and miR-192-5p as diagnostic biomarkers in epithelial ovarian cancer. Future Oncol. 2022;18(26):2919–31.
Song J, Lin Z, Liu Q, Huang S, Han L, Fang Y, Zhong P, Dou R, Xiang Z, Zheng J, et al. MiR-192-5p/RB1/NF-κBp65 signaling axis promotes IL-10 secretion during gastric cancer EMT to induce Treg cell differentiation in the tumour microenvironment. Clin Transl Med. 2022;12(8): e992.
Zhang X, Peng Y, Yuan Y, Gao Y, Hu F, Wang J, Zhu X, Feng X, Cheng Y, Wei Y, et al. Histone methyltransferase SET8 is regulated by miR-192/215 and induces oncogene-induced senescence via p53-dependent DNA damage in human gastric carcinoma cells. Cell Death Dis. 2020;11(10):937.
Deng S, Zhang X, Qin Y, Chen W, Fan H, Feng X, Wang J, Yan R, Zhao Y, Cheng Y, et al. miRNA-192 and -215 activate Wnt/β-catenin signaling pathway in gastric cancer via APC. J Cell Physiol. 2020;235(9):6218–29.
Jin Z, Selaru FM, Cheng Y, Kan T, Agarwal R, Mori Y, Olaru AV, Yang J, David S, Hamilton JP, et al. MicroRNA-192 and -215 are upregulated in human gastric cancer in vivo and suppress ALCAM expression in vitro. Oncogene. 2011;30(13):1577–85.
He J, Wu J, Dong S, Xu J, Wang J, Zhou X, Rao Z, Gao W. Exosome-encapsulated miR-31, miR-192, and miR-375 Serve as clinical biomarkers of gastric cancer. Journal of oncology. 2023;2023:7335456.
Chen Q, Ge X, Zhang Y, Xia H, Yuan D, Tang Q, Chen L, Pang X, Leng W, Bi F. Plasma miR-122 and miR-192 as potential novel biomarkers for the early detection of distant metastasis of gastric cancer. Oncol Rep. 2014;31(4):1863–70.
Chiang Y, Zhou X, Wang Z, Song Y, Liu Z, Zhao F, Zhu J, Xu H. Expression levels of microRNA-192 and -215 in gastric carcinoma. Pathol Oncol Res. 2012;18(3):585–91.
Xie X, Huang N, Zhang Y, Wei X, Gao M, Li M, Ning J, Liu W, Zhao Q, Wang H, et al. MiR-192-5p reverses cisplatin resistance by targeting ERCC3 and ERCC4 in SGC7901/DDP cells. J Cancer. 2019;10(4):1039–51.
Wang J, Yin G, Bian H, Yang J, Zhou P, Yan K, Liu C, Chen P, Zhu J, Li Z, et al. LncRNA XIST upregulates TRIM25 via negatively regulating miR-192 in hepatitis B virus-related hepatocellular carcinoma. Mol Med. 2021;27(1):41.
Lian J, Jing Y, Dong Q, Huan L, Chen D, Bao C, Wang Q, Zhao F, Li J, Yao M, et al. miR-192, a prognostic indicator, targets the SLC39A6/SNAIL pathway to reduce tumor metastasis in human hepatocellular carcinoma. Oncotarget. 2016;7(3):2672–83.
Wang X, Wu S, Yang Y, Zhao J. LncRNA CARMN affects hepatocellular carcinoma prognosis by regulating the miR-192-5p/LOXL2 axis. Oxid Med Cell Longev. 2022;2022:9277360.
Ashour H, Farghaly ME, Khowailed AA, Aboulhoda BE, Rashed LA, Elsebaie MM, Gaber SS. Modulation of miR-192/NF-κB/ TGF-β/ E-cadherin by thymoquinone protects against diethylnitrosamine/carbon tetrachloride hepatotoxicity. Physiol Internat. 2022. https://doiorg.publicaciones.saludcastillayleon.es/10.1556/2060.2022.00163.
Gu Y, Wei X, Sun Y, Gao H, Zheng X, Wong LL, Jin L, Liu N, Hernandez B, Peplowska K, et al. miR-192-5p silencing by genetic aberrations is a key event in hepatocellular carcinomas with cancer stem cell features. Cancer Res. 2019;79(5):941–53.
