Next Article in Journal
Tackling Insomnia Symptoms through Vestibular Stimulation in Patients with Breast Cancer: A Perspective Paper
Previous Article in Journal
Ready for Prime Time? Dendritic Cells in High-Grade Gliomas
Previous Article in Special Issue
Hypoxia-Inducible Factor-1: A Novel Therapeutic Target for the Management of Cancer, Drug Resistance, and Cancer-Related Pain
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

HIF-1-Induced hsa-miR-429: Understanding Its Direct Targets as the Key to Developing Cancer Diagnostics and Therapies

by
Sylwia Bartoszewska
1,
Jakub Sławski
2,
James F. Collawn
3 and
Rafal Bartoszewski
2,*
1
Department of Inorganic Chemistry, Medical University of Gdansk, 80-416 Gdansk, Poland
2
Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, 50-383 Wroclaw, Poland
3
Department of Cell, Developmental and Integrative Biology, University of Alabama, Birmingham, AL 35294, USA
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(11), 2903; https://doi.org/10.3390/cancers15112903
Submission received: 9 May 2023 / Revised: 21 May 2023 / Accepted: 24 May 2023 / Published: 25 May 2023
(This article belongs to the Special Issue The Role of Hypoxia Inducible Factor (HIF) in Cancers)

Abstract

:

Simple Summary

In this review, we discuss how miRNAs play a critical role in the regulation of mRNA stability and translation, how determining the direct targets of miRNAs in complex networks is extremely difficult, and how translating specific miRNAs to the clinic often results in failure. This has led to concerns that this approach may not be feasible. Using hsa-miR-429 as an example, we discuss the limitations encountered in the development of efficient miRNAs-related therapies and diagnostic approaches and how this can be improved. We also provide a literature analysis of the verified hsa-miR-429 targets in various human research models. A meta-analysis of this work should provide better insights into the role of hsa-miR-429 in cancer diagnosis and any potential therapeutic approaches.

Abstract

MicroRNAs (miRNAs) play a critical role in the regulation of mRNA stability and translation. In spite of our present knowledge on the mechanisms of mRNA regulation by miRNAs, the utilization and translation of these ncRNAs into clinical applications have been problematic. Using hsa-miR-429 as an example, we discuss the limitations encountered in the development of efficient miRNA-related therapies and diagnostic approaches. The miR-200 family members, which include hsa-miR-429, have been shown to be dysregulated in different types of cancer. Although these miR-200 family members have been shown to function in suppressing epithelial-to-mesenchymal transition, tumor metastasis, and chemoresistance, the experimental results have often been contradictory. These complications involve not only the complex networks involving these noncoding RNAs, but also the problem of identifying false positives. To overcome these limitations, a more comprehensive research strategy is needed to increase our understanding of the mechanisms underlying their biological role in mRNA regulation. Here, we provide a literature analysis of the verified hsa-miR-429 targets in various human research models. A meta-analysis of this work is presented to provide better insights into the role of hsa-miR-429 in cancer diagnosis and any potential therapeutic approach.

