Next Article in Journal
Resolution of RHCE Haplotype Ambiguities in Transfusion Settings
Previous Article in Journal
Toxicity of the New Psychoactive Substance (NPS) Clephedrone (4-Chloromethcathinone, 4-CMC): Prediction of Toxicity Using In Silico Methods for Clinical and Forensic Purposes
Previous Article in Special Issue
Serum microRNA Profiles and Pathways in Hepatitis B-Associated Hepatocellular Carcinoma: A South African Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

How MicroRNAs Command the Battle against Cancer

1
370 Heritage Medical Research Center, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2S2, Canada
2
Department of Laboratory Medicine and Pathology (5B4. 09), University of Alberta, Edmonton, AB T6G 2B7, Canada
3
Division of Anatomical Pathology, Children’s Hospital of Eastern Ontario (CHEO), University of Ottawa, 401 Smyth Road, Ottawa, ON K1H 8L1, Canada
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(11), 5865; https://doi.org/10.3390/ijms25115865
Submission received: 24 April 2024 / Revised: 23 May 2024 / Accepted: 26 May 2024 / Published: 28 May 2024
(This article belongs to the Special Issue MicroRNAs in Cancer Therapy 2.0)

Abstract

:
MicroRNAs (miRNAs) are small RNA molecules that regulate more than 30% of genes in humans. Recent studies have revealed that miRNAs play a crucial role in tumorigenesis. Large sets of miRNAs in human tumors are under-expressed compared to normal tissues. Furthermore, experiments have shown that interference with miRNA processing enhances tumorigenesis. Multiple studies have documented the causal role of miRNAs in cancer, and miRNA-based anticancer therapies are currently being developed. This review primarily focuses on two key points: (1) miRNAs and their role in human cancer and (2) the regulation of tumor suppressors by miRNAs. The review discusses (a) the regulation of the tumor suppressor p53 by miRNA, (b) the critical role of the miR-144/451 cluster in regulating the Itch-p63-Ago2 pathway, and (c) the regulation of PTEN by miRNAs. Future research and the perspectives of miRNA in cancer are also discussed. Understanding these pathways will open avenues for therapeutic interventions targeting miRNA regulation.
Keywords:
miRNA; p53; p63; PTEN; therapy

Graphical Abstract

1. miRNAs

MicroRNAs (miRNAs) are noncoding RNA molecules that regulate gene expression at transcription and translation levels [1,2,3,4]. They are 19–25 nucleotides (nt) long, with an average length of 22. The first miRNA, lin-4, was discovered in C. elegans by V. Ambros [5]. It was initially called stRNA (short temporal RNA) due to its restricted expression and its role in controlling the transition from larval to adult stage gene expression in C. elegans. The second miRNA, let-7, was subsequently identified in the development timing of C. elegans [6]. The homologues of the let-7 gene were then found in other animals, including humans [7]. The conservation of let-7 across species suggests a fundamental biological role for this small RNA. Humans have more than 1000 miRNAs that can target more than 30% of mammalian genes and are abundant in many types of human cells [8,9,10]. This makes miRNAs one of the most abundant classes of regulatory genes in humans. miRNAs are perceived as a key layer of post-transcriptional control within gene regulation. These miRNAs are highly conserved across species and have the common function of regulating gene expression at the post-transcriptional level [8,9,10].

2. miRNAs and Cancer

Numerous miRNAs have been found to have links to certain types of cancer [11,12]. Three observations made during the early stages of miRNA research suggested that they may play a role in human cancer. First, the first miRNAs discovered in roundworms C. elegans and fruit flies Drosophila were found to control cell growth and cell death, which can contribute to cancer if they are deregulated [5,13]. Second, when human miRNAs were discovered, many of them were found to be located in fragile sites in the genome or regions that are commonly amplified or deleted in human cancer [14]. Finally, miRNA expression was deregulated in malignant tumors and tumor cell lines compared to normal tissue [15,16,17]. It is still unknown whether altered miRNA expression is a cause or a consequence of malignant transformation.

3. miRNAs Function as an Oncogene or Tumor Suppressor

miRNAs are a type of RNA that plays a crucial role in the regulation of gene expression. They are short, non-coding RNAs that control target genes post-transcriptionally and have emerged as master regulators in various physiological and pathological processes, including oncogenesis [10,18,19,20,21,22]. Recent studies have shown that miRNAs regulate cell growth and apoptosis, making them potential targets for cancer therapies [19,22]. One of the first miRNAs discovered as an oncogene is miR-155, which is highly expressed in different types of cancer, including pediatric Burkitt lymphoma [21], Hodgkin disease [23], acute myeloid leukemia (AML) [24], lung cancer [25,26], and breast cancer [25,27]. However, the regulation of miR-155 and the mechanism of its overexpression in cancer are still unknown. Another oncogenic miRNA is miR-21, which is upregulated in various hematologic malignancies and solid tumors, including AML [24,28], glioblastoma [29], breast cancer [25], and liver tumors [30]. Understanding the role of miRNAs in cancer can help in the development of new diagnostic tools and therapies for cancer patients.
miRNAs can act as tumor suppressors by preventing the malignant transformation of a normal cell. In some cases, the expression of specific miRNAs is decreased in cancer cells, and these miRNAs are considered tumor suppressor genes [31]. For example, the miR15a/16-1 cluster is often downregulated or deleted in chronic lymphocytic leukemia (CLL) [32]. This cluster is inversely correlated with the expression of BCL-2, a key player in many types of human tumors [31]. This suggests that miRNAs may be involved in cancer formation by regulating cell growth and apoptosis. Ectopic expression of miR-16-1 has been shown to negatively regulate cell growth, induce apoptosis, and inhibit cell cycle progression in human cancer cell lines and in a leukemic xenograft model [33,34]. Similarly, the let-7 family of miRNAs is downregulated in many tumors, including lung and breast cancer [26,27,35], and overexpression of let-7 inhibited lung cancer cell growth in A549 lung adenocarcinoma cells [36]. However, miR-17-5p is described as a dual-role tumor suppressor/oncogene, which means that it can act according to the tissue and its transcriptome, including miRNA targets expressed in that particular tissue [25].

4. miRNAs and Cancer Therapy

miRNAs hold great promise as therapeutic agents because they regulate gene expression and influence cellular pathways involved in various diseases, including cancer, cardiovascular disorders, and neurological conditions [37,38,39,40,41,42,43,44,45,46,47,48]. The therapeutic potential of miRNAs stems from their ability to target multiple genes simultaneously, allowing for the modulation of complex biological processes. In this review, we focus on miRNAs and cancer therapy. (1) miRNA replacement therapy: In cancer, where miRNAs often act as tumor suppressors or oncogenes, restoring the expression of tumor suppressor miRNAs or inhibiting oncogenic miRNAs can be a therapeutic strategy. Synthetic miRNA mimics or viral vectors carrying miRNA sequences can be delivered to tumor cells to restore miRNA function and suppress tumor growth [41,42]. (2) miRNA inhibition therapy: In contrast, inhibition of oncogenic miRNAs using antisense oligonucleotides, antagomiRs, or miR-decoy can be effective in blocking their activity and inhibiting cancer progression [40,41]. (3) Drug delivery: Efficient delivery of miRNAs to target tissues remains a challenge for miRNA-based therapy. Various delivery systems, including lipid nanoparticles, viral vectors, and exosomes, are being developed to improve target cells’ stability, specificity, and uptake of miRNA [49,50]. (4) Combination therapy: miRNA-based therapies can be combined with conventional treatments such as chemotherapy, radiation therapy, or targeted therapies to improve treatment efficacy and overcome drug resistance. Combinatorial approaches can use synergistic interactions between miRNAs and other therapeutic modalities to improve patient outcomes [51,52,53].
Although miRNA-based therapy holds significant promise, several challenges need to be addressed, including optimization of delivery methods, minimizing off-target effects, and ensuring long-term safety and efficacy. However, ongoing research efforts continue to advance our understanding of miRNA biology and therapeutic applications, paving the way for the development of innovative treatments for a wide range of diseases. By expanding our discussion to include these additional miRNAs and their targets, we aim to provide a more comprehensive perspective on how miRNAs command the battle against cancer. This comprehensive approach underscores the potential as biomarkers and therapeutic agents, offering promising avenues for future cancer research and treatment.

5. Tumor Suppressor p53 and Tumors

The tumor suppressor p53 is inactivated in more than 50% of human tumors, and the p53 mutation is the most commonly observed genetic event in cancer cells [54,55,56,57]. Studies have demonstrated the crucial role of p53 in protecting genomes. Mice lacking p53 are highly susceptible to tumors [58], and germline mutations in p53 cause Li–Fraumeni syndrome, making individuals more prone to breast, brain, and adrenal gland cancers, as well as leukemias and sarcomas of bone and connective tissues [55,59]. The p53 gene controls the cell cycle, initiates apoptotic cell death, and regulates DNA repair [56,60,61,62,63]. Mutations in p53 have been associated with more aggressive diseases and worse overall survival [64,65,66,67]. Inactivation of p53 has been associated with a poor prognosis and drug resistance in malignant tumors [55,57,68], indicating its importance in cancer pathogenesis. The regulation of p53 expression and stability is crucial in maintaining normal cell growth. Many upstream regulators and downstream effectors of p53 are believed to play a role in carcinogenesis in humans and other species. The response to DNA damage in cancer progression is summarized in Figure 1. miRNAs have also been shown to target p53 and/or components of p53 regulatory pathways, thereby directly and/or indirectly affecting its activities [69,70,71,72,73].

5.1. Regulation of p53 by miRNAs

Numerous miRNAs have been identified as regulators of p53, influencing its expression, activity, and downstream signaling pathways [69,74,75,76,77,78]. One of the well-studied miRNAs targeting p53 is miR-34a [79,80,81,82,83,84,85]. It is directly transactivated by p53 and forms a feedback loop that regulates the p53 function. miR-34a suppresses tumors by targeting multiple components of the p53 pathway, including SIRT1, which deacetylates and inhibits p53, and various anti-apoptotic and cell cycle regulatory genes [86]. Through its regulation of p53 signaling and downstream effectors, miR-34a maintains genomic stability and suppresses tumorigenesis. Several miRNAs indirectly regulate p53 expression or activity by targeting key regulators of the p53 pathway. For example, miR-125b and miR-504 target p53 inhibitors such as MDM2, increasing the stability and activity [87]. miR-192/215 and miR-504 can directly target the 3’UTR of p53 mRNA, leading to reduced expression of p53 [87,88]. miR-125a and miR-504 target p53 cofactors, such as the p300/CBP-associated factor (PCAF), which affects p53 acetylation and transcriptional activity [89]. Dysregulation of miRNA-mediated p53 regulation is involved in various diseases, particularly cancer [90]. Alterations in miRNA expression levels or mutations that affect miRNA binding sites in the p53 pathway can disrupt p53 function and contribute to tumorigenesis. Understanding the complex interplay between miRNAs and p53 signaling pathways can provide insights into developing novel therapeutic strategies for cancer treatment, such as miRNA-based therapies targeting the p53 pathway.
Our research has identified a microRNA called miR-1301, which plays an important role in the activity and function of p53 by targeting UBE4B [76]. It is worth noting that when miR-1301 is overexpressed, it inhibits the dissemination and metastasis of tumor cells in a p53-dependent manner. Our findings suggest that miR-1301 is a tumor suppressor that inhibits tumor cell migration and invasion by regulating the UBE4B-p53 pathway [76]. As p53 is a critical tumor suppressor, its regulation by miRNAs is a complex and actively researched area. Table 1 highlights the complex interaction between miRNAs and the p53 pathway, reflecting the nuanced regulatory mechanisms that govern cellular responses to stress and damage, impacting cancer development and progression. Three important insights are summarized below: (1). Upregulators of p53: The miRNAs that upregulate p53 generally do not directly target p53 but instead target negative regulators of p53 or elements involved in the cell cycle and proliferation. However, miR-214 is an exception that directly targets PSMD10 and ASF1B, which affect p53 function. (2). Downregulators of p53: Certain miRNAs downregulate p53 by directly binding to its 3’UTR. Some examples of these miRNAs are miR-25, miR-30d, miR-504, miR-380-5p, miR-92, and miR-141. Furthermore, miR-125b targets not only p53, but also a broader range of components within the p53 pro-apoptotic network. (3). Contextual functions: The effects of these miRNAs on p53 can vary depending on the context, such as the specific cell type or the state of the disease. For example, miR-125b has been implicated in the regulation of apoptosis and proliferation, impacting the conserved p53 network across vertebrates.

