Role of miRNA‑122 in cancer (Review)
- Authors:
- Published online on: July 18, 2024 https://doi.org/10.3892/ijo.2024.5671
- Article Number: 83
-
Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
According to Global Cancer Statistics from 2018, there were 18.1 million new cancer cases and 9.6 million deaths attributed to cancer (1). Cancer ranks as the second leading cause of death globally, following ischemic heart disease (2). Current research on cancer diagnosis, treatment and prognosis primarily focuses on genetic and epigenetic factors, such as microRNAs (miRNAs). The first miRNA was discovered in nematodes, leading to further exploration of similar endogenous miRNAs across species using RNA interference (3). miRNAs not only translate proteins but also regulate gene transcription, influencing processes such as cell differentiation and organism development (3,4). Initially transcribed by RNA polymerase II in the nucleus, primary miRNAs are processed into precursor miRNAs by the Drosha enzyme-Dgcr8 complex. After translocating to the cytoplasm, these precursor miRNAs are cleaved into ~22-nucleotide-long double-stranded miRNAs by the enzyme Dicer (5). The double strands are then unwound, and the mature miRNA strand forms an RNA-induced silencing complex, which binds to the 3'-untranslated region (3'-UTR) of target mRNA to degrade or inhibit its translation, thereby negatively regulating gene expression (6). To date, >1,000 miRNAs have been identified in humans (7). Dysregulation of miRNA, such as miRNA-122, is associated with various diseases, particularly cancer (8). miRNA-122 constitutes ~72% of the total miRNAs found in the liver (9). Its expression is altered in multiple diseases. For example, elevated levels of miRNA-122 are associated with bronchiolitis. which may progress to asthma (10). miRNA-122 is also implicated in promoting diabetic retinopathy (11). Additionally, high levels of miRNA-122 suppress the release of inflammatory factors in osteoarthritis, suggesting potential therapeutic applications (12). Numerous studies have reported abnormal expression of miRNA-122 in various types of cancer, where it modulates tumor development by targeting specific genes (8,13). However, the exact role of miRNA-122 in cancer remains elusive.
The present review aimed to summarize how miRNA-122 influences various aspects of tumor cell behavior, including proliferation, migration, metastasis, invasion, angiogenesis and apoptosis, the role of miRNA-122 in modulating responses to radiotherapy and chemotherapy in tumor cells, as well as strategies for its systemic delivery and potential utility as a biomarker.
miRNA-122 and cancer
miRNAs are classified as oncogenic or tumor-suppressive based on their effects (14). miRNA-122 is derived from a single genomic locus on human chromosome 18 (15). miRNA-122 regulates tumor cell processes such as proliferation, angiogenesis, invasion, migration and apoptosis by targeting downstream genes. Due to its ability to target a diverse array of downstream genes, including both oncogenes and tumor suppressors, miRNA-122 exerts varied roles in cancer development, serving as either an oncogene or tumor suppressor, and may exhibit dual roles in certain types of cancer (16-34). Table I summarizes the role of miRNA-122 in various types of cancer.
Non-small cell carcinoma (NSCLC)
Lung cancer includes small cell carcinoma and NSCLC, with NSCLC being the predominant form, constituting 80-85% of cases and associated with 2-year relative survival rate of ~42% (35). NSCLC cells do not express endogenous miRNA-122. miRNA-122 inhibits the PI3K/AKT signaling pathway through suppression of its target gene insulin-like growth factor 1 receptor (IGF1R). This suppression blocks PI3K and AKT phosphorylation, enhances E-cadherin expression and decreases N-cadherin and vimentin expression. This disruption impedes epithelial-mesenchymal transition (EMT), thereby inhibiting migration and invasion of NSCLC cells (17).
Long-term exposure of NSCLC cells to gefitinib leads to emergence of gefitinib-resistant A549/GR cells. As a target of miRNA-122, peroxiredoxin II (Prx II) inhibition suppresses the self-renewal and EMT of A549/GR stem cells, which is characterized by an increase in E-cadherin and decrease in vimentin expression following miRNA-122 knockout. This process involves inhibition of Hedgehog, Notch and Wnt/β-catenin signaling pathways following Prx II targeting by miRNA-122. Knockout of miRNA-122 reverses these effects (Fig. 1) (18).
Nasopharyngeal carcinoma (NPC)
NPC arises from the nasopharyngeal crypt, originating from mucosal epithelium of the nasopharynx, and is characterized by high malignancy (36). NPC cells exhibit significantly decreased expression of miRNA-122. miRNA-122 suppresses PI3K and AKT phosphorylation, inhibits the PI3K/AKT signaling pathway and decreases the expression of E-cadherin, metastasis-associated gene 1, MMP2 and tissue inhibitor of metalloproteinase 2 by targeting tripartite motif-containing protein 29. This inhibition suppresses the proliferation, migration and invasion capabilities of NPC cells (20). Moreover, the long non-coding RNA (lncRNA) DRAIC (downregulated RNA in cancer) upregulates special AT-rich binding protein 1 (SATB1) expression by binding to miRNA-122, thereby alters the configuration of the miRNA-122 binding site on SATB1 and suppressing miRNA-122 expression. This promotes the proliferation, migration and invasion of NPC cells; these effects are reversed by miRNA-122 overexpression (Fig. 2) (21).
Prostate cancer
Prostate cancer is a prevalent malignancy in male patients, accounting for ~26% of newly diagnosed cancer cases (35). Tumor cells obtain their energy supply through relatively low-yield glycolysis, which does not involve oxygen or mitochondria (37). Pyruvate kinase M2 (PKM2) serves as a key rate-limiting enzyme in glycolysis, driving tumor cell proliferation (38). In docetaxel-resistant prostate cancer cells, miRNA-122 expression is notably decreased. This leads to increased PKM2 expression, enhancing glycolysis, promoting proliferation and reducing apoptosis in these resistant cells. The mechanism involves miRNA-122 targeting and inhibiting PKM2 to suppress prostate cancer progression (39).
miRNA-122 inhibits the proliferation of prostate cancer cells by downregulating Rho-associated protein kinase 2 (ROCK2) expression (22). ROCK2, a member of the Rho family, promotes invasion and metastasis in prostate cancer (40). Additionally, silencing ROCK2 expression counteracts enzalutamide resistance in enzalutamide-resistant prostate cancer cells, leading to inhibition of cancer cell proliferation (Fig. 3) (41).