1. Introduction

Short, endogenous non-coding double-stranded microRNAs (miRNAs) are involved in the repressive control of mRNA translation [1,2,3,4,5]. Understanding their mechanism of action in modulating mRNA stability and translation has opened new prospects for the diagnosis and therapy of human diseases, including cancers. Notably, the modulation of miRNA levels not only allows for the elimination of dysregulated proteins but also provides the opportunity to restore physiological proteostasis [6,7,8,9,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24]. Despite studies continuously reporting novel examples of disease-related dysregulation of miRNAs levels and novel targets of these molecules, the diagnostic approaches and clinical trials have often been unsuccessful [6,7,25,26,27,28,29,30,31]. These studies have led to skepticism regarding any possible miRNA clinical translational possibilities.
This specificity problem could result from a number of reasons including the following: (1) mammalian miRNAs are not required to be perfectly complementary to their target mRNA sequences; (2) each miRNA can affect up to hundreds of targets; (3) miRNAs can promote indirect effects through the modulation of transcription factors (TFs) and the inhibition of their downstream targets, thus generating false positives [32,33,34]. To make matters worse, mRNA stability and translational efficiency can be modulated by different miRNAs simultaneously. Furthermore, although some miRNAs have been shown to be potent post-transcriptional repressors [35,36,37,38,39,40,41,42,43,44,45,46,47,48], the vast majority of these molecules only provide a modest modulation of the subsequent protein levels and thus serve more as ‘molecular buffers’ than as strong repressors of gene expression [49,50]. Therefore, understanding the complex molecular networks modulated by miRNAs remains a challenging, but mandatory, step towards any successful utilization of these molecules in any therapeutic or diagnostic approach.
In this review, we discuss the molecular networks of miR-429, a member of the miR-200 family that is induced by HIF-1 during hypoxia [51]. miR-429 is commonly associated with the progression and metastatic potential of many human cancers [52,53,54,55,56,57]. The members of the miR-200 family are clustered in two genomic locations: one on chromosome 1 (1p36.33) that comprises miR-200b, miR-200a and miR-429, and the other on chromosome 12 (12p13.31) that includes miR-200c and miR-141 (Figure 1A) [58]. Despite the fact that these locations encode harpin precursors for both 5′ and 3′ miR-200 family members, the 3’ mature miRNA forms are the dominant ones [58], and therefore the -3p postfix is omitted in the majority of reports including in this review.
Although the miR-200 family members have been widely accepted as tumor suppressors, these studies have been controversial with regard to cancer prognoses [52,56,57]. Here, we discuss hsa-miR-429 and the challenges in understanding this miRNA role in cancer development. The first problem is the similarity of seed sequences in all of the miR-200 family members. This makes determining the specificity of each of the different members of this family a challenge. Certainly, not properly addressing this in an experimental model could eventually lead to false conclusions. The seed sequence AAUACUG is common for miRs-200b, miR-200c and miR-429, and there is only a one-nucleotide sequence difference, AACACUG, in miR-200a and miR-141 [23,58]. This identity and similarity make the mRNA target profiles of this family of miRNAs very similar (Figure 1).
As shown in Figure 1B, the vast majority of the predicted miR-429 high-probability targets (top 5%) are common for all of the miR-429 family members. The only difference is the “unique targets” of miR-429 can also be recognized with a probability higher than 95% [59,60]. A similar picture was obtained for miR-429 targets that were verified experimentally (Figure 1C) [61]. Furthermore, the expression of specific miR-200 family members such as hsa-miR-429 is modulated by the cellular context, cellular stress conditions, and the stage of tumor progression and metastasis [52,53,54,55,56,57]. How this relates to the other family members is still unclear.
Although the miR-200 family members including hsa-miR-429 were shown in numerous reports to be dysregulated in different types of cancer and assigned functions in suppressing epithelial–mesenchymal transition (EMT), tumor metastasis and chemoresistance [52,53,54,55,56,57], the data are often contradictory and difficult to understand. Therefore, a systematic analysis of the molecular network of their targets and context is needed. Here, we provide a comprehensive literature and meta-analysis of the reports verifying hsa-miR-429 targets in various human research models.

2. Methods

2.1. Literature Search Strategy

The review was prepared according to recommendations for systematic reviews and meta-analyses [62,63]. This comprehensive literature search included two online databases, PubMed and Scopus, up to February 2023. The terms “miR-429, hsa-miR-429, microRNA-429, miR-429-3p, hsa-miR-429-3p” were used as search identifiers in the relevant literature search.

2.2. Inclusion and Exclusion Criteria

The studies identified in these searches were included for further evaluation when the following criteria were fulfilled: (1) miR-429 was investigated in any type of human cell type; (2) the experimental approach aimed to determine miR-429 direct mRNA targets; (3) the study included experimental confirmation of miR-429 direct binding to the target sequence using a target mask (protector), luciferase reporters or RNA immunoprecipitation; (4) the target validation included miR-429 overexpression by synthetic analogs, such as mimics, pre-miRs, or vectors and inhibition by specific miR-429 inhibitors. Furthermore, studies were excluded according to following criteria: (1) non-English articles; (2) retracted articles; (3) errata and other types of articles such as conference records, abstracts, reviews or metanalyses; (4) insufficient data to confirm direct miR-429 interactions; (5) the prediction data were obtained from TCGA and GEO datasets. The schematic workflow of the data selection is presented in Figure 2.