5.2. p63 Regulation by miRNAs

p63 is a homolog of the tumor suppressor p53 and can induce apoptosis and cell cycle arrest [119,120,121,122,123,124]. Although p63 has apoptotic activity, it rarely mutates in human tumors [125,126]. p63 deficiency is embryonically lethal in mice, causing severe developmental abnormalities, including a lack of epithelium and abnormalities of limb development [127,128]. However, recent studies have found that p63 expression is lost or reduced in certain human tumors. This suggests that p63 can potentially suppress tumors [129,130,131,132]. Flores et al. (2005) have shown that loss of p63 can cooperate with loss of p53 in tumor suppression [129]. Furthermore, mutations in both p63 and p53 can lead to more aggressive tumor growth [133,134]. Metastatic tumors that progress faster tend to have lower levels of p63 expression, suggesting that loss of p63 accelerates tumorigenesis and metastatic spread [135].
Less is known about the regulation of p63 by miRNAs compared to p53. However, emerging evidence suggests that miRNAs may play an important role in modulating p63 expression and activity in various cellular processes, including development, differentiation, and cancer. Several miRNAs, such as miR-203, miR-130b, miR-205, and miR-944, have been implicated in regulating p63 expression or activity [136,137,138,139,140,141,142]. These miRNAs can target different isoforms of p63 and modulate its transcriptional activity, protein stability, or subcellular localization.
We have discovered that the expression of miR-144/451 leads to an increase in p63 protein levels [143]. Furthermore, TAp63 transactivates the miR-144/451 cluster promoter, resulting in a positive feedback loop. miR-144 triggers apoptosis and reduces cell invasion in a TAp63-dependent manner. We found that Itch, an E3 ubiquitin ligase for p63, is directly targeted by miR-144. We also observed that the association between Ago2 and miR-451 is dependent on TAp63, implying the critical role of TAp63 in the biogenesis of miR-451 mediated by Ago2 [143]. This report highlights that miR-144 is a crucial regulator of the Itch-p63-Ago2 pathway, which could have implications for developing novel therapies for treating human tumors. Table 2 summarizes the p63 regulation by miRNAs. The table represents the currently known interactions, but further research is likely to identify additional miRNAs. The impact of miRNAs on p63 can vary based on cell type, tissue context, and specific isoforms of p63 involved (TAp63 vs. ΔNp63). The regulation of p63 by miRNAs underscores its critical role in epithelial cell fate, tumorigenesis, and cellular stress responses.

5.3. Regulation of PTEN by miRNAs

The phosphatase and tensin homolog (PTEN) is a well-known tumor suppressor involved in the PI3K/AKT signaling pathway. PTEN’s loss or downregulation is frequently observed in various cancers. Regulation of PTEN by miRNAs is a crucial area of research. Multiple miRNAs have been identified to target PTEN directly, leading to decreased expression and tumor progression. Understanding the interplay between these miRNAs and PTEN offers potential therapeutic avenues to restore PTEN function in cancer cells [153,154].
Among the miRNAs involved in PTEN regulation, miR-21 stands out as particularly significant due to its frequent upregulation in various human cancers and its direct targeting of PTEN. miR-21 overexpression has been shown to promote the growth and metastasis of specific cancers, including non-small cell lung cancer, colorectal carcinoma, ovarian cancer, and triple-negative breast cancers [30,153,155,156,157,158,159]. Furthermore, miR-21 and miR-181b-1 have been implicated in inhibiting PTEN and cylindromatosis (CYLD) tumor suppressor functions, leading to increased NF-kB activity and linking inflammation to cancer [160]. This evidence highlights the pivotal role of miR-21 in the oncogenic process through the modulation of PTEN expression, underlining its importance among miRNAs involved in PTEN regulation.
Other miRNAs, such as miR-214, miR-93, and miR-130a, also modulate PTEN expression and are associated with resistance to therapies such as cisplatin in ovarian cancer cells [161,162,163]. Furthermore, miR-221 and miR-222 have been associated with aggressive forms of cancer, such as non-small cell lung cancer (NSCLC), hepatocarcinoma, and breast cancer, targeting PTEN and contributing to resistance to treatment [164,165].
The intricate regulation of PTEN by miRNAs is further complicated by the presence of competing endogenous RNAs (ceRNAs), which can sequester miRNAs, thus modulating the levels of PTEN. For example, the PTEN pseudogene (PTENpg1) can act as a ceRNA, sequestering miRNAs away from PTEN mRNA and, therefore, enhancing its expression [166,167].
We identified that miR-498 directly targeted the 3’UTR of PTEN mRNA and reduced PTEN protein levels in triple-negative breast cancer (TNBC) cells [168]. miR-498 promoted cell proliferation and cell cycle progression in TNBC cells in a PTEN-dependent manner. Suppressing miR-498 expression affected the oncogenic effects of miR-498 on cell proliferation and migration. These results provide new information on the downregulation of PTEN and indicate a potential therapeutic target for the treatment of TNBC [168]. Future directions in the regulation of PTEN by miRNAs include a deeper exploration of the complex network of miRNA-PTEN interactions and how they affect cancer progression and resistance to treatments. The challenges lie in the vast diversity of miRNAs and their targets and the context-dependent effects of miRNAs, which can vary between different types of tissues and stages of disease. Understanding these dynamics is crucial for developing miRNA-based therapeutic strategies that could potentially target PTEN regulation to combat cancer more effectively. Table 3 highlights the essential role of miRNAs in the regulation of PTEN expression, which influences cancer progression through various mechanisms. Several factors need to be considered when studying the interaction between miRNAs and PTEN. First, the relationship between them is complex and context-dependent, which means that it can vary depending on the type of cancer, the cellular environment, and the specific expression levels of miRNA. Second, some miRNAs may not directly target PTEN, but can still affect its expression through upstream signaling pathways or other regulatory molecules. Finally, studying the role of miRNAs in PTEN regulation can lead to developing new therapeutic strategies for cancers involving PTEN dysfunction. We propose a model to illustrate how miRNAs regulate tumor suppressors’ expression or activity (Figure 2).

6. Diagnostic, Prognostic, and Predictive Potential of miRNAs in Cancer

6.1. Diagnostic Potential of miRNAs

6.1.1. Early Detection of Cancers

miRNAs such as miR-34a, miR-125b, and miR-504, which are involved in p53 regulation [84,87,108], miR-203, miR-130b, and miR-944, which influence p63 expression or activity [142,145,148], and miRNAs such as miR-21 and miR-498 that directly target PTEN are frequently upregulated in various cancers such as triple-negative breast cancer and colorectal carcinoma [155,168]. Altered levels of these miRNAs might be detectable in bodily fluids before traditional clinical symptoms manifest, offering a non-invasive diagnostic tool.

6.1.2. Cancer Typing

miRNA profiles that regulate p63 may help differentiate cancer subtypes, particularly epithelial cancers where p63 plays a critical role in development and differentiation. Measuring specific miRNAs could help distinguish between cancer types and even subtypes, informing treatment strategies.

6.2. Prognostic Potential of miRNAs

6.2.1. Survival and Treatment Outcomes

miRNAs such as miR-34a and miR-1301, which regulate key components of the p53 pathway, have been linked to tumor suppression and genomic stability [76,84]. Their expression levels could potentially predict patient survival, likelihood of recurrence, and response to therapy. Higher levels of tumor suppressor miRNAs might correlate with better prognosis. miRNAs that modulate p63, such as miR-144 and miR-451, might be associated with better or worse outcomes based on their impact on p63 expression levels and tumor suppressor functions [143]. The role in triggering apoptosis and reducing cell invasion could be correlated with a favorable prognosis in cancers where p63 is active. miRNAs that downregulate PTEN, such as miR-21 and miR-221/222, have been linked with poor prognosis and more aggressive cancer phenotypes [155,164,165]. Patients exhibiting high levels of these miRNAs may have a worse prognosis, with faster disease progression and lower overall survival rates.

6.2.2. Assessing Disease Aggression

miRNAs that downregulate p53, such as miR-504 and miR-380-5p, could serve as biomarkers for more aggressive cancers [87,109]. Their presence and levels could inform clinicians about the likely disease course and aggressiveness, helping to tailor more aggressive treatment plans where necessary. Lower levels of p63 have been associated with more aggressive and metastatic tumor behaviors. miRNAs that decrease p63 expression or destabilize its protein could serve as indicators of more aggressive disease courses, aiding in predicting the likelihood of metastasis and overall disease aggressiveness. miRNAs such as miR-21 that promote cancer growth and metastasis by suppressing PTEN can also serve as markers to gauge disease progression [155,156]. Higher expression of such miRNAs could indicate a higher likelihood of metastatic spread and more aggressive disease, informing treatment intensity and monitoring strategies.

6.3. Predictive Potential of miRNAs

6.3.1. Prediction of Treatment Response

miRNAs such as miR-34a and miR-1301, targeting elements of the p53 pathway, could predict how well a patient might respond to specific therapies, particularly those that aim to activate p53. For example, patients with high expression of miR-34a might respond favorably to therapies that rely on intact p53 signaling. Since miRNAs such as miR-144 directly target components such as Itch, which are involved in the regulation of p63, their expression profiles could predict how a tumor might respond to therapies aiming to modulate the p63 pathway [143]. For example, tumors with high levels of miR-144 might respond better to therapies that aim to enhance p63’s tumor suppressive activities. miRNAs such as miR-214, miR-93, and miR-130a, which modulate PTEN expression, have been associated with resistance to therapies like cisplatin in ovarian cancer [161,162,163]. Profiling these miRNAs in patients could predict their response to specific treatments, enabling personalized therapy plans that could bypass or counteract resistance mechanisms.

6.3.2. Personalized Medicine Applications

Understanding the specific interactions between miRNAs and p53 can help in customizing therapeutic approaches. For example, if a patient has downregulated miR-1301, therapies designed to enhance its expression could be more effective in restoring normal p53 function and potentially inhibiting tumor growth and spread. Understanding the specific miRNA-p63 interactions could guide the development of targeted therapies, such as miRNA mimics or antagomiRs, to either upregulate or downregulate p63 activity in cancer treatments, depending on the desired therapeutic effect. Identifying the presence of specific miRNAs that target PTEN can guide the development and use of targeted therapies, such as miRNA mimics or inhibitors. For example, in cancers where miR-498 is found to downregulate PTEN, therapies aimed at inhibiting miR-498 could be effective.

7. Tissue-Specific Roles of miRNAs in Cancer

The tissue-specific expression and activity of miRNAs are crucial for understanding their roles in the pathogenesis and progression of various cancers.

7.1. Tissue-Specific Expression Patterns

The expression of specific miRNAs can be inherently tied to particular tissues. For example, miR-21 is upregulated in lung and breast tissues in cancerous states, distinctly influencing tumor progression in each tissue type.

7.2. Differential Roles in Cancer

miR-34a acts differently in colorectal cancer compared to its role in prostate cancer, according to its interaction with tissue-specific cofactors and targets [193].

7.3. Impact on Metastasis and Tissue Invasion

For example, miR-10b promotes metastasis in breast cancer but plays a different role in pancreatic cancer [194,195].

7.4. Clinical Implications

For example, miR-205 is a potent marker for skin and breast cancers, but plays a variety of roles in these tissues, which could influence therapeutic strategies [196].

8. Role of miRNAs in Modulating Immune cell Dynamics in the Cancer Microenvironment

8.1. miRNAs and Immune Cell Recruitment

miR-21 has been shown to reduce the expression of PDCD4, a pro-inflammatory protein, potentially diminishing the antitumor immune response to tumors and facilitating tumor growth [197].

8.2. Impact on Immune Evasion

miR-221 and miR-222 target PTEN and also influence immune surveillance by affecting the expression of immune checkpoint molecules such as PD-L1 [198]. This mechanism can lead to increased immune evasion, particularly in cancers where PTEN is already compromised.

8.3. Regulation of Immune-Related Signaling Pathways

miR-34a, directly transactivated by p53, influences tumor cells and impacts immune cells by modulating cytokine profiles and T-cell activation, critical for immune surveillance and antitumor immunity [193,199].

8.4. Interaction between p63, miRNAs, and Immune Cells

miR-203 and miR-205 regulate p63 and influence epithelial-to-mesenchymal transition (EMT) [200,201]. EMT is crucial for metastasis and for the creation of an immunosuppressive microenvironment that limits the efficacy of immune cells.

8.5. Therapeutic Implications

Understanding these interactions opens new avenues for therapies targeting these miRNAs, which could enhance immune response while controlling tumor growth. For example, inhibiting miR-21 or miR-221/222 could restore PTEN function and enhance immune cell activity within the tumor microenvironment.

9. Role of miRNAs in Tumor Differentiation, Evolution, and Metastasis

9.1. miRNAs and Tumor Differentiation

9.1.1. Influence on Cell Fate

Some members of the miR-200 family influence the epithelial–mesenchymal transition (EMT), a key process in tumor cell differentiation and metastasis.

9.1.2. Regulation of Differentiation Markers

miRNAs modulate the expression of markers that are crucial for maintaining stemness or differentiation within tumor cells, such as the role in regulating CD44, a cancer stem cell marker [202].

9.2. miRNAs and Tumor Evolution

9.2.1. Genetic and Epigenetic Modulation

miRNAs contribute to the genetic and epigenetic changes within tumors, affecting tumor heterogeneity and evolution; for example, miR-21’s role in promoting oncogenic mutations by targeting multiple tumor suppressor genes, including PTEN.

9.2.2. Adaptation to the Microenvironment

miR-155 influences the ability of tumor cells to adapt to and manipulate their microenvironment, facilitating survival and growth under various stress conditions.

9.3. miRNAs and Metastases

9.3.1. Promotion of the Metastatic Cascade

miRNAs such as miR-10b and miR-21 facilitate different stages of the metastatic cascade, from local invasion, intravasation, survival in circulation, and extravasation, to colonization at distant sites.

9.3.2. Interaction with the Metastatic Niche

miRNAs prepare distant organs for tumor cell colonization (premetastatic niche formation), including the impact on metastatic liver colonization in breast cancer.