Bile duct cancer (BDC)
BDC is a malignant tumor of the epithelial cells of the bile ducts; it is insidious and the 5-year survival rate drops to 2% if distant metastasis occurs (42). Expression of miRNA-122 is notably decreased in BDC compared with normal bile duct tissue. Overexpressed miRNA-122 in BDC cells significantly inhibits tumor cell proliferation and invasion while promoting apoptosis. However, the specific mechanism by which miRNA-122 inhibits BDC progression has not been fully elucidated (43). Wu et al (44) demonstrated that overexpression of miRNA-122 in BDC cells upregulates P53 expression, thereby suppressing tumor cell proliferation and invasion while promoting apoptosis. Additionally, miRNA-122 also inhibits BDC cell migration and invasion by targeting the downstream target gene chloride intracellular channel 1 (CLIC1). lncRNA urothelial cancer associated 1 (UCA1) regulates CLIC1 expression through sponging miRNA-122 to promote BDC metastasis (Fig. 4) (19).
Bladder cancer
In 2018, 549,393 patients were diagnosed with bladder cancer worldwide and 199,922 died from the disease (1). Angiogenesis is key for tumor development, making its inhibition a potential treatment strategy. Vascular endothelial growth factor (VEGF) is associated with tumor progression (45). miRNA-122 expression is downregulated in human bladder cancer tissue. By binding to the 3'-UTR of VEGFC, miRNA-122 significantly decreases VEGFC expression, inhibiting AKT and mTOR phosphorylation. This inhibition ultimately suppresses angiogenesis, invasion, migration and proliferation of bladder cancer cells (8). Furthermore, miRNA-122 overexpression enhances sensitivity of bladder cancer cells to cisplatin and promotes cancer cell apoptosis (8).
Prior research (16) has established that the cAMP-response element-binding protein (CREB) 1 serves as a downstream target of miRNA-122. miRNA-122 directly inhibits CREB1 to suppress proliferation and invasion in bladder cancer cells (Fig. 5) (16).
Breast cancer
Breast cancer is the most common cancer in female patients and the leading cause of cancer mortality in female patients worldwide (46). lncRNA ribonuclease P RNA component H1 (RPPH1) is highly expressed in breast cancer tissue. lncRNA RPPH1 promotes the expression of downstream genes, such as PKM2 and IGF1R, by sponging miRNA-122, which further promotes proliferation of breast cancer cells; by contrast, miRNA-122 overexpression reverses this process (24). miRNA-122 overexpression enhances sensitivity of breast cancer cells to radiotherapy, thereby inhibiting tumor cell survival (23). Radioresistant breast cancer cells show significantly increased miRNA-122 expression, whereas miRNA-122 knockout decreases the survival of these cells. This mechanism may involve miRNA-122 oncogenic potential through targeting zinc finger proteins 611 (ZNF611) in radioresistant breast cancer cells (23). Therefore, in primary breast cancer cells (before radiation therapy and without radioresistance), elevated miRNA-122 expression inhibits cancer progression and serves as a tumor suppressor. Conversely, in radiotherapy-resistant breast cancer cells, elevated miRNA-122 expression can promote radiotherapy resistance, demonstrating its dual role as both a tumor suppressor and oncogene depending on the cellular context (Fig. 6).
Liver cancer
Hepatocellular carcinoma (HCC), the predominant form of liver cancer, is the fifth most common cause of cancer deaths In the United States (47). miRNA-122 disrupts mesenchymal cytoskeleton, upregulates E-cadherin and α-catenin expression and downregulates vimentin and fibronectin expression by binding to the 3'-UTR of Ras homologous gene family member A (RhoA); this triggers mesenchymal-epithelial transition, reverses EMT and thus inhibits the invasion and migration of HCC cells (48).
In a study on adriamycin resistance in HCC (49), overexpression of miRNA-122 suppressed expression of ATP-binding cassette superfamily member 2 and multidrug resistance-associated protein 1. This overexpression enhances sensitivity of HCC cells to chemotherapeutic drugs and inhibits proliferation.
Polyploidy is a balanced amplification of the genome and is common in the liver. Hepatocytes become polyploid mostly due to failure of cytoplasmic division. Approximately 30% of hepatocytes in the human liver are polyploid. Liver polyploidy prevents gene mutations in hepatocytes and decreases the formation of liver tumors (50). miRNA-122 directly targets cytoplasmic cleavage genes cut-like homeobox protein-1, RhoA, microtubule-associated protein RP/EB family member 1, IQ-containing GTPase-activating protein 1, neural precursor cell-expressed developmentally down-regulated protein 4-like and solute carrier family 25 member 34, thereby resulting in cytoplasmic cleavage failure to increase hepatic polyploidization, thus suppressing liver tumorigenesis (51). Other studies have shown that frequent ploidy reduction in polyploid hepatocytes results in predisposition to liver tumor formation, which may imply that notable reduction in hepatic polyploidy contributes to cancer development, as this leads to genetic mutations (Fig. 7) (52,53).
Colorectal cancer (CRC)
CRC accounted for 9.4% of all new cases of cancer in 2020 (54). The liver is the most important target organ for hematogenous metastasis of CRC. Liver metastasis is the main cause of death due to CRC (54). Studies have found elevated miRNA-122 expression in liver cells of metastatic CRC, contrasting high expression was not detected in CRC cells. miRNA-122 correlates negatively with cationic amino acid transporter protein 1 (CAT1) expression and can enhance liver migration by targeting CAT1 (26). In oxaliplatin-resistant CRC cell lines, miRNA-122 expression is reduced while X-linked inhibitor of apoptosis protein (XIAP) expression is increased. miRNA-122 downregulates XIAP to restore oxaliplatin sensitivity in resistant cells, thereby suppressing CRC progression (27). Thus, elevated miRNA-122 levels in untreated CRC cells may indicate potential liver metastasis, suggesting an oncogenic role. Conversely, decreased miRNA-122 expression in oxaliplatin-treated CRC cells may indicate drug resistance, highlighting its role as a tumor suppressor. In summary, miRNA-122 exhibits dual roles as both a tumor suppressor and an oncogene in CRC (Fig. 8).
Esophageal cancer
Due to changes in dietary habits and genetic factors, esophageal cancer is a significant health challenge worldwide, with overall 5-year survival rate of ~10% and a propensity for early metastasis (55). KIF22, a kinesin-like DNA-binding protein, can promote cancer progression. Kinesin superfamily protein 22 (KIF22) is prominently expressed in esophageal squamous carcinoma tissue and cells, correlating significantly with poor prognosis (28); miRNA-122 negatively regulates KIF22, downregulates the expression of cyclin G1 (CCNG1), Cyclin dependent kinase 2, N-cadherin and vimentin and upregulates p21, p27 and E-cadherin expression; this induces S phase arrest and apoptosis of esophageal squamous carcinoma cells and inhibits EMT (28) (Fig. 8). Additionally, the response elements of miRNA-122 and miRNA-143 mediated by adenoviral vectors cause tumor necrosis factor-associated apoptosis-inducing ligand (TRAIL) to be highly expressed in esophageal cancer, but not in normal cells; this selectively induces apoptosis in esophageal cancer cells and protects against the toxicity of TRAIL to the liver (56). This may be an effective approach to treat esophageal cancer and prevent liver toxicity.