2.3. Data Extraction

All included studies were independently identified by two investigators (S.B. and R.B.), and uncertain data were verified by the third author (J.C.F). The following information was collected: (1) authors names, (2) DOI, (3) publication year, (4) sample types, (5) cell types, (6) type of miRNA, (7) type of biochemical strategy used to verify hsa-miR-429 targets, (8) assays used to evaluate functional role of identified interactions and (9) translational models.

2.4. Prediction of mRNA Targets and Gene Ontology Analysis

The miRNA–mRNA target interactions were predicted and analyzed with the use of mirDIP (microRNA Data Integration Portal and miRTarBase). The mirDIP database provides nearly 152 million human miRNA–target predictions, which were collected across 30 different resources and does not accumulate prediction bias toward biological processes or pathways [60,61]. miRTarBase contains miRNA–target interactions which were collected by manually surveying pertinent literature [61]. The Enrichr (https://amp.pharm.mssm.edu/Enrichr/, accessed on 2 February 2023) [64] and WebGestalt (https://www.webgestalt.org/, accessed on 2 February 2023) [65] webservers were applied to the analysis results into the “Gene Ontology categories” with the selection based on a Q value < 0.05. Furthermore, the analyses were limited to experimentally verified interactions, and no extended gene enrichment set analyses were performed.