10. Conclusions and Perspectives

miRNAs play critical roles in regulating tumor suppressor(s) or oncogene(s) expression, activity, and downstream signaling pathways. Through direct and indirect mechanisms, miRNAs can influence various aspects of tumor suppressor/or oncogene function, maintaining cell homeostasis and suppressing tumorigenesis. Future research efforts may focus on unraveling the functional significance of miRNA-mediated tumor suppressor/oncogene regulation in normal physiological processes and disease contexts, such as cancer and developmental disorders. Furthermore, elucidating the cross-talk between miRNA-regulated pathways and tumor suppressor/oncogene signaling networks could provide insight into complex regulatory mechanisms. Developing innovative approaches to targeted modulation of miRNA activity, such as miRNA mimics, inhibitors, or delivery systems, can offer new therapeutic opportunities for diseases associated with dysregulated p53/p63 signaling. Challenges in studying tumor suppressor/oncogene regulation by miRNAs include the complexity of the regulatory network, the context-dependent effects of miRNAs, and the need for robust experimental models and methodologies to validate miRNA–target interactions and functional outcomes. The challenges of studying the regulation of p63 by miRNAs include the diversity of p63 isoforms, the context-dependent functions of p63 in different cell types and tissues, and the need for sophisticated experimental approaches to dissect the molecular mechanisms underlying miRNA–p63 interactions. Future research efforts aimed at elucidating the complex interplay between miRNAs and tumor suppressor/oncogene signaling networks hold promise for uncovering new insights into cellular processes and disease mechanisms, with potential implications for developing novel therapeutic strategies.

Author Contributions

H.H.W. drafted the manuscript. S.L. and C.S. discussed and supported it. R.L. supervised the study and revised it. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from the Canadian Institutes of Health Research (CIHR, #115177) and the Natural Sciences and Engineering Research Council of Canada (NSERC, #06582) to R.L.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All other relevant data supporting the findings of this study are available in the article.

Acknowledgments

The authors thank the University of Alberta for the research equipment and technical support for this research.

Conflicts of Interest

The authors declare that they have no competing interest.