Glioma
Glioma is the most common primary malignant tumors of the brain. Glioma grows invasively and often involve surrounding normal brain tissue. RUNX2, part of the RUNX transcription factor family, regulates gene expression and enhances tumor cell proliferation by binding to specific DNA sequences (57). Additionally, miRNA-122 is downregulated in glioma compared with normal tissues (29). By targeting RUNX2, miRNA-122 inhibits proliferation and migration of glioma cells (29). Moreover, miRNA-122 induces cell cycle arrest, promotes apoptosis, and decreases proliferation in transglioma cells by targeting SOX6. However, this inhibitory effect on glioma cells can be reversed by circular RNA pleiotrophin, which serves as a miRNA-122 sponge (Fig. 8) (30).
Renal cell carcinoma (RCC)
RCC, is a prevalent form of kidney cancer, constituting for 2 to 3% of all adult malignancies, with 1.8% mortality rate (1). Fan et al (31) observed elevated miRNA-122 expression in RCC cells, correlating with poor prognosis. miRNA-122 induces EMT by suppressing Dicer, a downstream target gene, thereby enhancing migration and invasion of RCC cells (31). Similarly, Nie et al (32) found increased miRNA-122 expression in RCC cells, which promotes cell proliferation, migration and invasion by targeting FOXO3. Thus, miRNA-122 plays an oncogene role in RCC (Fig. 8).
Other types of cancer
Acute myeloid leukemia (AML) is a blood cancer characterized by abnormal cell proportions due to impaired differentiation of hematopoietic stem cells (58). Zhang et al (59) reported that patients with AML with low miRNA-122 levels in their bone marrow have poorer overall survival and lower rates of complete remission compared with those with high miRNA-122 expression (59). Yang et al (60) found that miRNA-122 expression is significantly reduced in AML bone marrow compared with non-malignant tissue and high miRNA-122 levels inhibit AML cell proliferation by affecting cell cycle pathways (60). However, further research is needed to understand how miRNA-122 suppresses AML progression.
Osteosarcoma, a common bone malignancy in children and adolescents, is associated with high mortality rates (61). miRNA-122 suppresses proliferation, migration, and invasion of osteosarcoma cells by decreasing the expression of CCNG1, Bcl-w and a disintegrin and matrix metalloproteinase-10 (33). However, this inhibitory effect is reversed by miRNA-122 sponging (33). Additionally, Liu et al (62) noted varied miRNA-122 expression between different osteosarcoma cell lines; while miRNA-122 is upregulated in HOS, Saos-2 and U2OS cell lines, it is downregulated in MG-63 cells. High miRNA-122 levels inhibit the proliferation, invasion and migration of Saos-2 osteosarcoma cells, indicating its role as a tumor suppressor despite being highly expressed in this cell line (62).
Cervical cancer is a common malignant tumor affecting female patients accounting for about 3.2% of all cancers (1). According to Yang et al (34), miRNA-122 targets RAD21, a component of the cohesin complex, thereby inhibiting the PI3K/AKT signaling pathway in cervical cancer cells. This inhibition suppresses cervical cancer cell proliferation and promotes apoptosis. Elevated levels of miRNA-122 are associated with improved prognosis in patients with cervical cancer (34).
miRNA-122 as a biomarker
With technological advancements, the quantification of miRNA-122 has become precise and convenient, highlighting its potential as a biomarker. Recent studies have indicated that miRNA-122 may be valuable for diagnosing cancer, predicting prognosis and assessing treatment response (Table II) (63-67).
Biomarker for diagnosis
The expression levels of miRNA-122 in the urine of patients with clear cell renal cell carcinoma (ccRCC) show a significant 13.9-fold elevation; alongside miRNA-1271 and miRNA-15b, this may be useful in diagnosing ccRCC (66). In prostate cancer, both tissue and serum levels of miRNA-122 are notably decreased and serum miRNA-122 levels effectively distinguish patients with prostate cancer from healthy individuals (22). Additionally, a model combining six miRNAs, including miRNA-122, has superior accuracy in differentiating patients with oral squamous cell carcinoma from healthy controls compared with serum squamous cell carcinoma antigen (68). The aforementioned studies suggest that miRNA-122 has potential to be used as an additional diagnostic marker for certain types of cancer.
Biomarker for prognosis
In CRC, miRNA-122 is associated with increased risk of tumor metastasis (69). Conversely, miRNA-122 expression is associated with improved prognosis in HCC (70), AML (59) and bile duct cancer (43). Low miRNA-122 expression also correlates with poor outcomes following radical resection in liver cancer (71). Yang et al (20) found that decreased miRNA-122 expression significantly correlates with advanced tumor node metastasis stage and distant metastasis in NPC. Therefore, miRNA-122 shows promise as a useful tool for predicting cancer prognosis.
Biomarker of therapeutic response
miRNA-122 expression is indicative of treatment response and aids in tailoring effective treatment plans. Elevated miRNA-122 levels predict early resistance to transcatheter arterial chemoembolization in patients with HCC (72). Additionally, patients with HCC who are responsive to sorafenib exhibit higher miRNA-122 expression post-chemotherapy compared with non-responsive patients, underscoring its potential to predict sorafenib efficacy (65).
Application of miRNA-122 in chemotherapy and radiotherapy
Chemotherapy and radiotherapy are frequently utilized in cancer treatment; nonetheless, addressing drug resistance in patients with advanced and recurrent cancer remains a challenge. Enhancing the sensitivity of patients to chemotherapy and radiotherapy is key (73).
miRNA-122 targets XIAP to reverse oxaliplatin resistance in CRC cells (27) and also sensitizes colon cancer cells to 5-fluorouracil (5-FU) by targeting PKM2 (74). In prostate cancer, miRNA-122 enhances cell sensitivity to docetaxel via PKM2 targeting (39). Moreover, miRNA-122 decreases the expression of drug-resistant P-glycoprotein and multidrug-resistance proteins by inhibiting small ubiquitin-like modifier sentrin-specific protease 1, thus overcoming adriamycin and sorafenib resistance in HCC cells (75). miRNA-122 directly targets the Wnt/β-catenin pathway, leading to decreased expression of multidrug resistance proteins 1 and increased sensitivity of HCC cells to oxaliplatin (76).
In radiation therapy, miRNA-122 decreases the expression of stress response regulators such as survivin, apoptosis inhibitory proteins 1 and 2 and IGF1R. This mechanism induces DNA double-strand breaks and promotes apoptosis in NSCLC cells, thereby enhancing inhibition of NSCLC cell proliferation and invasion (77). Additionally, miRNA-122 improves the effectiveness of radiation therapy by decreasing IGF1R expression (78). Thus, adjusting miRNA-122 levels enhances the efficacy of chemotherapy and radiotherapy in specific types of cancer.