3. Experimental Approaches to Define the Molecular Network of hsa-miR-429

The identification and solid experimental verification of both the mRNA targets as well as the transcriptional and posttranscriptional regulators of a specific miRNAs remain a major obstacle to understand their role in cellular signaling pathways. These types of analyses are mandatory for the further efficient translation of miRNAs into therapeutic approaches. Hence, although many reports regarding the mRNA targets and the biological function of hsa-miR-429 accumulated over the last decade, here we focused only on the rigorous ones that were based on experimental verification.
There is no single experimental method, unfortunately, that provides researchers with total confidence that any miRNA binding to mRNA is physiologically relevant. Therefore, multiple independent experimental approaches are required. For example, miRNA overexpression with the use of synthetic analogs (miRNA mimics containing the sequences that are identical to the guide strands of the mature miRNAs) results in the dramatic and non-physiological overexpression (from hundred- to thousandfold) of these molecules in cells. This level of overexpression oversaturates the RNA-induced silencing complexes (RISC complexes) and results in binding to mRNA targets that may not be physiologically relevant [66,67]. Alternative strategies to limit the miRNA overexpression effects on target gene expression in a more physiologically appropriate way include designing longer synthetic miRNA precursors such as pre-miRNAs that undergo Dicer processing in the cytoplasm or the use of pri-miRNAs that require delivery to the nucleus for processing [68,69,70,71].
A similar false positive bias can be generated by luciferase reporter approaches in which easily transfected human cell lines such as HEK293 cells serve as an environment for vectors expressing target 3’UTRs attached to the luciferase reporter as well as the miRNA analogs being tested [67,72,73,74,75,76]. In this type of system, both the target sequences and the miRNAs are elevated to nonphysiological levels, and the cell serves as the source of the RNAi machinery. Thus, even if mutagenesis, which often relies on removing the entire target sequence, confirms a potential miRNA–mRNA binding, it still needs to be considered as an overexpression approach.
Another approach for monitoring miRNA–mRNA interactions can be achieved by the utilization of specific miRNA inhibitors that impair the endogenous miRNA levels. AntagomiRs are synthetic chemically modified ssRNAs that are fully complementary miRNAs and thereby effectively sequester mature miRNAs [77,78]. The mechanism of miRNA inhibition by these molecules depends on the type of chemical modifications used. For example, high-affinity inhibitors form heteroduplexes with the targeted miRNAs, whereas low-affinity ones promote miRNA degradation [79,80,81,82]. There are concerns even about the use of antagomiRs. For example, this approach affects the entire miRNA target network, and this could allow other genes to modulate the expression of the candidate target and therefore lead to false positives based on indirect effects [83].
A more specific and appropriate approach to verify mRNA targets utilizes target protectors (morpholinos) that prevent endogenous miRNA binding to an mRNA region containing a specific miR seed sequence [84]. These molecules are ssRNAs (of ~25 bases), chemically modified to prevent them from triggering the RNAi pathway, and are complementary to an mRNA target sequence (of at least 14–15 contiguous bases). This approach blocks the relevant mRNA in question and provides the needed specificity that the other approaches lack [8,84,85,86,87]. That being said, although they provide a better physiological insight into miRNA–mRNA interactions, it remains possible that the binding in the 3′ mRNA regions may contain other miRNA target sites that could still be affected. If the target protector blocks the significant decreases in the target mRNA expression, then this concern is minimized. This target protector approach, unfortunately, would not differentiate between the targets of miR-429 and other miR-200 family members.
Another viable approach to verify miRNA–mRNA interactions relies on analyzing the composition of RISC complexes (RNA Immunoprecipitation Chip Assay, RIP assay) [88,89]. In these assays, immunoprecipitation of Argonaute RISC Catalytic Component 2 (Ago2), containing the other RISC components including all of the complexed mRNAs and miRNAs, is carried out. The verification of the miRNA and mRNA of interest is verified by qPCR or Next-Generation Sequencing (NGS) [50]. Although these assays are a solid prognostic of functional miRNA–mRNA interactions, the RISC complexes reflect all of the bound miRNAs and mRNAs in the cells. Therefore, the limitation of this approach is that, though it identifies the mRNA and miRNA contained in the RISC, it does not demonstrate that the two components actually exist in a functional hybrid. Furthermore, numerous research applications ignore the fact that even if the miRNA-related mRNA expression levels do change, it does not mean that this will lead to a change in the protein levels or the phenotype [90].
Taken together, the verification of miRNA–mRNA interactions is a complex challenge that should be based on multiple independent experimental strategies (Figure 3). One should also consider process-related changes in the endogenous miRNA levels as well as the complexity of signaling pathways that the miRNA’s target mRNA plays. Despite the large number of reports regarding hsa-miR-429 function, our inclusion criteria resulted in a final selection of 47 genes (Figure 2) that were identified either as a direct target of hsa-miR-429 or as transcriptional or posttranscriptional modulators, e.g., transcription factors or lncRNAs (sponges), respectively (Table 1).
To obtain insights into the biological role of the hsa-miR-429 network, we performed gene enrichment analyses using two well established integrative web tools, Enrichr and Webgestalt [64,65], and applied a strict False Discovery Rate-based (FDR) criteria analysis [133,134]. This approach assigned the hsa-miR-429-related target genes and indicated that this miRNA is an important regulator of processes that are either activated during development or dysregulated in human cancer and cardiovascular diseases.