References

  1. Bartel, D.P. Metazoan MicroRNAs. Cell 2018, 173, 20–51. [Google Scholar] [CrossRef] [PubMed]
  2. Diener, C.; Keller, A.; Meese, E. Emerging concepts of miRNA therapeutics: From cells to clinic. Trends Genet. 2022, 38, 613–626. [Google Scholar] [CrossRef]
  3. Ho, P.T.B.; Clark, I.M.; Le, L.T.T. MicroRNA-Based Diagnosis and Therapy. Int. J. Mol. Sci. 2022, 23, 7167. [Google Scholar] [CrossRef]
  4. Rupaimoole, R.; Slack, F.J. MicroRNA therapeutics: Towards a new era for the management of cancer and other diseases. Nat. Rev. Drug Discov. 2017, 16, 203–222. [Google Scholar] [CrossRef] [PubMed]
  5. Lee, R.C.; Feinbaum, R.L.; Ambros, V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 1993, 75, 843–854. [Google Scholar] [CrossRef]
  6. Reinhart, B.J.; Slack, F.J.; Basson, M.; Pasquinelli, A.E.; Bettinger, J.C.; Rougvie, A.E.; Horvitz, H.R.; Ruvkun, G. The 21-nucleotide let-7 RNA regulates developmental timing in Caenorhabditis elegans. Nature 2000, 403, 901–906. [Google Scholar] [CrossRef]
  7. Pasquinelli, A.E.; Reinhart, B.J.; Slack, F.; Martindale, M.Q.; Kuroda, M.I.; Maller, B.; Hayward, D.C.; Ball, E.E.; Degnan, B.; Müller, P.; et al. Conservation of the sequence and temporal expression of let-7 heterochronic regulatory RNA. Nature 2000, 408, 86–89. [Google Scholar] [CrossRef] [PubMed]
  8. Bentwich, I.; Avniel, A.; Karov, Y.; Aharonov, R.; Gilad, S.; Barad, O.; Barzilai, A.; Einat, P.; Einav, U.; Meiri, E.; et al. Identification of hundreds of conserved and nonconserved human microRNAs. Nat. Genet. 2005, 37, 766–770. [Google Scholar] [CrossRef]
  9. Friedman, R.C.; Farh, K.K.; Burge, C.B.; Bartel, D.P. Most mammalian mRNAs are conserved targets of microRNAs. Genome Res. 2009, 19, 92–105. [Google Scholar] [CrossRef]
  10. Lewis, B.P.; Burge, C.B.; Bartel, D.P. Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 2005, 120, 15–20. [Google Scholar] [CrossRef]
  11. He, L.; Thomson, J.M.; Hemann, M.T.; Hernando-Monge, E.; Mu, D.; Goodson, S.; Powers, S.; Cordon-Cardo, C.; Lowe, S.W.; Hannon, G.J.; et al. A microRNA polycistron as a potential human oncogene. Nature 2005, 435, 828–833. [Google Scholar] [CrossRef] [PubMed]
  12. Hwang, H.; Chang, H.R.; Baek, D. Determinants of Functional MicroRNA Targeting. Mol. Cells 2023, 46, 21–32. [Google Scholar] [CrossRef] [PubMed]
  13. Brennecke, J.; Hipfner, D.R.; Stark, A.; Russell, R.B.; Cohen, S.M. bantam encodes a developmentally regulated microRNA that controls cell proliferation and regulates the proapoptotic gene hid in Drosophila. Cell 2003, 113, 25–36. [Google Scholar] [CrossRef] [PubMed]
  14. Hutvagner, G.; Simard, M.J.; Mello, C.C.; Zamore, P.D. Sequence-specific inhibition of small RNA function. PLoS Biol. 2004, 2, E98. [Google Scholar] [CrossRef] [PubMed]
  15. Calin, G.A.; Croce, C.M. MicroRNA signatures in human cancers. Nat. Rev. Cancer 2006, 6, 857–866. [Google Scholar] [CrossRef] [PubMed]
  16. Lu, J.; Getz, G.; Miska, E.A.; Alvarez-Saavedra, E.; Lamb, J.; Peck, D.; Sweet-Cordero, A.; Ebert, B.L.; Mak, R.H.; Ferrando, A.A.; et al. MicroRNA expression profiles classify human cancers. Nature 2005, 435, 834–838. [Google Scholar] [CrossRef]
  17. Rupaimoole, R.; Calin, G.A.; Lopez-Berestein, G.; Sood, A.K. miRNA Deregulation in Cancer Cells and the Tumor Microenvironment. Cancer Discov. 2016, 6, 235–246. [Google Scholar] [CrossRef] [PubMed]
  18. Abba, M.L.; Patil, N.; Leupold, J.H.; Moniuszko, M.; Utikal, J.; Niklinski, J.; Allgayer, H. MicroRNAs as novel targets and tools in cancer therapy. Cancer Lett. 2017, 387, 84–94. [Google Scholar] [CrossRef] [PubMed]
  19. Cheng, A.M.; Byrom, M.W.; Shelton, J.; Ford, L.P. Antisense inhibition of human miRNAs and indications for an involvement of miRNA in cell growth and apoptosis. Nucleic Acids Res. 2005, 33, 1290–1297. [Google Scholar] [CrossRef]
  20. Lim, L.P.; Lau, N.C.; Garrett-Engele, P.; Grimson, A.; Schelter, J.M.; Castle, J.; Bartel, D.P.; Linsley, P.S.; Johnson, J.M. Microarray analysis shows that some microRNAs downregulate large numbers of target mRNAs. Nature 2005, 433, 769–773. [Google Scholar] [CrossRef]
  21. Metzler, M.; Wilda, M.; Busch, K.; Viehmann, S.; Borkhardt, A. High expression of precursor microRNA-155/BIC RNA in children with Burkitt lymphoma. Genes. Chromosomes Cancer 2004, 39, 167–169. [Google Scholar] [CrossRef] [PubMed]
  22. Tanno, B.; Cesi, V.; Vitali, R.; Sesti, F.; Giuffrida, M.L.; Mancini, C.; Calabretta, B.; Raschella, G. Silencing of endogenous IGFBP-5 by micro RNA interference affects proliferation, apoptosis and differentiation of neuroblastoma cells. Cell Death Differ. 2005, 12, 213–223. [Google Scholar] [CrossRef] [PubMed]
  23. Kluiver, J.; Poppema, S.; de Jong, D.; Blokzijl, T.; Harms, G.; Jacobs, S.; Kroesen, B.J.; van den Berg, A. BIC and miR-155 are highly expressed in Hodgkin, primary mediastinal and diffuse large B cell lymphomas. J. Pathol. 2005, 207, 243–249. [Google Scholar] [CrossRef]
  24. Garzon, R.; Volinia, S.; Liu, C.G.; Fernandez-Cymering, C.; Palumbo, T.; Pichiorri, F.; Fabbri, M.; Coombes, K.; Alder, H.; Nakamura, T.; et al. MicroRNA signatures associated with cytogenetics and prognosis in acute myeloid leukemia. Blood 2008, 111, 3183–3189. [Google Scholar] [CrossRef]
  25. Volinia, S.; Calin, G.A.; Liu, C.G.; Ambs, S.; Cimmino, A.; Petrocca, F.; Visone, R.; Iorio, M.; Roldo, C.; Ferracin, M.; et al. A microRNA expression signature of human solid tumors defines cancer gene targets. Proc. Natl. Acad. Sci. USA 2006, 103, 2257–2261. [Google Scholar] [CrossRef] [PubMed]
  26. Yanaihara, N.; Caplen, N.; Bowman, E.; Seike, M.; Kumamoto, K.; Yi, M.; Stephens, R.M.; Okamoto, A.; Yokota, J.; Tanaka, T.; et al. Unique microRNA molecular profiles in lung cancer diagnosis and prognosis. Cancer Cell 2006, 9, 189–198. [Google Scholar] [CrossRef] [PubMed]
  27. Iorio, M.V.; Ferracin, M.; Liu, C.G.; Veronese, A.; Spizzo, R.; Sabbioni, S.; Magri, E.; Pedriali, M.; Fabbri, M.; Campiglio, M.; et al. MicroRNA gene expression deregulation in human breast cancer. Cancer Res. 2005, 65, 7065–7070. [Google Scholar] [CrossRef] [PubMed]
  28. Garzon, R.; Garofalo, M.; Martelli, M.P.; Briesewitz, R.; Wang, L.; Fernandez-Cymering, C.; Volinia, S.; Liu, C.G.; Schnittger, S.; Haferlach, T.; et al. Distinctive microRNA signature of acute myeloid leukemia bearing cytoplasmic mutated nucleophosmin. Proc. Natl. Acad. Sci. USA 2008, 105, 3945–3950. [Google Scholar] [CrossRef] [PubMed]
  29. Ciafrè, S.A.; Galardi, S.; Mangiola, A.; Ferracin, M.; Liu, C.G.; Sabatino, G.; Negrini, M.; Maira, G.; Croce, C.M.; Farace, M.G. Extensive modulation of a set of microRNAs in primary glioblastoma. Biochem. Biophys. Res. Commun. 2005, 334, 1351–1358. [Google Scholar] [CrossRef]
  30. Meng, F.; Henson, R.; Wehbe-Janek, H.; Ghoshal, K.; Jacob, S.T.; Patel, T. MicroRNA-21 regulates expression of the PTEN tumor suppressor gene in human hepatocellular cancer. Gastroenterology 2007, 133, 647–658. [Google Scholar] [CrossRef]
  31. Cimmino, A.; Calin, G.A.; Fabbri, M.; Iorio, M.V.; Ferracin, M.; Shimizu, M.; Wojcik, S.E.; Aqeilan, R.I.; Zupo, S.; Dono, M.; et al. miR-15 and miR-16 induce apoptosis by targeting BCL2. Proc. Natl. Acad. Sci. USA 2005, 102, 13944–13949. [Google Scholar] [CrossRef] [PubMed]
  32. Calin, G.A.; Dumitru, C.D.; Shimizu, M.; Bichi, R.; Zupo, S.; Noch, E.; Aldler, H.; Rattan, S.; Keating, M.; Rai, K.; et al. Frequent deletions and down-regulation of micro- RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc. Natl. Acad. Sci. USA 2002, 99, 15524–15529. [Google Scholar] [CrossRef] [PubMed]
  33. Calin, G.A.; Cimmino, A.; Fabbri, M.; Ferracin, M.; Wojcik, S.E.; Shimizu, M.; Taccioli, C.; Zanesi, N.; Garzon, R.; Aqeilan, R.I.; et al. MiR-15a and miR-16-1 cluster functions in human leukemia. Proc. Natl. Acad. Sci. USA 2008, 105, 5166–5171. [Google Scholar] [CrossRef]
  34. Linsley, P.S.; Schelter, J.; Burchard, J.; Kibukawa, M.; Martin, M.M.; Bartz, S.R.; Johnson, J.M.; Cummins, J.M.; Raymond, C.K.; Dai, H.; et al. Transcripts targeted by the microRNA-16 family cooperatively regulate cell cycle progression. Mol. Cell Biol. 2007, 27, 2240–2252. [Google Scholar] [CrossRef] [PubMed]
  35. Johnson, S.M.; Grosshans, H.; Shingara, J.; Byrom, M.; Jarvis, R.; Cheng, A.; Labourier, E.; Reinert, K.L.; Brown, D.; Slack, F.J. RAS is regulated by the let-7 microRNA family. Cell 2005, 120, 635–647. [Google Scholar] [CrossRef] [PubMed]
  36. Takamizawa, J.; Konishi, H.; Yanagisawa, K.; Tomida, S.; Osada, H.; Endoh, H.; Harano, T.; Yatabe, Y.; Nagino, M.; Nimura, Y.; et al. Reduced expression of the let-7 microRNAs in human lung cancers in association with shortened postoperative survival. Cancer Res. 2004, 64, 3753–3756. [Google Scholar] [CrossRef] [PubMed]
  37. Badr, D.; Fouad, M.A.; Hussein, M.; Salem, S.; Zekri, A.; Shouman, S. Rebound increase in microRNA levels at the end of 5-FU-based therapy in colorectal cancer patients. Sci. Rep. 2023, 13, 14237. [Google Scholar] [CrossRef] [PubMed]
  38. Colpaert, R.M.W.; Calore, M. MicroRNAs in Cardiac Diseases. Cells 2019, 8, 737. [Google Scholar] [CrossRef] [PubMed]
  39. Kahraman, M.; Roske, A.; Laufer, T.; Fehlmann, T.; Backes, C.; Kern, F.; Kohlhaas, J.; Schrors, H.; Saiz, A.; Zabler, C.; et al. MicroRNA in diagnosis and therapy monitoring of early-stage triple-negative breast cancer. Sci. Rep. 2018, 8, 11584. [Google Scholar] [CrossRef]
  40. Kim, T.; Croce, C.M. MicroRNA: Trends in clinical trials of cancer diagnosis and therapy strategies. Exp. Mol. Med. 2023, 55, 1314–1321. [Google Scholar] [CrossRef]
  41. Menon, A.; Abd-Aziz, N.; Khalid, K.; Poh, C.L.; Naidu, R. miRNA: A Promising Therapeutic Target in Cancer. Int. J. Mol. Sci. 2022, 23, 11502. [Google Scholar] [CrossRef] [PubMed]
  42. Mollaei, H.; Safaralizadeh, R.; Rostami, Z. MicroRNA replacement therapy in cancer. J. Cell Physiol. 2019, 234, 12369–12384. [Google Scholar] [CrossRef] [PubMed]
  43. Musso, N.; Bivona, D.; Bonomo, C.; Bonacci, P.; D’Ippolito, M.E.; Boccagni, C.; Rubino, F.; De Tanti, A.; Lucca, L.F.; Pingue, V.; et al. Investigating microRNAs as biomarkers in disorders of consciousness: A longitudinal multicenter study. Sci. Rep. 2023, 13, 18415. [Google Scholar] [CrossRef] [PubMed]
  44. Shah, V.; Shah, J. Recent trends in targeting miRNAs for cancer therapy. J. Pharm. Pharmacol. 2020, 72, 1732–1749. [Google Scholar] [CrossRef]
  45. Szczepanek, J.; Skorupa, M.; Tretyn, A. MicroRNA as a Potential Therapeutic Molecule in Cancer. Cells 2022, 11, 1008. [Google Scholar] [CrossRef] [PubMed]
  46. Wang, H.; Taguchi, Y.H.; Liu, X. Editorial: miRNAs and Neurological Diseases. Front. Neurol. 2021, 12, 662373. [Google Scholar] [CrossRef]
  47. Zhang, G.Q.; Wang, S.Q.; Chen, Y.; Fu, L.Y.; Xu, Y.N.; Li, L.; Tao, L.; Shen, X.C. MicroRNAs Regulating Mitochondrial Function in Cardiac Diseases. Front. Pharmacol. 2021, 12, 663322. [Google Scholar] [CrossRef] [PubMed]
  48. Smith, E.S.; Whitty, E.; Yoo, B.; Moore, A.; Sempere, L.F.; Medarova, Z. Clinical Applications of Short Non-Coding RNA-Based Therapies in the Era of Precision Medicine. Cancers 2022, 14, 1588. [Google Scholar] [CrossRef]
  49. Dasgupta, I.; Chatterjee, A. Recent Advances in miRNA Delivery Systems. Methods Protoc. 2021, 4, 10. [Google Scholar] [CrossRef]
  50. Kang, E.; Kortylewski, M. Lipid Nanoparticle-Mediated Delivery of miRNA Mimics to Myeloid Cells. Methods Mol. Biol. 2023, 2691, 337–350. [Google Scholar]
  51. Costa, D.F.; Sarisozen, C.; Torchilin, V.P. Synthesis of Doxorubicin and miRNA Stimuli-Sensitive Conjugates for Combination Therapy. Methods Mol. Biol. 2019, 1974, 99–109. [Google Scholar] [PubMed]
  52. Normann, L.S.; Aure, M.R.; Leivonen, S.K.; Haugen, M.H.; Hongisto, V.; Kristensen, V.N.; Maelandsmo, G.M.; Sahlberg, K.K. MicroRNA in combination with HER2-targeting drugs reduces breast cancer cell viability in vitro. Sci. Rep. 2021, 11, 10893. [Google Scholar] [CrossRef] [PubMed]
  53. Seo, H.A.; Moeng, S.; Sim, S.; Kuh, H.J.; Choi, S.Y.; Park, J.K. MicroRNA-Based Combinatorial Cancer Therapy: Effects of MicroRNAs on the Efficacy of Anti-Cancer Therapies. Cells 2019, 9, 29. [Google Scholar] [CrossRef] [PubMed]