Systemic delivery strategies for miRNA-122
Due to the susceptibility of miRNAs to enzymatic degradation, protection from RNA hydrolases in extracellular serum is key during systemic delivery to ensure their delivery to target cells. This necessitates encapsulating miRNA in sealed carriers to prevent enzymatic hydrolysis during transportation. Therefore, effective delivery systems are essential for precise delivery of miRNA to tumor cells (79). Utilizing exosomal, viral and nanoparticle vectors for miRNA delivery can address this issue (80).
Exosome vectors
Exosomes are active vesicles secreted by cells to facilitate intercellular substance exchange and information transfer. They are commonly utilized as carriers for delivering miRNA (81). A recent study demonstrated that exosomes derived from adipose-derived mesenchymal stem cells effectively deliver miRNA-122 into HCC cells (82). This results in significant downregulation of CCNG1, disintegrin, MMP10 and IGF1R expression, enhancing the sensitivity of HCC cells to 5-FU and sorafenib (82).
Viral vectors
Viral vectors widely used include lentiviruses, retroviruses, adenoviruses, and adeno-associated viruses (AAVs) (83,84). In an in vivo mouse xenograft model, tumor growth of human HCC is notably suppressed by AAV3 along with miRNA-122 and miRNA-26a lentiviral delivery systems (85). Additionally, tumor necrosis factor-associated apoptosis-inducing ligands selectively expressed in osteosarcoma cells through adenoviral vectors of miRNA-122 and miRNA-34 promote apoptosis and inhibit cell proliferation (86). Notably, linking and cloning miRNA-21 and pre-miRNA-122 sequences into viral-like particle expression vectors and delivering them into HCC cells inhibits proliferation, migration and invasion of HCC cells and promotes apoptosis (87). While advantages such as easy cell access and high expression of introduced genes are offered by viral vectors, concerns about immunogenicity and potential promotion of gene mutagenesis have prompted a search for alternatives (88).
Nanocarriers
Nanocarriers offer advantages such as low toxicity, minimal immunogenicity, high permeability and non-integration of genes into the host cell genome, making them essential alternatives to viral vectors (89). Sendi et al (90) developed a galactose-targeted lipid calcium phosphate nanoformulation noted for its stability and efficient delivery of miRNA-122 to CRC liver metastatic hepatocytes. This formulation effectively prevents liver metastasis and prolongs survival in CRC mouse models without significant toxicity (90). Additionally, an ultrasound-triggered, phase-transitioning cationic nanodroplet delivers miRNA-122 to HCC cells, resulting in a significant increase in miRNA-122 expression and substantial inhibition of HCC cell proliferation, migration, and invasion (91). Zeng et al (92) created graphene-P-gluoprotein loaded with miR-122-InP@ZnS quantum dot nanocomposites, demonstrating effective delivery of miRNA-122 to multidrug-resistant HCC cells and induction of apoptosis (92). Furthermore, Zhang et al (93) utilized amphiphilic gemcitabine-oleic acid prodrug nanoparticles for encapsulating miRNA-122, effectively targeting HCC cells and significantly inhibiting proliferation in xenograft nude mice (93).
Clinical implications and limitations
Cell proliferation, metastasis and apoptosis are pivotal in tumor growth and development. The hallmark of cancer is aberrant cell proliferation, with the regulation of cell proliferation and survival being crucial in tumor formation (94). Thus, strategies that inhibit tumor proliferation and enhance apoptosis are key in cancer treatment. Additionally, cell metastasis significantly impacts cancer prognosis, as its occurrence signifies disease progression. Therefore, inhibiting tumor cell metastasis is a key therapeutic approach in cancer management (95).
miRNA-122 has garnered notable attention in cancer and liver disease research (96,97). Upregulation of miRNA-122 expression inhibits self-renewal, EMT and angiogenic capacity of NSCLC tumor stem cells (18). In HCC, elevated miRNA-122 levels enhance sensitivity to chemotherapy, suppress cell proliferation, metastasis and invasion and promote liver polyploidization (51,76). Conversely, as an oncogene, high miRNA-122 expression promotes migration and invasion in RCC (31). miRNA-122 functions by targeting various genes, exerting either oncogenic or tumor-suppressive effects. Moreover, miRNA-122 serves as a biomarker for diagnosing, prognosticating and monitoring response to treatment, thereby enhancing sensitivity to radiotherapy and chemotherapy (66,69). Systemic delivery strategies for miRNA-122 circumvent enzymatic hydrolysis, ensuring precise delivery to tumor cells and enhancing its stability during cancer treatment (90). Consequently, targeting miRNA-122 presents a promising avenue for future cancer therapies, although challenges remain. Determining specific diagnostic and prognostic thresholds for miRNA-122 given its varied roles is a primary challenge. Additionally, understanding interactions between miRNA-122 downstream target genes and cancer signaling pathways requires further investigation. Furthermore, while research on miRNA-122 and cancer treatment shows potential, its clinical translation remains incomplete, necessitating large-scale controlled experiments and clinical trials to elucidate its full mechanistic role in cancer.
Conclusion
The present review outlines the multifaceted role of miRNA-122 in various types of cancer. Dysregulation of miRNA-122 is observed in different types of cancer, influencing pathways key to tumor cell behavior such as proliferation, angiogenesis, differentiation, metastasis and apoptosis. Mechanistically, miRNA-122 modulates complex signaling pathways by targeting various genes, although certain lncRNAs and circ RNAs may serve as upstream regulators to regulate miRNA-122 expression. This complexity underscores miRNA-122 as a promising therapeutic target for cancer treatment. However, it lacks a universal or primary pathway across different types of cancer. Furthermore, miRNA-122 shows potential as a biomarker for cancer diagnosis, prognosis and treatment response, enhancing sensitivity to radiotherapy and chemotherapy in specific types of cancer. The systemic delivery of miRNA-122 holds promise in advancing cancer therapy. These insights suggest new avenues for research in cancer diagnosis and treatment.
Availability of data and materials
Not applicable.
Authors' contributions
JZ wrote the manuscript and constructed figures and tables. XD and RD revised the manuscript. ZC performed the literature review. LW conceptualized the study. Data authentication is not applicable. All authors have read and approved the final manuscript.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Acknowledgements
Not applicable.
Funding
The present study was supported by Taizhou Hailing District Science and Technology Development Program Project (grant no. HLKF-2019-4) and Guangxi University of Traditional Chinese Medicine Gui School of Chinese Medicine Inheritance Innovation Team Grant (grant no. 2022B004).