4. hsa-miR-429 and Cancer

The ability of epithelial cells to reversibly alter the expression of their cell adhesion proteins and their cytoskeleton to become motile mesenchymal cells is manifested during EMT. EMT is an essential and strictly regulated process in normal embryonic development, that includes the establishment of the neural crest and gastrulation, as well as tissue regeneration in adults [135]. Cancer cells, however, utilize an aberrant, partial reactivation of EMT in order to allow for the invasion of primary tumor cells [135,136,137]. Notably, hsa-miR-429 along with the other miR-200 family members have been identified as important modulators of the EMT through their interaction with the zinc finger E-box binding homeobox proteins ZEB1 and ZEB2 [53,56,91,92,93,96]. Furthermore, follow-up studies extended the regulatory role of hsa-miR-429 in this pathway to direct network interactions with AKT serine/threonine kinase 1 (AKT1) [108], the enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2) [123], the notch receptor 1 (NOTCH1) [111], occludin (OCLN) [106], RB binding protein 4, chromatin remodeling factor (RBBP4) [132], tight junction protein 1 (TJP1) [106] and hypoxia inducible factor 1 subunit alpha (HIF1A) [101,102,103].
In many cancers models, the repression of the endogenous levels of hsa-miR-429 leads to increased levels of ZEB1 and ZEB2 and consequently to reduced E-cadherin synthesis and increased cellular motility [138]. This mechanism is even more interesting in light of reports showing that all miR-200 family members are directly repressed transcriptional targets during ZEB1 and ZEB2 activation [139,140]. Thus, hsa-miR-429, along with the others miR-200 family members, and ZEB1 or ZEB2 function within a double-negative feedback loop, and consequently, changes in the levels of the miRNA or the ZEB factors have dramatic effects on each other. Thus, depending on the cellular signaling network, a cell can switch to either the epithelial or the mesenchymal phenotype [138].
The cellular signals that initiate increased ZEB factor expression and the related feedback loop activation include the TGF-β and TNF-α signaling pathways and the cellular responses from steroid hormones and hypoxia [138]. Interestingly, hsa-miR-429 participates in a negative feedback loop as an important direct regulator of HIF-1 signaling in normal human endothelial cells [101,103,141]. Thus, hsa-miR-429 could work as an additional safety mechanism by preventing extensive HIF-1-driven expression of ZEB factors during hypoxia. However, the consequences of such a mechanism for both the development processes and the hypoxic cancer microenvironment will require further validation.
Establishing the specific impact of hsa-miR-429 on ZEB factor expression is very challenging, since all miR-200 members are part of this negative feedback loop [138]. Furthermore, cancer-specific epigenetic changes such as silencing of Achaete-Scute Homolog (ACLS2) [96] or increasing the levels of lncRNA sponges such as MAPKAPK5 Antisense RNA 1 (MAPKAPK5-AS1) [94], circular RNA protein tyrosine kinase 2 (circPTK2) [115], circLIFR [116] or metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) [99,117] that limit the miR-200 family expression, were also shown to correlate with an increased metastatic potential. The role of hsa-miR-429, however, in modulating EMT is not limited to the ZEB factors, since other direct targets include other important positive transcriptional regulators of this pathway such as EZH2 [142], RBBP4 [143] and NOTCH1 [144]. Thus, depending on the cancer type, the reduction of hsa-miR-429 levels could also accelerate the EMT completely independently of the ZEB pathway.
EZH2 has been shown to increase the expression of circRNA_0082835, an hsa-miR-429 sponge in cancer cells, creating another negative feedback loop-based regulation between EZH2 and hsa-miR-429 [123]. Interestingly, however, AKT1, that is a negative regulator of EMT [145], has been shown to be a direct target of hsa-miR-429, along with tight junction protein 1 (TJP1) and occludin (OCLN) that are repressed during the EMT process [146,147]. Thus, reduced hsa-miR-429 levels can lead to increased AKT1, TJP1 and OCLN levels and consequently limit the EMT, counteracting this miRNA effect on ZEB factors and other EMT-favoring targets.
Furthermore, HIF-1 acts as a master regulator of cellular adaptation to low oxygen levels and can also directly stimulate the expression of ZEB1 [148,149,150] and modify Notch1 signaling [151,152,153]. Given that HIF1A forms a negative feedback loop with has-miR-429 [101], the interpretation of this miRNA role in the context of the hypoxic tumor microenvironment can be extremely challenging, given the HIF-1/ZEB1/Notch1 pathway crosstalk. Additionally, the expression of many lncRNAs that were proposed to reduce hsa-miR-429 levels, including MALAT1, LINC01234, XIST, MAPKAPK5-AS1 and PVT1, and were shown to be altered during hypoxia should be considered [154,155,156,157,158,159,160,161].
Both hypoxia and the activation of HIF-1 signaling are common features of tumor microenvironments [162,163,164]. They lead to both hsa-miR-429 induction and the accumulation of lncRNAs that are capable of reducing this miRNA expression levels. Given the inhibitory effect of hsa-miR-429 on EMT, we speculate that HIF-1 tumor-specific impact on lncRNAs levels, e.g., MALAT1 or XIST, could potentially help cancer cells to overcome the hsa-miR-429-related blockaid and shift the cells towards EMT. Furthermore, since oncogenic alterations in miRNA levels are often associated with drug resistance [165,166] and chemoresponses [167,168], the cancer-specific deregulation of HIF-1-related hsa-miR-429 induction may have a significant impact on therapeutic efficiency. This hypothesis, however, will require in-depth mechanistic tumor-specific studies in order to understand the impact of HIF-1-related tuning of hsa-miR-429 expression on the metastatic potential and the possibility of any viable drug responses.
This complex contradictory mechanism of hsa-miR-429 involvement in EMT signaling (Figure 4) requires further study to determine the usefulness of miR-200 family members to modulate EMT-related responses. Nevertheless, considering that miRNA acts as a buffer in gene expression profiles, the complex interactions of hsa-miR-429 may be critical for the proper tuning of developmental processes that include neurogenesis. Indeed, ASCL2, which is an important regulator of cellular differentiation [169], has been shown to induce the expression of hsa-miR-429 [96]. Obviously, further research is necessary to determine if feedback loop-based changes in hsa-miR-429 levels can work as an EMT switch during development.