Yin S, Jin W, Qiu Y, Fu L, Wang T, Yu H. Solamargine induces hepatocellular carcinoma cell apoptosis and autophagy via inhibiting LIF/miR-192-5p/CYR61/Akt signaling pathways and eliciting immunostimulatory tumor microenvironment. J Hematol Oncol. 2022;15(1):32.
Yan-Chun L, Hong-Mei Y, Zhi-Hong C, Qing H, Yan-Hong Z, Ji-Fang W. MicroRNA-192-5p promote the proliferation and metastasis of hepatocellular carcinoma cell by targeting SEMA3A. Appli Immunohistochem Mol Morphol. 2017;25(4):251–60.
Škrha P, Hořínek A, Anděl M, Škrha J. miRNA-192, miRNA-21 and miRNA-200: new pancreatic cancer markers in diabetic patients? Vnitr Lek. 2015;61(4):351–4.
Flammang I, Reese M, Yang Z, Eble JA, Dhayat SA. Tumor-Suppressive miR-192-5p Has Prognostic Value in Pancreatic Ductal Adenocarcinoma. Cancers (Basel). 2020;12(6):1693.
Zhao C, Zhang J, Zhang S, Yu D, Chen Y, Liu Q, Shi M, Ni C, Zhu M. Diagnostic and biological significance of microRNA-192 in pancreatic ductal adenocarcinoma. Oncol Rep. 2013;30(1):276–84.
Feng S, Cong S, Zhang X, Bao X, Wang W, Li H, Wang Z, Wang G, Xu J, Du B, et al. MicroRNA-192 targeting retinoblastoma 1 inhibits cell proliferation and induces cell apoptosis in lung cancer cells. Nucleic Acids Res. 2011;39(15):6669–78.
Zou P, Zhu M, Lian C, Wang J, Chen Z, Zhang X, Yang Y, Chen X, Cui X, Liu J, et al. miR-192-5p suppresses the progression of lung cancer bone metastasis by targeting TRIM44. Sci Rep. 2019;9(1):19619.
Wang T, Li W, Li H, Li W. Dysregulation of exosomal miR-192 and miR-194 expression in lung adenocarcinoma patients. Saudi J Biol Sci. 2021;28(3):1561–8.
Cao J, He Y, Liu HQ, Wang SB, Zhao BC, Cheng YS. MicroRNA 192 regulates chemo-resistance of lung adenocarcinoma for gemcitabine and cisplatin combined therapy by targeting Bcl-2. Int J Clin Exp Med. 2015;8(8):12397–403.
Jin H, Qiao F, Wang Y, Xu Y, Shang Y. Curcumin inhibits cell proliferation and induces apoptosis of human non-small cell lung cancer cells through the upregulation of miR-192-5p and suppression of PI3K/Akt signaling pathway. Oncol Rep. 2015;34(5):2782–9.
Filipska M, Skrzypski M, Czetyrbok K, Stokowy T, Stasiłojć G, Supernat A, Jassem J, Żaczek AJ, Bigda J. MiR-192 and miR-662 enhance chemoresistance and invasiveness of squamous cell lung carcinoma. Lung Cancer. 2018;118:111–8.
Li Y, Zu L, Wu H, Zhang F, Fan Y, Pan H, Du X, Guo F, Zhou Q. MiR-192/NKRF axis confers lung cancer cell chemoresistance to cisplatin via the NF-κB pathway. Thoracic cancer. 2022;13(3):430–41.
Zhang F, Li Y, Wu H, Qi K, You J, Li X, Zu L, Pan Z, Wang Y, Li Y, et al. MiR-192 confers cisplatin resistance by targeting Bim in lung cancer. Chin J Lung Cancer. 2014;17(5):384–90.
Chen ZJ, Yan YJ, Shen H, Zhou JJ, Yang GH, Liao YX, Zeng JM, Yang T. miR-192 is overexpressed and promotes cell proliferation in prostate cancer. Med Princ Pract. 2019;28(2):124–32.
Sun J, Fan Z, Lu S, Yang J, Hao T, Huo Q. MiR-192 suppresses the tumorigenicity of prostate cancer cells by targeting and inhibiting nin one binding protein. Int J Mol Med. 2016;37(2):485–92.