References
Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018. View Article : Google Scholar : PubMed/NCBI | |
Mattiuzzi C and Lippi G: Current cancer epidemiology. J Epidemiol Glob Health. 9:217–222. 2019. View Article : Google Scholar : PubMed/NCBI | |
Britton C, Laing R and Devaney E: Small RNAs in parasitic nematodes-forms and functions. Parasitology. 147:855–864. 2020. View Article : Google Scholar | |
Morales-Martínez M and Vega MI: Role of MicroRNA-7 (MiR-7) in cancer physiopathology. Int J Mol Sci. 23:90912022. View Article : Google Scholar : PubMed/NCBI | |
Hebbar S, Panzade G, Vashisht AA, Wohlschlegel JA, Veksler-Lublinsky I and Zinovyeva AY: Functional identification of microRNA-centered complexes in C: elegans. Sci Rep. 12:71332022. View Article : Google Scholar | |
Hill M and Tran N: miRNA interplay: Mechanisms and consequences in cancer. Dis Model Mech. 14:dmm0476622021. View Article : Google Scholar : PubMed/NCBI | |
Chen Y and Wang X: miRDB: An online database for prediction of functional microRNA targets. Nucleic Acids Res. 48(D1): D127–D131. 2020. View Article : Google Scholar : | |
Wang Y, Xing QF, Liu XQ, Guo ZJ, Li CY and Sun G: MiR-122 targets VEGFC in bladder cancer to inhibit tumor growth and angiogenesis. Am J Transl Res. 8:3056–3066. 2016.PubMed/NCBI | |
Chun KH: Molecular Targets and signaling pathways of microRNA-122 in hepatocellular carcinoma. Pharmaceutics. 14:13802022. View Article : Google Scholar : PubMed/NCBI | |
Collison AM, Sokulsky LA, Kepreotes E, Pereira de Siqueira A, Morten M, Edwards MR, Walton RP, Bartlett NW, Yang M, Nguyen TH, et al: miR-122 promotes virus-induced lung disease by targeting SOCS1. JCI Insight. 6:e1279332021. View Article : Google Scholar : PubMed/NCBI | |
Wang M, Zheng H, Zhou X, Zhang J and Shao G: miR-122 promotes diabetic retinopathy through targeting TIMP3. Anim Cells Syst (Seoul). 24:275–281. 2020. View Article : Google Scholar : PubMed/NCBI | |
Scott KM, Cohen DJ, Boyan BD and Schwartz Z: miR-122 and the WNT/β-catenin pathway inhibit effects of both interleukin-1β and tumor necrosis factor-α in articular chondrocytes in vitro. J Cell Biochem. 123:1053–1063. 2022. View Article : Google Scholar : PubMed/NCBI | |
Li A, Wu J, Zhai A, Qian J, Qian J, Wang X, Qaria MA, Zhang Q, Li Y, Fang Y, Kao W, et al: HBV triggers APOBEC2 expression through miR-122 regulation and affects the proliferation of liver cancer cells. Int J Oncol. 55:1137–1148. 2019.PubMed/NCBI | |
Smolarz B, Durczyński A, Romanowicz H, Szyłło K and Hogendorf P: miRNAs in cancer (review of literature). Int J Mol Sci. 23:28052022. View Article : Google Scholar : PubMed/NCBI | |
Liu Y, Song JW, Lin JY, Miao R and Zhong JC: Roles of MicroRNA-122 in cardiovascular fibrosis and related diseases. Cardiovasc Toxicol. 20:463–473. 2020. View Article : Google Scholar : PubMed/NCBI | |
Guo L, Yin M and Wang Y: CREB1, a direct target of miR-122, promotes cell proliferation and invasion in bladder cancer. Oncol Lett. 16:3842–3848. 2018.PubMed/NCBI | |
Qin H, Sha J, Jiang C, Gao X, Qu L, Yan H, Xu T, Jiang Q and Gao H: miR-122 inhibits metastasis and epithelial-mesenchymal transition of non-small-cell lung cancer cells. Onco Targets Ther. 8:3175–3184. 2015.PubMed/NCBI | |
Chandimali N, Huynh DL, Zhang JJ, Lee JC, Yu DY, Jeong DK and Kwon T: MicroRNA-122 negatively associates with peroxiredoxin-II expression in human gefitinib-resistant lung cancer stem cells. Cancer Gene Ther. 26:292–304. 2019. View Article : Google Scholar : | |
Kong L, Wu Q, Zhao L, Ye J, Li N and Yang H: Upregulated lncRNA-UCA1 contributes to metastasis of bile duct carcinoma through regulation of miR-122/CLIC1 and activation of the ERK/MAPK signaling pathway. Cell Cycle. 18:1212–1228. 2019. View Article : Google Scholar : PubMed/NCBI | |
Yang Y, Li Q and Guo L: MicroRNA-122 acts as tumor suppressor by targeting TRIM29 and blocking the activity of PI3K/AKT signaling in nasopharyngeal carcinoma in vitro. Mol Med Rep. 17:8244–8252. 2018.PubMed/NCBI | |
Liao B, Wang Z, Zhu Y, Wang M and Liu Y: Long noncoding RNA DRAIC acts as a microRNA-122 sponge to facilitate nasopharyngeal carcinoma cell proliferation, migration and invasion via regulating SATB1. Artif Cells Nanomed Biotechnol. 47:3585–3597. 2019. View Article : Google Scholar : PubMed/NCBI | |
Liu H, Hou T, Ju W, Xing Y, Zhang X and Yang J: MicroRNA-122 downregulates Rho-associated protein kinase 2 expression and inhibits the proliferation of prostate carcinoma cells. Mol Med Rep. 19:3882–3888. 2019.PubMed/NCBI | |
Perez-Añorve IX, Gonzalez-De la Rosa CH, Soto-Reyes E, Beltran-Anaya FO, Del Moral-Hernandez O, Salgado-Albarran M, Angeles-Zaragoza O, Gonzalez-Barrios JA, Landero-Huerta DA and Chavez-Saldaña M: New insights into radioresistance in breast cancer identify a dual function of miR-122 as a tumor suppressor and oncomiR. Mol Oncol. 13:1249–1267. 2019. View Article : Google Scholar : PubMed/NCBI | |
Zhang Y and Tang L: Inhibition of breast cancer cell proliferation and tumorigenesis by long non-coding RNA RPPH1 down-regulation of miR-122 expression. Cancer Cell Int. 17:1092017. View Article : Google Scholar : PubMed/NCBI | |
Li XN, Yang H and Yang T: miR-122 inhibits hepatocarcinoma cell progression by targeting LMNB2. Oncol Res. 28:41–49. 2020. View Article : Google Scholar | |
Iino I, Kikuchi H, Miyazaki S, Hiramatsu Y, Ohta M, Kamiya K, Kusama Y, Baba S, Setou M and Konno H: Effect of miR-122 and its target gene cationic amino acid transporter 1 on colorectal liver metastasis. Cancer Sci. 104:624–630. 2013. View Article : Google Scholar : PubMed/NCBI | |