  54. Hafner, A.; Bulyk, M.L.; Jambhekar, A.; Lahav, G. The multiple mechanisms that regulate p53 activity and cell fate. Nat. Rev. Mol. Cell Biol. 2019, 20, 199–210. [Google Scholar] [CrossRef] [PubMed]
  55. Levine, A.J. p53, the cellular gatekeeper for growth and division. Cell 1997, 88, 323–331. [Google Scholar] [CrossRef] [PubMed]
  56. Stracquadanio, G.; Wang, X.; Wallace, M.D.; Grawenda, A.M.; Zhang, P.; Hewitt, J.; Zeron-Medina, J.; Castro-Giner, F.; Tomlinson, I.P.; Goding, C.R.; et al. The importance of p53 pathway genetics in inherited and somatic cancer genomes. Nat. Rev. Cancer 2016, 16, 251–265. [Google Scholar] [CrossRef]
  57. Vogelstein, B.; Lane, D.; Levine, A.J. Surfing the p53 network. Nature 2000, 408, 307–310. [Google Scholar] [CrossRef] [PubMed]
  58. Donehower, L.A.; Harvey, M.; Slagle, B.L.; McArthur, M.J.; Montgomery, C.A., Jr.; Butel, J.S.; Bradley, A. Mice deficient for p53 are developmentally normal but susceptible to spontaneous tumours. Nature 1992, 356, 215–221. [Google Scholar] [CrossRef] [PubMed]
  59. Malkin, D.; Li, F.P.; Strong, L.C.; Fraumeni, J.F., Jr.; Nelson, C.E.; Kim, D.H.; Kassel, J.; Gryka, M.A.; Bischoff, F.Z.; Tainsky, M.A.; et al. Germ line p53 mutations in a familial syndrome of breast cancer, sarcomas, and other neoplasms. Science 1990, 250, 1233–1238. [Google Scholar] [CrossRef]
  60. Bieging, K.T.; Mello, S.S.; Attardi, L.D. Unravelling mechanisms of p53-mediated tumour suppression. Nat. Rev. Cancer 2014, 14, 359–370. [Google Scholar] [CrossRef]
  61. Kastenhuber, E.R.; Lowe, S.W. Putting p53 in Context. Cell 2017, 170, 1062–1078. [Google Scholar] [CrossRef]
  62. Laptenko, O.; Prives, C. p53: Master of life, death, and the epigenome. Genes. Dev. 2017, 31, 955–956. [Google Scholar] [CrossRef]
  63. Powell, E.; Piwnica-Worms, D.; Piwnica-Worms, H. Contribution of p53 to metastasis. Cancer Discov. 2014, 4, 405–414. [Google Scholar] [CrossRef]
  64. Alexander, K.A.; Cote, A.; Nguyen, S.C.; Zhang, L.; Gholamalamdari, O.; Agudelo-Garcia, P.; Lin-Shiao, E.; Tanim, K.M.A.; Lim, J.; Biddle, N.; et al. p53 mediates target gene association with nuclear speckles for amplified RNA expression. Mol. Cell 2021, 81, 1666–1681.e6. [Google Scholar] [CrossRef]
  65. Aubrey, B.J.; Kelly, G.L.; Janic, A.; Herold, M.J.; Strasser, A. How does p53 induce apoptosis and how does this relate to p53-mediated tumour suppression? Cell Death Differ. 2018, 25, 104–113. [Google Scholar] [CrossRef]
  66. Sabapathy, K.; Lane, D.P. Therapeutic targeting of p53: All mutants are equal, but some mutants are more equal than others. Nat. Rev. Clin. Oncol. 2018, 15, 13–30. [Google Scholar] [CrossRef]
  67. Wang, M.; Attardi, L.D. A Balancing Act: p53 Activity from Tumor Suppression to Pathology and Therapeutic Implications. Annu. Rev. Pathol. 2022, 17, 205–226. [Google Scholar] [CrossRef] [PubMed]
  68. Greenblatt, M.S.; Bennett, W.P.; Hollstein, M.; Harris, C.C. Mutations in the p53 tumor suppressor gene: Clues to cancer etiology and molecular pathogenesis. Cancer Res. 1994, 54, 4855–4878. [Google Scholar] [PubMed]
  69. Hermeking, H. MicroRNAs in the p53 network: Micromanagement of tumour suppression. Nat. Rev. Cancer 2012, 12, 613–626. [Google Scholar] [CrossRef]
  70. Kumar, M.; Lu, Z.; Takwi, A.A.; Chen, W.; Callander, N.S.; Ramos, K.S.; Young, K.H.; Li, Y. Negative regulation of the tumor suppressor p53 gene by microRNAs. Oncogene 2011, 30, 843–853. [Google Scholar] [CrossRef]
  71. Liu, J.; Zhang, C.; Zhao, Y.; Feng, Z. MicroRNA Control of p53. J. Cell Biochem. 2017, 118, 7–14. [Google Scholar] [CrossRef] [PubMed]
  72. Sargolzaei, J.; Etemadi, T.; Alyasin, A. The P53/microRNA network: A potential tumor suppressor with a role in anticancer therapy. Pharmacol. Res. 2020, 160, 105179. [Google Scholar] [CrossRef] [PubMed]
  73. Suzuki, H.I.; Yamagata, K.; Sugimoto, K.; Iwamoto, T.; Kato, S.; Miyazono, K. Modulation of microRNA processing by p53. Nature 2009, 460, 529–533. [Google Scholar] [CrossRef] [PubMed]
  74. Hermeking, H. p53 enters the microRNA world. Cancer Cell 2007, 12, 414–418. [Google Scholar] [CrossRef]
  75. Hoffman, Y.; Bublik, D.R.; Pilpel, Y.; Oren, M. miR-661 downregulates both Mdm2 and Mdm4 to activate p53. Cell Death Differ. 2014, 21, 302–309. [Google Scholar] [CrossRef] [PubMed]
  76. Wang, B.; Wu, H.; Chai, C.; Lewis, J.; Pichiorri, F.; Eisenstat, D.D.; Pomeroy, S.L.; Leng, R.P. MicroRNA-1301 suppresses tumor cell migration and invasion by targeting the p53/UBE4B pathway in multiple human cancer cells. Cancer Lett. 2017, 401, 20–32. [Google Scholar] [CrossRef] [PubMed]
  77. Xiao, J.; Lin, H.; Luo, X.; Luo, X.; Wang, Z. miR-605 joins p53 network to form a p53:miR-605:Mdm2 positive feedback loop in response to stress. EMBO J. 2011, 30, 5021. [Google Scholar] [CrossRef]
  78. Capaccia, C.; Diverio, S.; Zampini, D.; Guelfi, G. The Complex Interaction between P53 and miRNAs Joins New Awareness in Physiological Stress Responses. Cells 2022, 11, 1631. [Google Scholar] [CrossRef] [PubMed]
  79. Choi, Y.J.; Lin, C.P.; Risso, D.; Chen, S.; Kim, T.A.; Tan, M.H.; Li, J.B.; Wu, Y.; Chen, C.; Xuan, Z.; et al. Deficiency of microRNA miR-34a expands cell fate potential in pluripotent stem cells. Science 2017, 355, eaag1927. [Google Scholar] [CrossRef]
  80. Hermeking, H. The miR-34 family in cancer and apoptosis. Cell Death Differ. 2010, 17, 193–199. [Google Scholar] [CrossRef]
  81. Kim, N.H.; Kim, H.S.; Kim, N.G.; Lee, I.; Choi, H.S.; Li, X.Y.; Kang, S.E.; Cha, S.Y.; Ryu, J.K.; Na, J.M.; et al. p53 and microRNA-34 are suppressors of canonical Wnt signaling. Sci. Signal 2011, 4, ra71. [Google Scholar] [CrossRef] [PubMed]
  82. Kim, N.H.; Kim, H.S.; Li, X.Y.; Lee, I.; Choi, H.S.; Kang, S.E.; Cha, S.Y.; Ryu, J.K.; Yoon, D.; Fearon, E.R.; et al. A p53/miRNA-34 axis regulates Snail1-dependent cancer cell epithelial-mesenchymal transition. J. Cell Biol. 2011, 195, 417–433. [Google Scholar] [CrossRef] [PubMed]
  83. Navarro, F.; Lieberman, J. miR-34 and p53: New Insights into a Complex Functional Relationship. PLoS ONE 2015, 10, e0132767. [Google Scholar] [CrossRef] [PubMed]
  84. Okada, N.; Lin, C.P.; Ribeiro, M.C.; Biton, A.; Lai, G.; He, X.; Bu, P.; Vogel, H.; Jablons, D.M.; Keller, A.C.; et al. A positive feedback between p53 and miR-34 miRNAs mediates tumor suppression. Genes. Dev. 2014, 28, 438–450. [Google Scholar] [CrossRef] [PubMed]
  85. Zhang, D.G.; Zheng, J.N.; Pei, D.S. P53/microRNA-34-induced metabolic regulation: New opportunities in anticancer therapy. Mol. Cancer 2014, 13, 115. [Google Scholar] [CrossRef] [PubMed]
  86. Yamakuchi, M.; Ferlito, M.; Lowenstein, C.J. miR-34a repression of SIRT1 regulates apoptosis. Proc. Natl. Acad. Sci. USA 2008, 105, 13421–13426. [Google Scholar] [CrossRef] [PubMed]
  87. Hu, W.; Chan, C.S.; Wu, R.; Zhang, C.; Sun, Y.; Song, J.S.; Tang, L.H.; Levine, A.J.; Feng, Z. Negative regulation of tumor suppressor p53 by microRNA miR-504. Mol. Cell 2010, 38, 689–699. [Google Scholar] [CrossRef] [PubMed]
  88. Georges, S.A.; Biery, M.C.; Kim, S.Y.; Schelter, J.M.; Guo, J.; Chang, A.N.; Jackson, A.L.; Carleton, M.O.; Linsley, P.S.; Cleary, M.A.; et al. Coordinated regulation of cell cycle transcripts by p53-Inducible microRNAs, miR-192 and miR-215. Cancer Res. 2008, 68, 10105–10112. [Google Scholar] [CrossRef]
  89. Abdi, J.; Rastgoo, N.; Li, L.; Chen, W.; Chang, H. Role of tumor suppressor p53 and micro-RNA interplay in multiple myeloma pathogenesis. J. Hematol. Oncol. 2017, 10, 169. [Google Scholar] [CrossRef]
  90. Madrigal, T.; Hernandez-Monge, J.; Herrera, L.A.; Gonzalez-De la Rosa, C.H.; Dominguez-Gomez, G.; Candelaria, M.; Luna-Maldonado, F.; Calderon Gonzalez, K.G.; Diaz-Chavez, J. Regulation of miRNAs Expression by Mutant p53 Gain of Function in Cancer. Front. Cell Dev. Biol. 2021, 9, 695723. [Google Scholar] [CrossRef]
  91. Fornari, F.; Gramantieri, L.; Giovannini, C.; Veronese, A.; Ferracin, M.; Sabbioni, S.; Calin, G.A.; Grazi, G.L.; Croce, C.M.; Tavolari, S.; et al. MiR-122/cyclin G1 interaction modulates p53 activity and affects doxorubicin sensitivity of human hepatocarcinoma cells. Cancer Res. 2009, 69, 5761–5767. [Google Scholar] [CrossRef] [PubMed]
  92. Jones, M.; Lal, A. MicroRNAs, wild-type and mutant p53: More questions than answers. RNA Biol. 2012, 9, 781–791. [Google Scholar] [CrossRef]
  93. Kwon, J.J.; Factora, T.D.; Dey, S.; Kota, J. A Systematic Review of miR-29 in Cancer. Mol. Ther. Oncolytics 2019, 12, 173–194. [Google Scholar] [CrossRef]
  94. Nguyen, T.T.P.; Suman, K.H.; Nguyen, T.B.; Nguyen, H.T.; Do, D.N. The Role of miR-29s in Human Cancers-An Update. Biomedicines 2022, 10, 2121. [Google Scholar] [CrossRef] [PubMed]
  95. Sun, C.Y.; Zhang, X.P.; Wang, W. Coordination of miR-192 and miR-22 in p53-Mediated Cell Fate Decision. Int. J. Mol. Sci. 2019, 10, 4768. [Google Scholar] [CrossRef]
  96. Feng, Z.; Zhang, C.; Wu, R.; Hu, W. Tumor suppressor p53 meets microRNAs. J. Mol. Cell Biol. 2011, 3, 44–50. [Google Scholar] [CrossRef]
  97. Hoffman, Y.; Pilpel, Y.; Oren, M. microRNAs and Alu elements in the p53-Mdm2-Mdm4 regulatory network. J. Mol. Cell Biol. 2014, 6, 192–197. [Google Scholar] [CrossRef] [PubMed]
  98. Zhang, C.; Liu, J.; Wang, X.; Wu, R.; Lin, M.; Laddha, S.V.; Yang, Q.; Chan, C.S.; Feng, Z. MicroRNA-339-5p inhibits colorectal tumorigenesis through regulation of the MDM2/p53 signaling. Oncotarget 2014, 5, 9106–9117. [Google Scholar] [CrossRef] [PubMed]
  99. Misiewicz-Krzeminska, I.; Sarasquete, M.E.; Quwaider, D.; Krzeminski, P.; Ticona, F.V.; Paíno, T.; Delgado, M.; Aires, A.; Ocio, E.M.; García-Sanz, R.; et al. Restoration of microRNA-214 expression reduces growth of myeloma cells through positive regulation of P53 and inhibition of DNA replication. Haematologica 2013, 98, 640–648. [Google Scholar] [CrossRef]
  100. Spizzo, R.; Nicoloso, M.S.; Lupini, L.; Lu, Y.; Fogarty, J.; Rossi, S.; Zagatti, B.; Fabbri, M.; Veronese, A.; Liu, X.; et al. miR-145 participates with TP53 in a death-promoting regulatory loop and targets estrogen receptor-alpha in human breast cancer cells. Cell Death Differ. 2010, 17, 246–254. [Google Scholar] [CrossRef]
  101. Zhang, C.; Liu, J.; Tan, C.; Yue, X.; Zhao, Y.; Peng, J.; Wang, X.; Laddha, S.V.; Chan, C.S.; Zheng, S.; et al. microRNA-1827 represses MDM2 to positively regulate tumor suppressor p53 and suppress tumorigenesis. Oncotarget 2016, 7, 8783–8796. [Google Scholar] [CrossRef] [PubMed]
  102. Dar, A.A.; Majid, S.; Rittsteuer, C.; de Semir, D.; Bezrookove, V.; Tong, S.; Nosrati, M.; Sagebiel, R.; Miller, J.R., 3rd; Kashani-Sabet, M. The role of miR-18b in MDM2-p53 pathway signaling and melanoma progression. J. Natl. Cancer Inst. 2013, 105, 433–442. [Google Scholar] [CrossRef] [PubMed]
  103. Jansson, M.D.; Damas, N.D.; Lees, M.; Jacobsen, A.; Lund, A.H. miR-339-5p regulates the p53 tumor-suppressor pathway by targeting MDM2. Oncogene 2015, 34, 1908–1918. [Google Scholar] [CrossRef]
  104. Bou Kheir, T.; Futoma-Kazmierczak, E.; Jacobsen, A.; Krogh, A.; Bardram, L.; Hother, C.; Grønbæk, K.; Federspiel, B.; Lund, A.H.; Friis-Hansen, L. miR-449 inhibits cell proliferation and is down-regulated in gastric cancer. Mol. Cancer 2011, 10, 29. [Google Scholar] [CrossRef] [PubMed]
  105. Suh, S.S.; Yoo, J.Y.; Nuovo, G.J.; Jeon, Y.J.; Kim, S.; Lee, T.J.; Kim, T.; Bakacs, A.; Alder, H.; Kaur, B.; et al. MicroRNAs/TP53 feedback circuitry in glioblastoma multiforme. Proc. Natl. Acad. Sci. USA 2012, 109, 5316–5321. [Google Scholar] [CrossRef] [PubMed]
  106. Wang, Y.; Huang, J.W.; Castella, M.; Huntsman, D.G.; Taniguchi, T. p53 is positively regulated by miR-542-3p. Cancer Res. 2014, 74, 3218–3227. [Google Scholar] [CrossRef] [PubMed]
  107. Mulcahy, E.Q.X.; Zhang, Y.; Colόn, R.R.; Cain, S.R.; Gibert, M.K., Jr.; Dube, C.J.; Hafner, M.; Abounader, R. MicroRNA 3928 Suppresses Glioblastoma through Downregulation of Several Oncogenes and Upregulation of p53. Int. J. Mol. Sci. 2022, 23, 3930. [Google Scholar] [CrossRef]