Park MN, Park H, Rahman MA, Kim JW, Park SS, Cho Y, Choi J, Son SR, Jang DS, Shim BS, et al. BK002 induces miR-192-5p-mediated apoptosis in castration-resistant prostate cancer cells via modulation of PI3K/CHOP. Front Oncol. 2022;12: 791365.
Zhang X, Peng Y, Huang Y, Deng S, Feng X, Hou G, Lin H, Wang J, Yan R, Zhao Y, et al. Inhibition of the miR-192/215-Rab11-FIP2 axis suppresses human gastric cancer progression. Cell Death Dis. 2018;9(7):778.
Liu J, Wen Y, Liu Z, Liu S, Xu P, Xu Y, Deng S, Hu S, Luo R, Jiang J, et al. VPS33B modulates c-Myc/p53/miR-192-3p to target CCNB1 suppressing the growth of non-small cell lung cancer. Mol Ther Nucleic Acids. 2021;23:324–35.
Pan Y, Sun Y, Liu Z, Zhang C. miR-192-5p upregulation mediates the suppression of curcumin in human NSCLC cell proliferation, migration and invasion by targeting c-Myc and inactivating the Wnt/β-catenin signaling pathway. Mol Med Rep. 2020;22(2):1594–604.
Du X, Qi F, Lu S, Li Y, Han W. Nicotine upregulates FGFR3 and RB1 expression and promotes non-small cell lung cancer cell proliferation and epithelial-to-mesenchymal transition via downregulation of miR-99b and miR-192. Biomed Pharmacother. 2018;101:656–62.
Fei Z, Wang Y, Gu Y, Xie R, Hao Q, Jiang Y. CircKIF5B promotes hepatocellular carcinoma progression by regulating the miR-192 Family/XIAP Axis. Front Oncol. 2022;12: 916246.
Gu Y, Ji F, Liu N, Zhao Y, Wei X, Hu S, Jia W, Wang XW, Budhu A, Ji J, et al. Loss of miR-192-5p initiates a hyperglycolysis and stemness positive feedback in hepatocellular carcinoma. J Exp Clin Cancer Res. 2020;39(1):268.
Sun L, Zhou X, Li Y, Chen W, Wu S, Zhang B, Yao J, Xu A. KLF5 regulates epithelial-mesenchymal transition of liver cancer cells in the context of p53 loss through miR-192 targeting of ZEB2. Cell Adh Migr. 2020;14(1):182–94.
Rokavec M, Bouznad N, Hermeking H. Paracrine induction of epithelial-mesenchymal transition between colorectal cancer cells and its suppression by a p53/miR-192/215/NID1 Axis. Cell Mol Gastroenterol Hepatol. 2019;7(4):783–802.
Song B, Wang Y, Kudo K, Gavin EJ, Xi Y, Ju J. miR-192 Regulates dihydrofolate reductase and cellular proliferation through the p53-microRNA circuit. Clin Cancer Res. 2008;14(24):8080–6.
Vajen B, Greiwe L, Schäffer V, Eilers M, Huge N, Stalke A, Schlegelberger B, Illig T, Skawran B. MicroRNA-192-5p inhibits migration of triple negative breast cancer cells and directly regulates Rho GTPase activating protein 19. Genes Chromosomes Cancer. 2021;60(11):733–42.
Dong RF, Zhuang YJ, Wang Y, Zhang ZY, Xu XZ, Mao YR, Yu JJ. Tumor suppressor miR-192-5p targets TRPM7 and inhibits proliferation and invasion in cervical cancer. Kaohsiung J Med Sci. 2021;37(8):699–708.
Zhang T, Wang C, Wang K, Liang Y, Liu T, Feng L, Yang X. RacGAP1 promotes the malignant progression of cervical cancer by regulating AP-1 via miR-192 and p-JNK. Cell Death Dis. 2022;13(7):604.
Bazavar M, Fazli J, Valizadeh A, Ma B, Mohammadi E, Asemi Z, Alemi F, Maleki M, Xing S, Yousefi B. miR-192 enhances sensitivity of methotrexate drug to MG-63 osteosarcoma cancer cells. Pathol Res Pract. 2020;216(11): 153176.
Feinberg-Gorenshtein G, Guedj A, Shichrur K, Jeison M, Luria D, Kodman Y, Ash S, Feinmesser M, Edry L, Shomron N, et al. MiR-192 directly binds and regulates Dicer1 expression in neuroblastoma. PLoS ONE. 2013;8(11): e78713.