Hua Y, Zhu Y, Zhang J, Zhu Z, Ning Z, Chen H, Liu L, Chen Z and Meng Z: miR-122 targets X-linked inhibitor of apoptosis protein to sensitize oxaliplatin-resistant colorectal cancer cells to oxaliplatin-mediated cytotoxicit. Cell Physiol Biochem. 51:2148–2159. 2018. View Article : Google Scholar | |
Wang J, Yu PY, Yu JP, Luo JD, Sun ZQ, Sun F, Kong Z and Wang JL: KIF22 promotes progress of esophageal squamous cell carcinoma cells and is negatively regulated by miR-122. Am J Transl Res. 13:4152–4166. 2021.PubMed/NCBI | |
Ding CQ, Deng WS, Yin XF and Ding XD: MiR-122 inhibits cell proliferation and induces apoptosis by targeting runt-related transcription factors 2 in human glioma. Eur Rev Med Pharmacol Sci. 22:4925–4933. 2018.PubMed/NCBI | |
Chen C, Deng L, Nie DK, Jia F, Fu LS, Wan ZQ and Lan Q: Circular RNA Pleiotrophin promotes carcinogenesis in glioma via regulation of microRNA-122/SRY-box transcription factor 6 axis. Eur J Cancer Prev. 29:165–173. 2020. View Article : Google Scholar | |
Fan Y, Ma X, Li H, Gao Y, Huang Q, Zhang Y, Bao X, Du Q, Luo G, Liu K, et al: miR-122 promotes metastasis of clear-cell renal cell carcinoma by downregulating Dicer. Int J Cancer. 142:547–560. 2018. View Article : Google Scholar | |
Nie W, Ni D, Ma X, Zhang Y, Gao Y, Peng C and Zhang X: miR-122 promotes proliferation and invasion of clear cell renal cell carcinoma by suppressing Forkhead box O3. Int J Oncol. 54:559–571. 2019. | |
Ma J, Wu Q, Zhang Y, Li J, Yu Y, Pan Q and Sun F: MicroRNA sponge blocks the tumor-suppressing functions of microRNA-122 in human hepatoma and osteosarcoma cells. Oncol Rep. 32:2744–2752. 2014. View Article : Google Scholar : PubMed/NCBI | |
Yang Y, Liu Y, Liu W, Li C, Liu Y, Hu W and Song H: miR-122 inhibits the cervical cancer development by targeting the oncogene RAD21. Biochem Genet. 60:303–314. 2022. View Article : Google Scholar | |
Siegel RL, Miller KD, Fuchs HE and Jemal A: Cancer statistics, 2021. CA Cancer J Clin. 71:7–33. 2021. View Article : Google Scholar : PubMed/NCBI | |
Bossi P, Chan AT, Licitra L, Trama A, Orlandi E, Hui EP, Halámková J, Mattheis S, Baujat B, Hardillo J, et al: Nasopharyngeal carcinoma: ESMO-EURACAN clinical practice guidelines for diagnosis, treatment and follow-up†. Ann Oncol. 32:452–465. 2021. View Article : Google Scholar | |
Chelakkot C, Chelakkot VS, Shin Y and Song K: Modulating glycolysis to improve cancer therapy. Int J Mol Sci. 24:26062023. View Article : Google Scholar : PubMed/NCBI | |
Li TE, Wang S, Shen XT, Zhang Z, Chen M, Wang H, Zhu Y, Xu D, Hu BY, Wei R, et al: PKM2 drives hepatocellular carcinoma progression by inducing immunosuppressive microenvironment. Front Immunol. 11:5899972020. View Article : Google Scholar : PubMed/NCBI | |
Zhu Z, Tang G and Yan J: MicroRNA-122 regulates docetaxel resistance of prostate cancer cells by regulating PKM2. Exp Ther Med. 20:2472020. View Article : Google Scholar : PubMed/NCBI | |
Gong H, Zhou L, Khelfat L, Qiu G, Wang Y, Mao K and Chen W: Rho-associated protein kinase (ROCK) promotes proliferation and migration of PC-3 and DU145 prostate cancer cells by targeting LIM kinase 1 (LIMK1) and matrix metalloproteinase-2 (MMP-2). Med Sci Monit. 25:3090–3099. 2019. View Article : Google Scholar : PubMed/NCBI | |
Chen X, Yin L, Xu H, Rong J, Feng M, Jiang D and Bai Y: Knockdown of RhoA expression reverts enzalutamide resistance via the P38 MAPK pathway in castration-resistant prostate cancer. Recent Pat Anticancer Drug Discov. 18:92–99. 2023. View Article : Google Scholar | |
Sato K, Glaser S, Alvaro D, Meng F, Francis H and Alpini G: Cholangiocarcinoma: Novel therapeutic targets. Expert Opin Ther Targets. 24:345–357. 2020. View Article : Google Scholar : PubMed/NCBI | |
Liu N, Jiang F, He TL, Zhang JK, Zhao J, Wang C, Jiang GX, Cao LP, Kang PC, Zhong XY, et al: The roles of MicroRNA-122 overexpression in inhibiting proliferation and invasion and stimulating apoptosis of human cholangiocarcinoma cells. Sci Rep. 5:165662015. View Article : Google Scholar : PubMed/NCBI | |
Wu C, Zhang J, Cao X, Yang Q and Xia D: Effect of Mir-122 on human cholangiocarcinoma proliferation, invasion, and apoptosis through P53 expression. Med Sci Monit. 22:2685–2690. 2016. View Article : Google Scholar : PubMed/NCBI | |
Haibe Y, Kreidieh M, El Hajj H, Khalifeh I, Mukherji D, Temraz S and Shamseddine A: Resistance mechanisms to anti-angiogenic therapies in cancer. Front Oncol. 10:2212020. View Article : Google Scholar : PubMed/NCBI | |
Wilkinson L and Gathani T: Understanding breast cancer as a global health concern. Br J Radiol. 95:202110332022. View Article : Google Scholar : | |
Anwanwan D, Singh SK, Singh S, Saikam V and Singh R: Challenges in liver cancer and possible treatment approaches. Biochim Biophys Acta Rev Cancer. 1873:1883142020. View Article : Google Scholar : | |
Wang SC, Lin XL, Li J, Zhang TT, Wang HY, Shi JW, Yang S, Zhao WT, Xie RY, Wei F, et al: MicroRNA-122 triggers mesenchymal-epithelial transition and suppresses hepatocellular carcinoma cell motility and invasion by targeting RhoA. PloS One. 9:e1013302014. View Article : Google Scholar : PubMed/NCBI | |
Yahya SMM, Fathy SA, El-Khayat ZA, El-Toukhy SE, Hamed AR, Hegazy MGA and Nabih HK: Possible role of microRNA-122 in Modulating multidrug resistance of hepatocellular carcinoma. Indian J Clin Biochem. 33:21–30. 2018. View Article : Google Scholar : PubMed/NCBI | |