  108. Le, M.T.; Teh, C.; Shyh-Chang, N.; Xie, H.; Zhou, B.; Korzh, V.; Lodish, H.F.; Lim, B. MicroRNA-125b is a novel negative regulator of p53. Genes. Dev. 2009, 23, 862–876. [Google Scholar] [CrossRef]
  109. Swarbrick, A.; Woods, S.L.; Shaw, A.; Balakrishnan, A.; Phua, Y.; Nguyen, A.; Chanthery, Y.; Lim, L.; Ashton, L.J.; Judson, R.L.; et al. miR-380-5p represses p53 to control cellular survival and is associated with poor outcome in MYCN-amplified neuroblastoma. Nat. Med. 2010, 16, 1134–1140. [Google Scholar] [CrossRef]
  110. Zhang, S.; Yu, J.; Sun, B.F.; Hou, G.Z.; Yu, Z.J.; Luo, H. MicroRNA-92a Targets SERTAD3 and Regulates the Growth, Invasion, and Migration of Prostate Cancer Cells via the P53 Pathway. Onco Targets Ther. 2020, 13, 5495–5514. [Google Scholar] [CrossRef]
  111. Zhou, X.; Wu, W.; Zeng, A.; Nie, E.; Jin, X.; Yu, T.; Zhi, T.; Jiang, K.; Wang, Y.; Zhang, J.; et al. MicroRNA-141-3p promotes glioma cell growth and temozolomide resistance by directly targeting p53. Oncotarget 2017, 8, 71080–71094. [Google Scholar] [CrossRef] [PubMed]
  112. Herrera-Merchan, A.; Cerrato, C.; Luengo, G.; Dominguez, O.; Piris, M.A.; Serrano, M.; Gonzalez, S. miR-33-mediated downregulation of p53 controls hematopoietic stem cell self-renewal. Cell Cycle 2010, 9, 3277–3285. [Google Scholar] [CrossRef]
  113. Fan, Y.; Yin, S.; Hao, Y.; Yang, J.; Zhang, H.; Sun, C.; Ma, M.; Chang, Q.; Xi, J.J. miR-19b promotes tumor growth and metastasis via targeting TP53. RNA 2014, 20, 765–772. [Google Scholar] [CrossRef] [PubMed]
  114. Zhang, S.; Zhang, C.; Li, Y.; Wang, P.; Yue, Z.; Xie, S. miR-98 regulates cisplatin-induced A549 cell death by inhibiting TP53 pathway. Biomed. Pharmacother. 2011, 65, 436–442. [Google Scholar] [CrossRef] [PubMed]
  115. Liu, F.; Di Wang, X. miR-150-5p represses TP53 tumor suppressor gene to promote proliferation of colon adenocarcinoma. Sci. Rep. 2019, 9, 6740. [Google Scholar] [CrossRef] [PubMed]
  116. Liu, Y.; Xing, R.; Zhang, X.; Dong, W.; Zhang, J.; Yan, Z.; Li, W.; Cui, J.; Lu, Y. miR-375 targets the p53 gene to regulate cellular response to ionizing radiation and etoposide in gastric cancer cells. DNA Repair 2013, 12, 741–750. [Google Scholar] [CrossRef]
  117. Tian, S.; Huang, S.; Wu, S.; Guo, W.; Li, J.; He, X. MicroRNA-1285 inhibits the expression of p53 by directly targeting its 3’ untranslated region. Biochem. Biophys. Res. Commun. 2010, 396, 435–439. [Google Scholar] [CrossRef] [PubMed]
  118. Eisfeld, A.K.; Schwind, S.; Patel, R.; Huang, X.; Santhanam, R.; Walker, C.J.; Markowitz, J.; Hoag, K.W.; Jarvinen, T.M.; Leffel, B.; et al. Intronic miR-3151 within BAALC drives leukemogenesis by deregulating the TP53 pathway. Sci. Signal 2014, 7, ra36. [Google Scholar] [CrossRef] [PubMed]
  119. Armstrong, S.R.; Wu, H.; Wang, B.; Abuetabh, Y.; Sergi, C.; Leng, R.P. The Regulation of Tumor Suppressor p63 by the Ubiquitin-Proteasome System. Int. J. Mol. Sci. 2016, 17, 2041. [Google Scholar] [CrossRef]
  120. Melino, G. p63 is a suppressor of tumorigenesis and metastasis interacting with mutant p53. Cell Death Differ. 2011, 18, 1487–1499. [Google Scholar] [CrossRef]
  121. Melino, G.; Lu, X.; Gasco, M.; Crook, T.; Knight, R.A. Functional regulation of p73 and p63: Development and cancer. Trends Biochem. Sci. 2003, 28, 663–670. [Google Scholar] [CrossRef]
  122. Wu, H.H.; Wang, B.; Armstrong, S.R.; Abuetabh, Y.; Leng, S.; Roa, W.H.Y.; Atfi, A.; Marchese, A.; Wilson, B.; Sergi, C.; et al. Hsp70 acts as a fine-switch that controls E3 ligase CHIP-mediated TAp63 and ΔNp63 ubiquitination and degradation. Nucleic Acids Res. 2021, 49, 2740–2758. [Google Scholar] [CrossRef]
  123. Yang, A.; Kaghad, M.; Wang, Y.; Gillett, E.; Fleming, M.D.; Dötsch, V.; Andrews, N.C.; Caput, D.; McKeon, F. p63, a p53 homolog at 3q27-29, encodes multiple products with transactivating, death-inducing, and dominant-negative activities. Mol. Cell 1998, 2, 305–316. [Google Scholar] [CrossRef] [PubMed]
  124. Yang, A.; McKeon, F. P63 and P73: P53 mimics, menaces and more. Nat. Rev. Mol. Cell Biol. 2000, 1, 199–207. [Google Scholar] [CrossRef] [PubMed]
  125. De Laurenzi, V.; Melino, G. Evolution of functions within the p53/p63/p73 family. Ann. N. Y. Acad. Sci. 2000, 926, 90–100. [Google Scholar] [CrossRef]
  126. Moll, U.M.; Slade, N. p63 and p73: Roles in development and tumor formation. Mol. Cancer Res. 2004, 2, 371–386. [Google Scholar] [CrossRef]
  127. Mills, A.A.; Zheng, B.; Wang, X.J.; Vogel, H.; Roop, D.R.; Bradley, A. p63 is a p53 homologue required for limb and epidermal morphogenesis. Nature 1999, 398, 708–713. [Google Scholar] [CrossRef]
  128. Yang, A.; Schweitzer, R.; Sun, D.; Kaghad, M.; Walker, N.; Bronson, R.T.; Tabin, C.; Sharpe, A.; Caput, D.; Crum, C.; et al. p63 is essential for regenerative proliferation in limb, craniofacial and epithelial development. Nature 1999, 398, 714–718. [Google Scholar] [CrossRef] [PubMed]
  129. Flores, E.R.; Sengupta, S.; Miller, J.B.; Newman, J.J.; Bronson, R.; Crowley, D.; Yang, A.; McKeon, F.; Jacks, T. Tumor predisposition in mice mutant for p63 and p73: Evidence for broader tumor suppressor functions for the p53 family. Cancer Cell 2005, 7, 363–373. [Google Scholar] [CrossRef]
  130. Guo, X.; Keyes, W.M.; Papazoglu, C.; Zuber, J.; Li, W.; Lowe, S.W.; Vogel, H.; Mills, A.A. TAp63 induces senescence and suppresses tumorigenesis in vivo. Nat. Cell Biol. 2009, 11, 1451–1457. [Google Scholar] [CrossRef]
  131. Su, X.; Chakravarti, D.; Cho, M.S.; Liu, L.; Gi, Y.J.; Lin, Y.L.; Leung, M.L.; El-Naggar, A.; Creighton, C.J.; Suraokar, M.B.; et al. TAp63 suppresses metastasis through coordinate regulation of Dicer and miRNAs. Nature 2010, 467, 986–990. [Google Scholar] [CrossRef] [PubMed]
  132. Su, X.; Gi, Y.J.; Chakravarti, D.; Chan, I.L.; Zhang, A.; Xia, X.; Tsai, K.Y.; Flores, E.R. TAp63 is a master transcriptional regulator of lipid and glucose metabolism. Cell Metab. 2012, 16, 511–525. [Google Scholar] [CrossRef] [PubMed]
  133. Flores, E.R.; Tsai, K.Y.; Crowley, D.; Sengupta, S.; Yang, A.; McKeon, F.; Jacks, T. p63 and p73 are required for p53-dependent apoptosis in response to DNA damage. Nature 2002, 416, 560–564. [Google Scholar] [CrossRef] [PubMed]
  134. Rocco, J.W.; Leong, C.O.; Kuperwasser, N.; DeYoung, M.P.; Ellisen, L.W. p63 mediates survival in squamous cell carcinoma by suppression of p73-dependent apoptosis. Cancer Cell 2006, 9, 45–56. [Google Scholar] [CrossRef] [PubMed]
  135. Massion, P.P.; Taflan, P.M.; Jamshedur Rahman, S.M.; Yildiz, P.; Shyr, Y.; Edgerton, M.E.; Westfall, M.D.; Roberts, J.R.; Pietenpol, J.A.; Carbone, D.P.; et al. Significance of p63 amplification and overexpression in lung cancer development and prognosis. Cancer Res. 2003, 63, 7113–7121. [Google Scholar] [PubMed]
  136. Chen, H.L.; Chiang, P.C.; Lo, C.H.; Lo, Y.H.; Hsu, D.K.; Chen, H.Y.; Liu, F.T. Galectin-7 Regulates Keratinocyte Proliferation and Differentiation through JNK-miR-203-p63 Signaling. J. Investig. Dermatol. 2016, 136, 182–191. [Google Scholar] [CrossRef]
  137. Gunaratne, P.H.; Pan, Y.; Rao, A.K.; Lin, C.; Hernandez-Herrera, A.; Liang, K.; Rait, A.S.; Venkatanarayan, A.; Benham, A.L.; Rubab, F.; et al. Activating p53 family member TAp63: A novel therapeutic strategy for targeting p53-altered tumors. Cancer 2019, 125, 2409–2422. [Google Scholar] [CrossRef] [PubMed]
  138. Kim, J.; Park, S.; Chang, Y.; Park, K.H.; Lee, H. Synergetic Effects of Intronic Mature miR-944 and ΔNp63 Isoforms on Tumorigenesis in a Cervical Cancer Cell Line. Int. J. Mol. Sci. 2020, 21, 5612. [Google Scholar] [CrossRef] [PubMed]
  139. Odar, K.; Boštjančič, E.; Gale, N.; Glavač, D.; Zidar, N. Differential expression of microRNAs miR-21, miR-31, miR-203, miR-125a-5p and miR-125b and proteins PTEN and p63 in verrucous carcinoma of the head and neck. Histopathology 2012, 61, 257–265. [Google Scholar] [CrossRef]
  140. Tran, M.N.; Choi, W.; Wszolek, M.F.; Navai, N.; Lee, I.L.; Nitti, G.; Wen, S.; Flores, E.R.; Siefker-Radtke, A.; Czerniak, B.; et al. The p63 protein isoform ΔNp63α inhibits epithelial-mesenchymal transition in human bladder cancer cells: Role of MIR-205. J. Biol. Chem. 2013, 288, 3275–3288. [Google Scholar] [CrossRef]
  141. Tucci, P.; Agostini, M.; Grespi, F.; Markert, E.K.; Terrinoni, A.; Vousden, K.H.; Muller, P.A.; Dötsch, V.; Kehrloesser, S.; Sayan, B.S.; et al. Loss of p63 and its microRNA-205 target results in enhanced cell migration and metastasis in prostate cancer. Proc. Natl. Acad. Sci. USA 2012, 109, 15312–15317. [Google Scholar] [CrossRef]
  142. Xie, H.; Lee, L.; Scicluna, P.; Kavak, E.; Larsson, C.; Sandberg, R.; Lui, W.O. Novel functions and targets of miR-944 in human cervical cancer cells. Int. J. Cancer 2015, 136, E230–E241. [Google Scholar] [CrossRef] [PubMed]
  143. Wang, B.; Wu, H.H.; Abuetabh, Y.; Leng, S.; Davidge, S.T.; Flores, E.R.; Eisenstat, D.D.; Leng, R. p63, a key regulator of Ago2, links to the microRNA-144 cluster. Cell Death Dis. 2022, 13, 397. [Google Scholar] [CrossRef] [PubMed]
  144. Chikh, A.; Matin, R.N.; Senatore, V.; Hufbauer, M.; Lavery, D.; Raimondi, C.; Ostano, P.; Mello-Grand, M.; Ghimenti, C.; Bahta, A.; et al. iASPP/p63 autoregulatory feedback loop is required for the homeostasis of stratified epithelia. EMBO J. 2011, 30, 4261–4273. [Google Scholar] [CrossRef] [PubMed]
  145. Lena, A.M.; Shalom-Feuerstein, R.; Rivetti di Val Cervo, P.; Aberdam, D.; Knight, R.A.; Melino, G.; Candi, E. miR-203 represses ‘stemness’ by repressing DeltaNp63. Cell Death Differ. 2008, 15, 1187–1195. [Google Scholar] [CrossRef] [PubMed]
  146. Manni, I.; Artuso, S.; Careccia, S.; Rizzo, M.G.; Baserga, R.; Piaggio, G.; Sacchi, A. The microRNA miR-92 increases proliferation of myeloid cells and by targeting p63 modulates the abundance of its isoforms. FASEB J. 2009, 23, 3957–3966. [Google Scholar] [CrossRef]
  147. Scheel, A.H.; Beyer, U.; Agami, R.; Dobbelstein, M. Immunofluorescence-based screening identifies germ cell associated microRNA 302 as an antagonist to p63 expression. Cell Cycle 2009, 8, 1426–1432. [Google Scholar] [CrossRef]
  148. Rivetti di Val Cervo, P.; Lena, A.M.; Nicoloso, M.; Rossi, S.; Mancini, M.; Zhou, H.; Saintigny, G.; Dellambra, E.; Odorisio, T.; Mahe, C.; et al. p63-microRNA feedback in keratinocyte senescence. Proc. Natl. Acad. Sci. USA 2012, 109, 1133–1138. [Google Scholar] [CrossRef] [PubMed]
  149. Huang, Y.; Kesselman, D.; Kizub, D.; Guerrero-Preston, R.; Ratovitski, E.A. Phospho-ΔNp63α/microRNA feedback regulation in squamous carcinoma cells upon cisplatin exposure. Cell Cycle 2013, 12, 684–697. [Google Scholar] [CrossRef]
  150. Kim, K.; Madak-Erdogan, Z.; Ventrella, R.; Katzenellenbogen, B.S. A MicroRNA196a2* and TP63 circuit regulated by estrogen receptor-α and ERK2 that controls breast cancer proliferation and invasiveness properties. Horm. Cancer 2013, 4, 78–91. [Google Scholar] [CrossRef]
  151. Papagiannakopoulos, T.; Shapiro, A.; Kosik, K.S. MicroRNA-21 targets a network of key tumor-suppressive pathways in glioblastoma cells. Cancer Res. 2008, 68, 8164–8172. [Google Scholar] [CrossRef] [PubMed]
  152. Quintavalle, C.; Donnarumma, E.; Iaboni, M.; Roscigno, G.; Garofalo, M.; Romano, G.; Fiore, D.; De Marinis, P.; Croce, C.M.; Condorelli, G. Effect of miR-21 and miR-30b/c on TRAIL-induced apoptosis in glioma cells. Oncogene 2013, 32, 4001–4008. [Google Scholar] [CrossRef] [PubMed]
  153. Ghafouri-Fard, S.; Abak, A.; Shoorei, H.; Mohaqiq, M.; Majidpoor, J.; Sayad, A.; Taheri, M. Regulatory role of microRNAs on PTEN signaling. Biomed. Pharmacother. 2021, 133, 110986. [Google Scholar] [CrossRef] [PubMed]
  154. Travis, G.; McGowan, E.M.; Simpson, A.M.; Marsh, D.J.; Nassif, N.T. PTEN, PTENP1, microRNAs, and ceRNA Networks: Precision Targeting in Cancer Therapeutics. Cancers 2023, 15, 4954. [Google Scholar] [CrossRef] [PubMed]
  155. Bautista-Sánchez, D.; Arriaga-Canon, C.; Pedroza-Torres, A.; De La Rosa-Velázquez, I.A.; González-Barrios, R.; Contreras-Espinosa, L.; Montiel-Manríquez, R.; Castro-Hernández, C.; Fragoso-Ontiveros, V.; Álvarez-Gómez, R.M.; et al. The Promising Role of miR-21 as a Cancer Biomarker and Its Importance in RNA-Based Therapeutics. Mol. Ther. Nucleic Acids 2020, 20, 409–420. [Google Scholar] [CrossRef] [PubMed]