5. Responses to Hypoxia

In both physiological and pathological conditions, the process of cellular adaptation to insufficient oxygen levels is based on the wide transcriptional reprogramming of gene expression. Reprograming is needed in order to alter metabolism toward glycolysis, prevent cell death and restore oxygen homeostasis through the activation of angiogenesis and erythropoiesis [170]. The master regulators of these cellular activities are hypoxia-inducible factors (HIFs) that during hypoxia form active heterodimers of alpha subunits with their oxygen-independent beta subunits [171,172,173]. The HIF-1 complex mediates the initial and very wide reprograming of gene expression during acute hypoxia [174]. Extensive cellular adaptation to low oxygen levels, along with the intensive effort to restore the normal oxygen pressure through the upregulation of angiogenesis and erythropoiesis [50,51,174], creates a potential risk of oxidative damage (reoxygenation injury) upon the rapid restoration of the cellular oxygen levels [175].
During chronic hypoxia in endothelial cells in vascularized organs such as the lung, heart, placenta and kidney [176,177,178], the other HIF isoform, HIF-2α (EPAS1), eventually replaces HIF-1 and modulates its activity to a more balanced adaptative response [173,179,180,181]. Importantly, passing the transcriptional signal from HIF-1 to HIF-2 is necessary for proper vascularization during development [182,183]. However, the switch from HIF-1 and HIF-2 is also utilized by many cancer cells as an adaptive mechanism to the chronic oxygen deficiency in the tumor microenvironment [179,184]. Furthermore, another tissue-specific HIF alpha isoform, HIF-3α-2, accumulates during prolonged hypoxia in order to either support erythropoietin production in hepatoma cells [185] or induce the mTOR pathway in endothelial cells and eventually to direct the cells toward apoptosis [186,187].
Thus, the switch from HIF-1 to HIF-2 and HIF-3 is necessary for the proper adjustment of cellular homeostasis towards survival (HIF-2) or apoptosis (HIF-3) during prolonged hypoxia. Importantly, during hypoxia, the levels of both HIF-1 and HIF-3 are dynamically modulated by hsa-miR-429. Interestingly, both HIF3A and hsa-miR-429 are induced by HIF-1 [101,103,188], while HIF1A and HIF3A are direct targets of hsa-miR-429 [101,103]. Notably, HIF-2 levels are independent of HIF-1, whereas the EPAS1 transcript (HIF-2) seems to be less prone to miRNA-related degradation [83].
This relationship between the HIFs and hsa-miR-429 has two important functional implications in endothelial cells. First, HIF-1 creates a negative regulatory loop with hsa-miR-429 that is responsible for decreasing the HIF1A message during hypoxia as the EPAS1 levels increase. Secondly, it prevents an extensive HIF3A accumulation during acute hypoxia that could potentially limit HIF-1 activity and induce cell death. In contrast, during prolonged hypoxia, it attenuates the continuous HIF-1 transcriptional activity through the dominant negative properties of HIF-3α [189] as well as promotes the initiation of apoptosis [186].