Duncan AW: Hepatocyte ploidy modulation in liver cancer. EMBO Rep. 21:e519222020. View Article : Google Scholar : PubMed/NCBI | |
Hsu SH, Delgado ER, Otero PA, Teng KY, Kutay H, Meehan KM, Moroney JB, Monga JK, Hand NJ, Friedman JR, et al: MicroRNA-122 regulates polyploidization in the murine liver. Hepatology. 64:599–615. 2016. View Article : Google Scholar : PubMed/NCBI | |
Matsumoto T, Wakefield L, Peters A, Peto M, Spellman P and Grompe M: Proliferative polyploid cells give rise to tumors via ploidy reduction. Nat Commun. 12:6462021. View Article : Google Scholar : PubMed/NCBI | |
Matsumoto T: Implications of polyploidy and ploidy alterations in hepatocytes in liver injuries and cancers. Int J Mol Sci. 23:94092022. View Article : Google Scholar : PubMed/NCBI | |
Wang F, Long J, Li L, Wu ZX, Da TT, Wang XQ, Huang C, Jiang YH, Yao XQ, Ma HQ, et al: Single-cell and spatial transcriptome analysis reveals the cellular heterogeneity of liver metastatic colorectal cancer. Sci Adv. 9:eadf54642023. View Article : Google Scholar : PubMed/NCBI | |
Huang FL and Yu SJ: Esophageal cancer: Risk factors, genetic association, and treatment. Asian J Surg. 41:210–215. 2018. View Article : Google Scholar | |
Zhou K, Yan Y and Zhao S: Esophageal cancer-selective expression of TRAIL mediated by MREs of miR-143 and miR-122. Tumour Biology: Tumour Biol. 35:5787–5795. 2014. View Article : Google Scholar : PubMed/NCBI | |
Samarakkody AS, Shin NY and Cantor AB: Role of RUNX family transcription factors in DNA damage response. Mol Cells. 43:99–106. 2020.PubMed/NCBI | |
DiNardo CD, Erba HP, Freeman SD and Wei AH: Acute myeloid leukaemia. Lancet. 401:2073–2086. 2023. View Article : Google Scholar : PubMed/NCBI | |
Zhang TJ, Qian Z, Wen XM, Zhou JD, Li XX, Xu ZJ, Ma JC, Zhang ZH, Lin J and Qian J: Lower expression of bone marrow miR-122 is an independent risk factor for overall survival in cytogenetically normal acute myeloid leukemia. Pathol Res Pract. 214:896–901. 2018. View Article : Google Scholar : PubMed/NCBI | |
Yang J, Yuan Y, Yang X, Hong Z and Yang L: Decreased expression of microRNA-122 is associated with an unfavorable prognosis in childhood acute myeloid leukemia and function analysis indicates a therapeutic potential. Pathol Res Pract. 213:1166–1172. 2017. View Article : Google Scholar : PubMed/NCBI | |
Wang Y, Wang C, Xia M, Tian Z, Zhou J, Berger JM, Zhang XH and Xiao H: Engineering small-molecule and protein drugs for targeting bone tumors. Mol Ther. 32:1219–1237. 2024. View Article : Google Scholar : PubMed/NCBI | |
Liu B, Yao S and Zhou J: Micro-RNA 122 and micro-RNA 96 affected human osteosarcoma biological behavior and associated with prognosis of patients with osteosarcoma. Biosci Rep. 40:BSR202015292020. View Article : Google Scholar : PubMed/NCBI | |
Sun L, Liu X, Pan B, Hu X, Zhu Y, Su Y, Guo Z, Zhang G, Xu M, Xu X, et al: Serum exosomal miR-122 as a potential diagnostic and prognostic biomarker of colorectal cancer with liver metastasis. J Cancer. 11:630–637. 2020. View Article : Google Scholar : PubMed/NCBI | |
Tang Y, Zhao S, Wang J, Li D, Ren Q and Tang Y: Plasma miR-122 as a potential diagnostic and prognostic indicator in human glioma. Neurol Sci. 38:1087–1092. 2017. View Article : Google Scholar : PubMed/NCBI | |
Zhan G, Jiang H, Yang R and Yang K: miR-122 and miR-197 expressions in hepatic carcinoma patients before and after chemotherapy and their effect on patient prognosis. Am J Transl Res. 13:6731–6737. 2021.PubMed/NCBI | |
Cochetti G, Cari L, Nocentini G, Maulà V, Suvieri C, Cagnani R, Rossi De Vermandois JA and Mearini E: Detection of urinary miRNAs for diagnosis of clear cell renal cell carcinoma. Sci Rep. 10:212902020. View Article : Google Scholar : PubMed/NCBI | |
Chen Q, Ge X, Zhang Y, Xia H, Yuan D, Tang Q, Chen L, Pang X, Leng W and Bi F: Plasma miR-122 and miR-192 as potential novel biomarkers for the early detection of distant metastasis of gastric cancer. Oncol Rep. 31:1863–1870. 2014. View Article : Google Scholar : PubMed/NCBI | |
Nakamura K, Hiyake N, Hamada T, Yokoyama S, Mori K, Yamashiro K, Beppu M, Sagara Y, Sagara Y and Sugiura T: Circulating microRNA panel as a potential novel biomarker for oral squamous cell carcinoma diagnosis. Cancers (Basel). 13:4492021. View Article : Google Scholar : PubMed/NCBI | |
Maierthaler M, Benner A, Hoffmeister M, Surowy H, Jansen L, Knebel P, Chang-Claude J, Brenner H and Burwinkel B: Plasma miR-122 and miR-200 family are prognostic markers in colorectal cancer. Int J Cancer. 140:176–187. 2017. View Article : Google Scholar | |
Deng P, Li M and Wu Y: The predictive efficacy of serum exosomal microRNA-122 and microRNA-148a for hepatocellular carcinoma based on smart healthcare. J Healthc Eng. 2022:59145412022. View Article : Google Scholar : PubMed/NCBI | |
Ha SY, Yu JI, Choi C, Kang SY, Joh JW, Paik SW, Kim S, Kim M, Park HC and Park CK: Prognostic significance of miR-122 expression after curative resection in patients with hepatocellular carcinoma. Sci Rep. 9:147382019. View Article : Google Scholar : PubMed/NCBI | |
Kim SS, Nam JS, Cho HJ, Won JH, Kim JW, Ji JH, Yang MJ, Park JH, Noh CK, Shin SJ, et al: Plasma micoRNA-122 as a predictive marker for treatment response following transarterial chemoembolization in patients with hepatocellular carcinoma. J Gastroenterol Hepatol. 32:199–207. 2017. View Article : Google Scholar | |
Wang S, Liu Y, Feng Y, Zhang J, Swinnen J, Li Y and Ni Y: A review on curability of cancers: More efforts for novel therapeutic options are needed. Cancers (Basel). 11:17822019. View Article : Google Scholar : PubMed/NCBI | |