Jin Y, Lu J, Wen J, Shen Y, Wen X. Regulation of growth of human bladder cancer by miR-192 the journal of the international society for oncodevelopmental biology and medicine. Tumour Biol. 2015;36(5):3791–7.
Song Y, Du Y, Qin C, Liang H, Yang W, Lin J, Ding M, Han J, Xu T. Gemcitabine-resistant biomarkers in bladder cancer are associated with tumor-immune microenvironment. Front Develop Biol. 2021;9: 809620.
Sun Y, Pan J, Mao S, Jin J. IL-17/miR-192/IL-17Rs regulatory feedback loop facilitates multiple myeloma progression. PLoS ONE. 2014;9(12): e114647.
Tuerxun N, Wang J, Qin YT, Zhao F, Wang H, Qu JH, Uddin MN, Hao JP. Identification of key genes and miRNA-mRNA regulatory networks associated with bone marrow immune microenvironment regulations in multiple myeloma by integrative bioinformatics analysis. Hematology. 2022;27(1):506–17.
Li Q, Luan Q, Zhu H, Zhao Y, Ji J, Wu F, Yan J. Circular RNA circ_0005774 contributes to proliferation and suppresses apoptosis of acute myeloid leukemia cells via circ_0005774/miR-192-5p/ULK1 ceRNA pathway. Biochem Biophys Res Commun. 2021;551:78–85.
Wu W, Deng J, Chen C, Ma X, Yu L, Chen L. Circ_0001602 aggravates the progression of acute myeloid leukemia by regulating the miR-192-5p/ZBTB20 axis. Hematology. 2023;28(1):2240133.
Zhang H, Zhang C, Feng R, Zhang H, Gao M, Ye L. Investigating the microRNA-mRNA regulatory network in acute myeloid leukemia. Oncol Lett. 2017;14(4):3981–8.
Khella HW, Bakhet M, Allo G, Jewett MA, Girgis AH, Latif A, Girgis H, Von Both I, Bjarnason GA, Yousef GM. miR-192, miR-194 and miR-215: a convergent microRNA network suppressing tumor progression in renal cell carcinoma. Carcinogenesis. 2013;34(10):2231–9.
Wu SY, Rupaimoole R, Shen F, Pradeep S, Pecot CV, Ivan C, Nagaraja AS, Gharpure KM, Pham E, Hatakeyama H, et al. A miR-192-EGR1-HOXB9 regulatory network controls the angiogenic switch in cancer. Nat Commun. 2016;7:11169.
Gao Y, Zhang H, Zhao S, He D, Gu C. Nanofluorescence probes to detect miR-192/Integrin alpha 1 and their correlations with retinoblastoma. J Biomed Nanotechnol. 2021;17(11):2176–85.
Honar YS, Javaher S, Soleimani M, Zarebkohan A, Farhadihosseinabadi B, Tohidfar M, Abdollahpour-Alitappeh M. Advanced stage, high-grade primary tumor ovarian cancer: a multi-omics dissection and biomarker prediction process. Sci Rep. 2023;13(1):17265.
Wang L, Peng B, Yan Y, Liu G, Yang D, Wang Q, Li Y, Mao Q, Chen Q. The tRF-3024b hijacks miR-192-5p to increase BCL-2-mediated resistance to cytotoxic T lymphocytes in esophageal squamous cell carcinoma. Int Immunopharmacol. 2024;126: 111135.
Khalafizadeh A, Hashemizadegan SD, Shokri F, Bakhshinejad B, Jabbari K, Motavaf M, Babashah S. Competitive endogenous RNA networks: decoding the role of long non-coding RNAs and circular RNAs in colorectal cancer chemoresistance. J Cell Mol Med. 2024;28(7): e18197.
Rahnama S, Bakhshinejad B, Farzam F, Bitaraf A, Ghazimoradi MH, Babashah S. Identification of dysregulated competing endogenous RNA networks in glioblastoma: a way toward improved therapeutic opportunities. Life Sci. 2021;277: 119488.
Ye M, Zhang J, Zhang J, Miao Q, Yao L, Zhang J. Curcumin promotes apoptosis by activating the p53-miR-192-5p/215-XIAP pathway in non-small cell lung cancer. Cancer Lett. 2015;357(1):196–205.