  156. Chawra, H.S.; Agarwal, M.; Mishra, A.; Chandel, S.S.; Singh, R.P.; Dubey, G.; Kukreti, N.; Singh, M. MicroRNA-21’s role in PTEN suppression and PI3K/AKT activation: Implications for cancer biology. Pathol. Res. Pract. 2024, 254, 155091. [Google Scholar] [CrossRef] [PubMed]
  157. Rhim, J.; Baek, W.; Seo, Y.; Kim, J.H. From Molecular Mechanisms to Therapeutics: Understanding MicroRNA-21 in Cancer. Cells 2022, 11, 2791. [Google Scholar] [CrossRef] [PubMed]
  158. Xu, L.F.; Wu, Z.P.; Chen, Y.; Zhu, Q.S.; Hamidi, S.; Navab, R. MicroRNA-21 (miR-21) regulates cellular proliferation, invasion, migration, and apoptosis by targeting PTEN, RECK and Bcl-2 in lung squamous carcinoma, Gejiu City, China. PLoS ONE 2014, 9, e103698. [Google Scholar] [CrossRef]
  159. Zhu, S.; Wu, H.; Wu, F.; Nie, D.; Sheng, S.; Mo, Y.Y. MicroRNA-21 targets tumor suppressor genes in invasion and metastasis. Cell Res. 2008, 18, 350–359. [Google Scholar] [CrossRef]
  160. Iliopoulos, D.; Jaeger, S.A.; Hirsch, H.A.; Bulyk, M.L.; Struhl, K. STAT3 activation of miR-21 and miR-181b-1 via PTEN and CYLD are part of the epigenetic switch linking inflammation to cancer. Mol. Cell 2010, 39, 493–506. [Google Scholar] [CrossRef]
  161. Chu, S.; Liu, G.; Xia, P.; Chen, G.; Shi, F.; Yi, T.; Zhou, H. miR-93 and PTEN: Key regulators of doxorubicin-resistance and EMT in breast cancer. Oncol. Rep. 2017, 38, 2401–2407. [Google Scholar] [CrossRef] [PubMed]
  162. Han, J.B.; Huang, M.L.; Li, F.; Yang, R.; Chen, S.M.; Tao, Z.Z. MiR-214 Mediates Cell Proliferation and Apoptosis of Nasopharyngeal Carcinoma Through Targeting Both WWOX and PTEN. Cancer Biother. Radiopharm. 2020, 35, 615–625. [Google Scholar] [CrossRef] [PubMed]
  163. Wei, H.; Cui, R.; Bahr, J.; Zanesi, N.; Luo, Z.; Meng, W.; Liang, G.; Croce, C.M. miR-130a Deregulates PTEN and Stimulates Tumor Growth. Cancer Res. 2017, 77, 6168–6178. [Google Scholar] [CrossRef] [PubMed]
  164. Yamashita, R.; Sato, M.; Kakumu, T.; Hase, T.; Yogo, N.; Maruyama, E.; Sekido, Y.; Kondo, M.; Hasegawa, Y. Growth inhibitory effects of miR-221 and miR-222 in non-small cell lung cancer cells. Cancer Med. 2015, 4, 551–564. [Google Scholar] [CrossRef] [PubMed]
  165. Li, B.; Lu, Y.; Yu, L.; Han, X.; Wang, H.; Mao, J.; Shen, J.; Wang, B.; Tang, J.; Li, C.; et al. miR-221/222 promote cancer stem-like cell properties and tumor growth of breast cancer via targeting PTEN and sustained Akt/NF-κB/COX-2 activation. Chem. Biol. Interact. 2017, 277, 33–42. [Google Scholar] [CrossRef] [PubMed]
  166. Johnsson, P.; Ackley, A.; Vidarsdottir, L.; Lui, W.O.; Corcoran, M.; Grandér, D.; Morris, K.V. A pseudogene long-noncoding-RNA network regulates PTEN transcription and translation in human cells. Nat. Struct. Mol. Biol. 2013, 20, 440–446. [Google Scholar] [CrossRef] [PubMed]
  167. Lister, N.; Shevchenko, G.; Walshe, J.L.; Groen, J.; Johnsson, P.; Vidarsdóttir, L.; Grander, D.; Ataide, S.F.; Morris, K.V. The molecular dynamics of long noncoding RNA control of transcription in PTEN and its pseudogene. Proc. Natl. Acad. Sci. USA 2017, 114, 9942–9947. [Google Scholar] [CrossRef] [PubMed]
  168. Chai, C.; Wu, H.; Wang, B.; Eisenstat, D.D.; Leng, R.P. MicroRNA-498 promotes proliferation and migration by targeting the tumor suppressor PTEN in breast cancer cells. Carcinogenesis 2018, 39, 1185–1196. [Google Scholar] [CrossRef] [PubMed]
  169. Li, B.; Sun, H. MiR-26a promotes neurite outgrowth by repressing PTEN expression. Mol. Med. Rep. 2013, 8, 676–680. [Google Scholar] [CrossRef]
  170. Coronel-Hernandez, J.; Lopez-Urrutia, E.; Contreras-Romero, C.; Delgado-Waldo, I.; Figueroa-Gonzalez, G.; Campos-Parra, A.D.; Salgado-Garcia, R.; Martinez-Gutierrez, A.; Rodriguez-Morales, M.; Jacobo-Herrera, N.; et al. Cell migration and proliferation are regulated by miR-26a in colorectal cancer via the PTEN-AKT axis. Cancer Cell Int. 2019, 19, 80. [Google Scholar] [CrossRef]
  171. Wang, W.; Liu, L.; Tian, Y. miR-19-3p Targets PTEN to Regulate Cervical Cancer Cell Proliferation, Invasion, and Autophagy. Genet Res. 2023, 2023, 4784500. [Google Scholar] [CrossRef] [PubMed]
  172. Liu, H.; Pan, Y.; Han, X.; Liu, J.; Li, R. MicroRNA-216a promotes the metastasis and epithelial-mesenchymal transition of ovarian cancer by suppressing the PTEN/AKT pathway. OncoTargets Ther. 2017, 10, 2701–2709. [Google Scholar] [CrossRef] [PubMed]
  173. Kato, M.; Putta, S.; Wang, M.; Yuan, H.; Lanting, L.; Nair, I.; Gunn, A.; Nakagawa, Y.; Shimano, H.; Todorov, I.; et al. TGF-beta activates Akt kinase through a microRNA-dependent amplifying circuit targeting PTEN. Nat. Cell Biol. 2009, 11, 881–889. [Google Scholar] [CrossRef] [PubMed]
  174. Bar, N.; Dikstein, R. miR-22 forms a regulatory loop in PTEN/AKT pathway and modulates signaling kinetics. PLoS ONE 2010, 5, e10859. [Google Scholar] [CrossRef] [PubMed]
  175. Fan, W.; Huang, J.; Xiao, H.; Liang, Z. MicroRNA-22 is downregulated in clear cell renal cell carcinoma, and inhibits cell growth, migration and invasion by targeting PTEN. Mol. Med. Rep. 2016, 13, 4800–4806. [Google Scholar] [CrossRef] [PubMed]
  176. Zhao, Y.S.; Yang, W.C.; Xin, H.W.; Han, J.X.; Ma, S.G. MiR-182-5p Knockdown Targeting PTEN Inhibits Cell Proliferation and Invasion of Breast Cancer Cells. Yonsei Med. J. 2019, 60, 148–157. [Google Scholar] [CrossRef] [PubMed]
  177. Wu, Z.; He, B.; He, J.; Mao, X. Upregulation of miR-153 promotes cell proliferation via downregulation of the PTEN tumor suppressor gene in human prostate cancer. Prostate 2013, 73, 596–604. [Google Scholar] [CrossRef] [PubMed]
  178. Yousefnia, S. A comprehensive review on miR-153: Mechanistic and controversial roles of miR-153 in tumorigenicity of cancer cells. Front. Oncol. 2022, 12, 985897. [Google Scholar] [CrossRef] [PubMed]
  179. Sagar, S.K. miR-106b as an emerging therapeutic target in cancer. Genes. Dis. 2022, 9, 889–899. [Google Scholar] [CrossRef]
  180. Li, N.; Miao, Y.; Shan, Y.; Liu, B.; Li, Y.; Zhao, L.; Jia, L. MiR-106b and miR-93 regulate cell progression by suppression of PTEN via PI3K/Akt pathway in breast cancer. Cell Death Dis. 2017, 8, e2796. [Google Scholar] [CrossRef]
  181. Liu, D.; Zhong, L.; Yuan, Z.; Yao, J.; Zhong, P.; Liu, J.; Yao, S.; Zhao, Y.; Liu, L.; Chen, M.; et al. miR-382-5p modulates the ATRA-induced differentiation of acute promyelocytic leukemia by targeting tumor suppressor PTEN. Cell Signal 2019, 54, 1–9. [Google Scholar] [CrossRef] [PubMed]
  182. Jiang, X.; Shan, A.; Su, Y.; Cheng, Y.; Gu, W.; Wang, W.; Ning, G.; Cao, Y. miR-144/451 Promote Cell Proliferation via Targeting PTEN/AKT Pathway in Insulinomas. Endocrinology 2015, 156, 2429–2439. [Google Scholar] [CrossRef] [PubMed]
  183. Liu, Q.; Geng, P.; Shi, L.; Wang, Q.; Wang, P. miR-29 promotes osteosarcoma cell proliferation and migration by targeting PTEN. Oncol. Lett. 2019, 17, 883–890. [Google Scholar] [CrossRef]
  184. Suo, H.B.; Zhang, K.C.; Zhao, J. MiR-200a promotes cell invasion and migration of ovarian carcinoma by targeting PTEN. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 4080–4089. [Google Scholar]
  185. Zhao, W.; Han, T.; Li, B.; Ma, Q.; Yang, P.; Li, H. miR-552 promotes ovarian cancer progression by regulating PTEN pathway. J. Ovarian Res. 2019, 12, 121. [Google Scholar] [CrossRef] [PubMed]
  186. Xiang, Y.; Chen, Y.J.; Yan, Y.B.; Liu, Y.; Qiu, J.; Tan, R.Q.; Tian, Q.; Guan, L.; Niu, S.S.; Xin, H.W. MiR-186 bidirectionally regulates cisplatin sensitivity of ovarian cancer cells via suppressing targets PIK3R3 and PTEN and upregulating APAF1 expression. J. Cancer 2020, 11, 3446–3453. [Google Scholar] [CrossRef]
  187. Wang, H.; Zhao, Y.; Chen, T.; Liu, G.; He, N.; Hu, H. MiR-371 promotes proliferation and metastasis in hepatocellular carcinoma by targeting PTEN. BMB Rep. 2019, 52, 312–317. [Google Scholar] [CrossRef]
  188. Sun, J.F.; Zhang, D.; Gao, C.J.; Zhang, Y.W.; Dai, Q.S. Exosome-Mediated MiR-155 Transfer Contributes to Hepatocellular Carcinoma Cell Proliferation by Targeting PTEN. Med. Sci. Monit. Basic. Res. 2019, 25, 218–228. [Google Scholar] [CrossRef]
  189. Lu, J.; Mu, X.; Yin, Q.; Hu, K. miR-106a contributes to prostate carcinoma progression through PTEN. Oncol. Lett. 2019, 17, 1327–1332. [Google Scholar] [CrossRef]
  190. Wang, H.; Ma, Z.; Liu, X.; Zhang, C.; Hu, Y.; Ding, L.; Qi, P.; Wang, J.; Lu, S.; Li, Y. MiR-183-5p is required for non-small cell lung cancer progression by repressing PTEN. Biomed. Pharmacother. 2019, 111, 1103–1111. [Google Scholar] [CrossRef]
  191. Xia, L.; Song, M.; Sun, M.; Chen, W.; Yang, C. miR-486 Promotes Capan-2 Pancreatic Cancer Cell Proliferation by Targeting Phosphatase and Tensin Homolog Deleted on Chromosome 10 (PTEN). Front. Genet. 2019, 10, 541. [Google Scholar] [CrossRef] [PubMed]
  192. Wei, G.; Yuan, Y.; He, X.; Jin, L.; Jin, D. Enhanced plasma miR-142-5p promotes the progression of intrahepatic cholangiocarcinoma via targeting PTEN. Exp. Ther. Med. 2019, 17, 4190–4196. [Google Scholar] [CrossRef] [PubMed]
  193. Fu, J.; Imani, S.; Wu, M.Y.; Wu, R.C. MicroRNA-34 Family in Cancers: Role, Mechanism, and Therapeutic Potential. Cancers 2023, 15, 4723. [Google Scholar] [CrossRef]
  194. Kim, J.; Siverly, A.N.; Chen, D.; Wang, M.; Yuan, Y.; Wang, Y.; Lee, H.; Zhang, J.; Muller, W.J.; Liang, H.; et al. Ablation of miR-10b Suppresses Oncogene-Induced Mammary Tumorigenesis and Metastasis and Reactivates Tumor-Suppressive Pathways. Cancer Res. 2016, 76, 6424–6435. [Google Scholar] [CrossRef] [PubMed]
  195. Xu, C.; Qi, X. MiR-10b inhibits migration and invasion of pancreatic ductal adenocarcinoma via regulating E2F7. J. Clin. Lab. Anal. 2020, 34, e23442. [Google Scholar] [CrossRef] [PubMed]
  196. Chauhan, N.; Dhasmana, A.; Jaggi, M.; Chauhan, S.C.; Yallapu, M.M. miR-205: A Potential Biomedicine for Cancer Therapy. Cells 2020, 9, 1957. [Google Scholar] [CrossRef] [PubMed]
  197. Asangani, I.A.; Rasheed, S.A.; Nikolova, D.A.; Leupold, J.H.; Colburn, N.H.; Post, S.; Allgayer, H. MicroRNA-21 (miR-21) post-transcriptionally downregulates tumor suppressor Pdcd4 and stimulates invasion, intravasation and metastasis in colorectal cancer. Oncogene 2008, 27, 2128–2136. [Google Scholar] [CrossRef] [PubMed]
  198. Di Martino, M.T.; Arbitrio, M.; Caracciolo, D.; Cordua, A.; Cuomo, O.; Grillone, K.; Riillo, C.; Caridà, G.; Scionti, F.; Labanca, C.; et al. miR-221/222 as biomarkers and targets for therapeutic intervention on cancer and other diseases: A systematic review. Mol. Ther. Nucleic Acids 2022, 27, 1191–1224. [Google Scholar] [CrossRef] [PubMed]
  199. Li, X.J.; Ren, Z.J.; Tang, J.H. MicroRNA-34a: A potential therapeutic target in human cancer. Cell Death Dis. 2014, 5, e1327. [Google Scholar] [CrossRef]
  200. Candi, E.; Amelio, I.; Agostini, M.; Melino, G. MicroRNAs and p63 in epithelial stemness. Cell Death Differ. 2015, 22, 12–21. [Google Scholar] [CrossRef]
  201. Taube, J.H.; Malouf, G.G.; Lu, E.; Sphyris, N.; Vijay, V.; Ramachandran, P.P.; Ueno, K.R.; Gaur, S.; Nicoloso, M.S.; Rossi, S.; et al. Epigenetic silencing of microRNA-203 is required for EMT and cancer stem cell properties. Sci. Rep. 2013, 3, 2687. [Google Scholar] [CrossRef] [PubMed]
  202. Li, W.J.; Wang, Y.; Liu, R.; Kasinski, A.L.; Shen, H.; Slack, F.J.; Tang, D.G. MicroRNA-34a: Potent Tumor Suppressor, Cancer Stem Cell Inhibitor, and Potential Anticancer Therapeutic. Front. Cell Dev. Biol. 2021, 9, 640587. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Response to DNA damage in cancer progression. This figure emphasizes the critical balance between tumor suppressor activation and oncogene activation in determining cellular fate following DNA damage. The proper function of tumor suppressors is essential for preventing cancer, while their inhibition or the activation of oncogenes can lead to malignant transformation.