He J, Xie G, Tong J, Peng Y, Huang H, Li J, Wang N and Liang H: Overexpression of microRNA-122 re-sensitizes 5-FU-resistant colon cancer cells to 5-FU through the inhibition of PKM2 in vitro and in vivo. Cell Biochem Biophys. 70:1343–1350. 2014. View Article : Google Scholar : PubMed/NCBI | |
Dai J, Hao Y, Chen X, Yu Q and Wang B: miR-122/SENP1 axis confers stemness and chemoresistance to liver cancer through Wnt/β-catenin signaling. Oncol Lett. 26:3902023. View Article : Google Scholar | |
Cao F and Yin LX: miR-122 enhances sensitivity of hepatocellular carcinoma to oxaliplatin via inhibiting MDR1 by targeting Wnt/β-catenin pathway. Exp Mol Pathol. 106:34–43. 2019. View Article : Google Scholar | |
Ma D, Jia H, Qin M, Dai W, Wang T, Liang E, Dong G, Wang Z, Zhang Z and Feng F: MiR-122 induces radiosensitization in non-small cell lung cancer cell line. Int J Mol Sci. 16:22137–22150. 2015. View Article : Google Scholar : PubMed/NCBI | |
Xu G, Bu S, Wang X and Ge H: MiR-122 radiosensitize hepatocellular carcinoma cells by suppressing cyclin G1. Int J Radiat Biol. 98:11–17. 2022. View Article : Google Scholar | |
Zhao ZL, Liu C, Wang QZ, Wu HW and Zheng JW: Engineered exosomes for targeted delivery of miR-187-3p suppress the viability of hemangioma stem cells by targeting notch signaling. Ann Transl Med. 10:6212022. View Article : Google Scholar : PubMed/NCBI | |
Wu L, Zhou W, Lin L, Chen A, Feng J, Qu X, Zhang H and Yue J: Delivery of therapeutic oligonucleotides in nanoscale. Bioact Mater. 7:292–323. 2021.PubMed/NCBI | |
Liang Y, Xu X, Li X, Xiong J, Li B, Duan L, Wang D and Xia J: Chondrocyte-targeted MicroRNA delivery by engineered exosomes toward a cell-free osteoarthritis therapy. ACS Appl Mater Interfaces. 12:36938–36947. 2020. View Article : Google Scholar : PubMed/NCBI | |
Lou G, Song X, Yang F, Wu S, Wang J, Chen Z and Liu Y: Exosomes derived from miR-122-modified adipose tissue-derived MSCs increase chemosensitivity of hepatocellular carcinoma. J Hematol Oncol. 8:1222015. View Article : Google Scholar : PubMed/NCBI | |
Sweef O, Zaabout E, Bakheet A, Halawa M, Gad I, Akela M, Tousson E, Abdelghany A and Furuta S: Unraveling therapeutic opportunities and the diagnostic potential of microRNAs for human lung cancer. Pharmaceutics. 15:20612023. View Article : Google Scholar : PubMed/NCBI | |
Li X, Le Y, Zhang Z, Nian X, Liu B and Yang X: Viral vector-based gene therapy. Int J Mol Sci. 24:77362023. View Article : Google Scholar : PubMed/NCBI | |
Yin L, Keeler GD, Zhang Y, Hoffman BE, Ling C, Qing K and Srivastava A: AAV3-miRNA vectors for growth suppression of human hepatocellular carcinoma cells in vitro and human liver tumors in a murine xenograft model in vivo. Gene Ther. 28:422–434. 2021. View Article : Google Scholar : | |
Xiao F, Chen J, Lian C, Han P and Zhang C: Tumor necrosis factor-related apoptosis-inducing ligand induces cytotoxicity specific to osteosarcoma by microRNA response elements. Mol Med Rep. 11:739–745. 2015. View Article : Google Scholar | |
Zhang J, Li D, Zhang R, Peng R and Li J: Delivery of microRNA-21-sponge and pre-microRNA-122 by MS2 virus-like particles to therapeutically target hepatocellular carcinoma cells. Exp Biol Med (Maywood). 246:2463–2472. 2021. View Article : Google Scholar : PubMed/NCBI | |
Shui M, Chen Z, Chen Y, Yuan Q, Li H, Vong CT, Farag MA and Wang S: Engineering polyphenol-based carriers for nucleic acid delivery. Theranostics. 13:3204–3223. 2023. View Article : Google Scholar : PubMed/NCBI | |
Yan Y, Liu XY, Lu A, Wang XY, Jiang LX and Wang JC: Non-viral vectors for RNA delivery. J Control Release. 342:241–279. 2022. View Article : Google Scholar : PubMed/NCBI | |
Sendi H, Yazdimamaghani M, Hu M, Sultanpuram N, Wang J, Moody AS, McCabe E, Zhang J, Graboski A, Li L, et al: Nanoparticle delivery of miR-122 inhibits colorectal cancer liver metastasis. Cancer Res. 82:105–113. 2022. View Article : Google Scholar : | |
Guo H, Xu M, Cao Z, Li W, Chen L, Xie X, Wang W and Liu J: Ultrasound-assisted miR-122-loaded polymeric nanodroplets for hepatocellular carcinoma gene therapy. Mol Pharm. 17:541–553. 2020. | |
Zeng X, Yuan Y, Wang T, Wang H, Hu X, Fu Z, Zhang G, Liu B and Lu G: Targeted imaging and induction of apoptosis of drug-resistant hepatoma cells by miR-122-loaded graphene-InP nanocompounds. J Nanobiotechnology. 15:92017. View Article : Google Scholar : PubMed/NCBI | |
Zhang HT, Sun J, Yan Y, Cui SH, Wang H, Wang CH, Qiu C, Chen X, Ding JS, Qian HG, et al: Encapsulated microRNA by gemcitabine prodrug for cancer treatment. J Control Release. 316:317–330. 2019. View Article : Google Scholar : PubMed/NCBI | |
Tang Z, Xu Z, Zhu X and Zhang J: New insights into molecules and pathways of cancer metabolism and therapeutic implications. Cancer Commun (Lond). 41:16–36. 2021. View Article : Google Scholar | |
Novikov NM, Zolotaryova SY, Gautreau AM and Denisov EV: Mutational drivers of cancer cell migration and invasion. Br J Cancer. 124:102–114. 2021. View Article : Google Scholar : | |
Hsu KH, Wei CW, Su YR, Chou T, Lin YL, Yang FC, Tsou AP, Hsu CL, Tseng PH, Chen NJ, et al: Upregulation of RelB in the miR-122 knockout mice contributes to increased levels of proinflammatory chemokines/cytokines in the liver and macrophages. Immunol Lett. 226:22–30. 2020. View Article : Google Scholar : PubMed/NCBI | |
Nabih HK: The significance of HCV viral load in the incidence of HCC: A correlation between Mir-122 and CCL2. J Gastrointest Cancer. 51:412–417. 2020. View Article : Google Scholar |