Guo Q, Jin Y, Chen X, Ye X, Shen X, Lin M, Zeng C, Zhou T, Zhang J. NF-κB in biology and targeted therapy: new insights and translational implications. Signal Transduct Target Ther. 2024;9(1):53.
Yang Q, Zhuge X, Lin W, Yu W, Zhu Y, Shi C, Shi Z. Hydrogel-based miR-192 delivery inhibits the development of hepatocellular carcinoma by suppressing the GSK3β/Wnt/β-catenin pathway. Neoplasma. 2023;70(4):555–65.
Sasaki CT, Vageli DP. miR-21, miR-155, miR-192, and miR-375 deregulations related to NF-kappaB activation in gastroduodenal fluid-induced early preneoplastic lesions of laryngeal mucosa in vivo. Neoplasia. 2016;18(6):329–38.
Doukas SG, Vageli DP, Sasaki CT. NF-κB inhibition reverses acidic bile-induced miR-21, miR-155, miR-192, miR-34a, miR-375 and miR-451a deregulations in human hypopharyngeal cells. J Cell Mol Med. 2018;22(5):2922–34.
Zhang X, Peng Y, Huang Y, Yang M, Yan R, Zhao Y, Cheng Y, Liu X, Deng S, Feng X, et al. SMG-1 inhibition by miR-192/-215 causes epithelial-mesenchymal transition in gastric carcinogenesis via activation of Wnt signaling. Cancer Med. 2018;7(1):146–56.
Zhang J, Zhao CY, Zhang SH, Yu DH, Chen Y, Liu QH, Shi M, Ni CR, Zhu MH. Upregulation of miR-194 contributes to tumor growth and progression in pancreatic ductal adenocarcinoma. Oncol Rep. 2014;31(3):1157–64.
Kandimalla R, Shimura T, Mallik S, Sonohara F, Tsai S, Evans DB, Kim SC, Baba H, Kodera Y, Von Hoff D, et al. Identification of serum miRNA signature and establishment of a nomogram for risk stratification in patients With pancreatic ductal adenocarcinoma. Ann Surg. 2022;275(1):e229–37.
Khan IA, Rashid S, Singh N, Rashid S, Singh V, Gunjan D, Das P, Dash NR, Pandey RM, Chauhan SS, et al. Panel of serum miRNAs as potential non-invasive biomarkers for pancreatic ductal adenocarcinoma. Sci Rep. 2021;11(1):2824.
Fründt T, Krause L, Hussey E, Steinbach B, Köhler D, von Felden J, Schulze K, Lohse AW, Wege H, Schwarzenbach H. Diagnostic and prognostic value of miR-16, miR-146a, miR-192 and miR-221 in exosomes of hepatocellular carcinoma and liver cirrhosis patients. Cancers (Basel). 2021;13(10):2484.
Li G, Yin J, Wu Z, Li S, He A, Sun Z. Expression level of miRNA in the peripheral blood of patients with multiple myeloma and its clinical significance. Am J Transl Res. 2021;13(5):5343–9.
Wang G, Chan ES, Kwan BC, Li PK, Yip SK, Szeto CC, Ng CF. Expression of microRNAs in the urine of patients with bladder cancer. Clin Genitourin Cancer. 2012;10(2):106–13.
Poel D, Rustenburg F, Sie D, van Essen HF, Eijk PP, Bloemena E, Elhorst Benites T, van den Berg MC, Vergeer MR, Leemans RC, et al. Expression of let-7i and miR-192 is associated with resistance to cisplatin-based chemoradiotherapy in patients with larynx and hypopharynx cancer. Oral Oncol. 2020;109: 104851.
Acknowledgements
Not applicable.
Funding
This work was supported by the Universiti Sains Malaysia, Bridging with Project No: R501-LR-RND003-0000001472-0000.
Author information
Authors and Affiliations
Contributions
SRAR and ISI contributed to search strategy, design this manuscript sections and prepared tables. YY wrote the original draft and visualized the results. YM designed, structured and edited the manuscript. MAY reviewed and edited the manuscript. All authors read and approved the final manuscript.
Corresponding authors
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors declare no competing interests.
Additional information
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
About this article
Cite this article
Yang, Y., Razak, S.R.A., Ismail, I.S. et al. Molecular mechanisms of miR-192 in cancer: a biomarker and therapeutic target. Cancer Cell Int 25, 94 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12935-025-03666-5
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12935-025-03666-5