Figure 1. Response to DNA damage in cancer progression. This figure emphasizes the critical balance between tumor suppressor activation and oncogene activation in determining cellular fate following DNA damage. The proper function of tumor suppressors is essential for preventing cancer, while their inhibition or the activation of oncogenes can lead to malignant transformation.
Ijms 25 05865 g001
Figure 2. Illustrates how miRNAs regulate tumor suppressors’ expression or activity. They can also affect their stability and activity by targeting an E3 ligase, thus influencing cell proliferation and cancer progression. Moreover, miRNAs can promote cell death. The figure provides a summary of tumor suppressors that are regulated by miRNAs.
Figure 2. Illustrates how miRNAs regulate tumor suppressors’ expression or activity. They can also affect their stability and activity by targeting an E3 ligase, thus influencing cell proliferation and cancer progression. Moreover, miRNAs can promote cell death. The figure provides a summary of tumor suppressors that are regulated by miRNAs.
Ijms 25 05865 g002
Table 1. p53 regulation by miRNAs: a more comprehensive view.
Table 1. p53 regulation by miRNAs: a more comprehensive view.
miRNAsFunction to p53Regulation of p53Cancer TypeModelType of EvidenceReferences
miR-122Activates p53 by interacting with p53-regulating factors such as MDM2, SIRT1, YY1.UpregulatesHepatocellular carcinomaIn vitro cell lineqPCR, Western blot[91,92]
miR-34Directly regulated by p53, it represses several cell cycle and proliferation targets.UpregulatesMultiple typesIn vivo xenograft, in vitro cell lineqPCR, Western blot, RIP[79,80,81,82,83,84,85]
miR-29Activates p53 indirectly by targeting MDM2.UpregulatesLung cancerIn vitro cell lineqPCR, Western blot[93,94]
miR-192Activates p53 indirectly by targeting MDM2.UpregulatesColorectal cancerIn vitro cell lineqPCR, Western blot[88,95]
miR-194Works similarly to miR-192 in activating p53.UpregulatesMultiple typesIn vitro cell lineqPCR, Western blot[69,89,96,97,98]
miR-215Along with miR-192 and miR-194, MDM2 is targeted to activate p53.UpregulatesMultiple typesIn vitro cell lineqPCR, Western blot[69,88,96,97,98]
miR-605Activates p53 indirectly by targeting MDM2.UpregulatesLung cancerIn vitro cell lineWestern blot[69,96,97,98]
miR-214Positively regulates p53 by suppressing PSMD10 and ASF1B, direct targets of miR-214.UpregulatesMultiple typesIn vitro cell lineqPCR, Western blot[89,99]
miR-145Activates p53 indirectly by targeting MDM2.UpregulatesMultiple typesIn vitro cell lineqPCR, Western blot[100]
miR-1827Activates p53 indirectly by targeting MDM2.UpregulatesMultiple typesIn vivo xenograft, in vitro cell lineqPCR, Western blot[101]
miR-18bActivates p53 indirectly by targeting MDM2.UpregulatesmelanomaIn vivo xenograft, in vitro cell lineqPCR, Western blot[102]
miR-339-5pActivates p53 indirectly by targeting MDM2.UpregulatesMultiple typesIn vitro cell lineqPCR, Western blot[103]
miR-1301Activates p53 indirectly by targeting UBE4B.UpregulatesMultiple typesIn vitro cell lineqPCR, Western blot[76]
miR-449Induces p53 activation by inhibiting HDAC1 and SIRT1.UpregulatesGastric cancerIn vitro cell lineqPCR, Western blot[104]
miR-32Activates p53 indirectly by targeting MDM2 and TSC1.UpregulatesGlioblastoma In vitro cell lineqPCR, Western blot[105]
miR-542-3pActivates p53 indirectly by targeting MDM2 and RPS23.UpregulatesNeuroblastomaIn vitro cell lineqPCR, Western blot[106]
miR-661Activates p53 indirectly by targeting MDM2 and MDM4.UpregulatesBreast cancerIn vitro cell lineqPCR, Western blot[75]
miR-3928Activates p53 indirectly, by targeting MDM2, CD44, DDX3X, HMGA2, etc. UpregulatesGlioblastomaIn vivo xenograft, in vitro cell lineqPCR, Western blot[107]
miR-25Negatively regulates p53 by directly binding to its 3′UTR.DownregulatesMultiple myelomaIn vitro cell lineqPCR, Western blot[70]
miR-30dNegatively regulates p53, similar to miR-25.DownregulatesMultiple myelomaIn vitro cell lineqPCR, Western blot[70]
miR-125bTarget p53 and several components of the p53 pro-apoptotic network, including BAK1 and PUMA.DownregulatesBrain cancerIn vitro cell line, zebrafishqPCR, Western blot[108]
miR-504Directly targets p53’s 3′UTR, negatively regulating it.DownregulatesMultiple typesIn vivo xenograft, in vitro cell lineqPCR, Western blot[87]
miR-380-5pDirectly targets p53’s 3’UTR, acting as a negative regulator.DownregulatesBrain cancerIn vivo xenograft, in vitro cell lineqPCR, Western blot[109]
miR-92Negatively regulates p53.DownregulatesProstate cancerIn vitro cell lineqPCR, Western blot[110]
miR-141Negatively affects p53 by binding to its 3′UTR.DownregulatesGlioblastomaIn vitro cell lineqPCR, Western blot[111]
miR-33Directly targets p53’s 3′UTR.DownregulatesHepatocellular carcinomaIn vitro cell lineqPCR, Western blot[112]
miR-19bDirectly targets p53’s 3′UTR.DownregulatesMultiple typesIn vivo xenograft, in vitro cell lineqPCR, Western blot[113]
miR-98Directly targets p53’s 3′UTR.DownregulatesLung cancerIn vitro cell lineqPCR, Western blot[114]
miR-150-5pDirectly targets p53’s 3′UTR.DownregulatesColorectal cancerIn vitro cell lineqPCR, Western blot[115]
miR-375Directly targets p53’s 3′UTR.DownregulatesGastric cancerIn vitro cell lineqPCR, Western blot[116]
miR-1285Directly targets p53’s 3′UTR.DownregulatesMultiple typesIn vitro cell lineqPCR, Western blot[117]
miR-3151Directly targets p53’s 3′UTR.DownregulatesAcute leukemiaIn vitro cell lineqPCR, Western blot[118]
Please note that this is not an exhaustive list, and numerous other miRNAs have been shown to regulate p53 expression.
Table 2. p63 regulation by miRNAs.
Table 2. p63 regulation by miRNAs.
miRNAFunction to p63Regulation of p63Cancer TypeModelType of EvidenceReferences
miR-574-3pInhibited by iASPP, p63 expression is maintained due to inhibition of miR-574-3p, which targets p63.UpregulatesSkin cancerIn vitro cell lineqPCR, Western blot[144]
miR-720Like miR-574-3p, its expression is inhibited by iASPP to maintain p63 expression.UpregulatesSkin cancerIn vitro cell lineqPCR, Western blot[144]
miR-144Activates p63 indirectly by targeting Itch.UpregulatesBreast cancerIn vitro cell lineqPCR, Western blot[143]
miR-203Represses p63 expression in supra-basal epithelial cells, contributing to the border between progenitor and differentiated epithelial cells.DownregulatesHead and neck cancerIn vitro cell lineqPCR, Western blot[145]
miR-92Targets ΔNp63α and β, affecting expression in keratinocyte and myeloid cells, respectively.DownregulatesLeukemiaIn vitro cell lineqPCR, Western blot, RIP[146]
miR-302Suppresses p63 expression in germ cells.DownregulatesTesticular cancerIn vitro cell lineqPCR, Western blot[147]
miR-130bDirectly targets p63’s 3′UTR.DownregulatesMultiple typesIn vitro cell lineqPCR, Western blot[148]
miR-181aDirectly targets p63’s 3′UTR.DownregulatesSquamous carcinomaIn vitro cell lineqPCR, Western blot[149]
miR-196aDirectly targets p63’s 3′UTR.DownregulatesBreast cancerIn vitro cell lineqPCR, Western blot[150]
miR-21Directly targets p63’s 3′UTR.DownregulatesMultiple typesIn vivo xenograft, in vitro cell lineqPCR, Western blot[151,152]
miR-30b/cDirectly targets p63’s 3′UTR.DownregulatesGlioblastomaIn vitro cell lineqPCR, Western blot[152]
miR-374aDirectly targets p63’s 3′UTR.DownregulatesSquamous carcinomaIn vitro cell lineqPCR, Western blot[149]
miR-519aDirectly targets p63’s 3′UTR.DownregulatesSquamous carcinomaIn vitro cell lineqPCR, Western blot[149]
miR-630Directly targets p63’s 3′UTR.DownregulatesSquamous carcinomaIn vitro cell lineqPCR, Western blot[149]
miR-885-3pDirectly targets p63’s 3′UTR.DownregulatesSquamous carcinomaIn vitro cell lineqPCR, Western blot[149]
miR-138Directly targets p63’s 3′UTR.DownregulatesMultiple typesIn vitro cell lineqPCR, Western blot[148]
miR-944Directly targets p63’s 3′UTR.DownregulatesCervical cancerIn vitro cell lineqPCR, Western blot[142]
Table 3. Regulation of PTEN by miRNAs.
Table 3. Regulation of PTEN by miRNAs.
miRNAsFunction to PTENRegulation of PTENCancer TypeModelType of EvidenceReferences
miR-21Promotes cancer growth and metastasis by targeting PTEN.DownregulatesVarious, including lung and breastIn vitro cell line, in vivo xenograftqPCR, Western blot[30,153,155,156,157,158,159]
miR-214Reduces PTEN expression, enhancing cell survival and drug resistance.DownregulatesNasopharyngeal carcinomaIn vitro cell lineqPCR, Western blot[162]
miR-93It is associated with resistance to cisplatin in ovarian cancer cells by targeting PTEN.DownregulatesBreast cancerIn vitro cell lineqPCR, Western blot[161]
miR-130aLinked to cisplatin resistance by directly targeting PTEN,DownregulatesBreast cancerIn vitro cell lineqPCR, Western blot[163]
miR-498Directly targeting the 3’UTR of PTEN.DownregulatesBreast cancer (TNBC)In vitro cell lineqPCR, Western blot[168]
miR-221/222Directly targeting the 3’UTR of PTEN.DownregulatesNon-small cell lung cancerIn vitro cell lineqPCR, Western blot[164,165]
miR-26aDirectly targeting the 3’UTR of PTEN.DownregulatesMultiple typesIn vitro cell lineqPCR, Western blot[169,170]
miR-19-3pTargets PTEN to regulate cervical cancer cell proliferation, invasion, and Autophagy.DownregulatesCervical cancerIn vitro cell lineqPCR, Western blot[153,171]
miR-216a/217promote metastasis by targeting PTEN.DownregulatesOvarian cancerIn vitro cell lineqPCR, Western blot[172,173]
miR-22Directly targeting the 3’UTR of PTEN.DownregulatesMultiple typesIn vitro cell lineqPCR, Western blot[174,175]
miR-182-5pInhibits cell proliferation and Invasion of Breast Cancer Cells by targeting PTEN.DownregulatesBreast cancerIn vitro cell lineqPCR, Western blot[176]
miR-153activated the AKT signaling and downregulated FOXO1 transcriptional activity. By targeting PTEN.DownregulatesProstate cancerIn vitro cell lineqPCR, Western blot[177,178]
miR-106bDirectly targeting the 3’UTR of PTEN.DownregulatesBreast cancerIn vitro cell lineqPCR, Western blot[179,180]
miR-382-5pDirectly targeting the 3’UTR of PTEN.DownregulatesLeukemiaIn vitro cell lineqPCR, Western blot[181]
miR-144/451Promote cell proliferation by targeting PTEN.DownregulatesInsulinomasIn vitro cell lineqPCR, Western blot[182]
miR-29promotes osteosarcoma cell proliferation and migration by targeting PTEN.DownregulatesOsteosarcomaIn vitro cell lineqPCR, Western blot[183]
miR-200aDirectly targeting the 3’UTR of PTEN.DownregulatesOvarian cancerIn vitro cell lineqPCR, Western blot[184]
miR-552Directly targeting the 3’UTR of PTEN.DownregulatesOvarian cancerIn vitro cell lineqPCR, Western blot[185]
miR-186Directly targeting the 3’UTR of PTEN.DownregulatesOvarian cancerIn vitro cell lineqPCR, Western blot[186]
miR-371Directly targeting the 3’UTR of PTEN.DownregulatesHepatocellular carcinomaIn vitro cell lineqPCR, Western blot[187]
miR-155Directly targeting the 3’UTR of PTEN.DownregulatesHepatocellular carcinomaIn vitro cell lineqPCR, Western blot[188]
miR-106aDirectly targeting the 3’UTR of PTEN.DownregulatesProstate cancerIn vitro cell lineqPCR, Western blot[189]
miR-183-5pDirectly targeting the 3’UTR of PTEN.DownregulatesNon-small cell lung cancerIn vitro cell lineqPCR, Western blot[190]
miR-486Directly targeting the 3’UTR of PTEN.DownregulatesPancreatic cancerIn vitro cell lineqPCR, Western blot[191]
miR-142-5pDirectly targeting the 3’UTR of PTEN.DownregulatesIntrahepatic cholangiocarcinomaIn vitro cell lineqPCR, Western blot[192]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Wu, H.H.; Leng, S.; Sergi, C.; Leng, R. How MicroRNAs Command the Battle against Cancer. Int. J. Mol. Sci. 2024, 25, 5865. https://doi.org/10.3390/ijms25115865

AMA Style

Wu HH, Leng S, Sergi C, Leng R. How MicroRNAs Command the Battle against Cancer. International Journal of Molecular Sciences. 2024; 25(11):5865. https://doi.org/10.3390/ijms25115865

Chicago/Turabian Style

Wu, Hong Helena, Sarah Leng, Consolato Sergi, and Roger Leng. 2024. "How MicroRNAs Command the Battle against Cancer" International Journal of Molecular Sciences 25, no. 11: 5865. https://doi.org/10.3390/ijms25115865

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

Article Metrics

Back to TopTop