Taken together, in endothelial cells at least, hsa-miR-429 serves as a crucial modulator of HIF signaling. This miRNA maintains low levels of HIF1A and HIF3A expression during normal oxygen conditions, while during the early stages of hypoxia, when HIF-1α is dramatically elevated, hsa-miR-429 elevated expression maintains low levels of HIF3A mRNA and protein. During prolonged hypoxia, the HIF-1α levels decline via hsa-miR-429 mediated effects on HIF1A mRNA levels. This then leads to a reduction in has-miR-429 levels and a subsequent elevation of the HIF3A message and HIF-3α protein accumulation over time. Thus, hsa-miR-429 regulates the transitional switch between HIF-1-, HIF-2- and HIF-3-mediated transcriptional reprogramming in endothelial cells during hypoxia (Figure 5). Although the described mechanisms are well characterized in normal endothelial cells, it should be emphasized that they may also be active in the vessels of hypoxic solid tumors [190,191]. Therefore, the tumor microenvironment may be an important determinant of hsa-miR-429 pathological function. Importantly, additional hsa-miR-429 mRNA targets were reported in hypoxia-exposed cancer cells, stressing this miRNA role in controlling both HIF signaling and hypoxia-induced angiogenesis. Similar to what was found for EMT, AKT1 as an hsa-miR-429 target, was shown to limit HIF-1α expression [192]. Thus, the induction of hsa-miR-429 by HIF-1 during acute hypoxia can serve a similar role as that of HIF-3 and prevent AKT1-related HIF-1 inactivation early during hypoxia, while this miRNA’s decline during prolonged hypoxia could facilitate the HIF switch.
Additional levels of hsa-miR-429 impact on the HIF pathway can also be provided by the direct modulation of PTEN levels [129]. PTEN is a repressor of HIF-1 transcriptional activity [193], and interestingly, the loss of expression of this gene is observed in many types of cancers [194]. Hence, hypoxic changes in hsa-miR-429 can also modulate the extent of HIF-1 transcriptional activity. Furthermore, the hypoxic elevation of hsa-miR-429 can also contribute to HIF-1-mediated reduction in peroxisome proliferator-activated receptor alpha (PPAR-α) expression during hypoxia [195]. This mechanism complements the role of the HIF switch in the adaptative response to hypoxia, since a decrease in PPARα will increase the efficiency of cellular oxygen use through the modulation of mitochondrial metabolism by decreasing the beta-oxidation of fatty acids [196].
hsa-miR-429 can also modulate the extent of hypoxia-induced angiogenesis indirectly by controlling the extent of HIF signaling and thus VEGF expression [197]. hsa-miR-429 can also modulate angiogenesis through direct interactions with other regulators including v-Crk avian sarcoma virus CT10 oncogene homolog-like (CRKL) [198], fibroblast growth factor receptor substrate 2 (FRS2) [199], tissue inhibitor of metalloproteinases 2 (TIMP2) [200] and Janus kinase 1 (JAK1) [201]. Since some of these hsa-miR-429 targets either have antiangiogenic functions (e.g., TIMP2) [200] or are cancer-type specific, further studies are necessary in order to understand how this miRNA modulates angiogenesis in different types of cancer.
As already mentioned, numerous lncRNAs that sequester (sponges) hsa-miR-429, such as MALAT1, LINC01234, XIST, MAPKAPK5-AS1 and PVT1, are dysregulated by hypoxia (usually, induced) [154,155,156,157,158,159,160,161] or promote oncological transformation [202]. However, many of them bind many other miRNAs under the very same conditions, including all miR-200 family members [203]. Our understanding of their dynamic changes during hypoxia and their preferences and efficiency in binding specific miRNAs in more physiologically relevant models remains limited. Thus, more studies are needed to establish how effective these lncRNAs are in terms of reducing the cellular hsa-miR-429 expression levels when other mRNA targets are available.

6. Discussion

Taken together, hsa-miR-429 appears to be critical for the regulation of cellular adaptation to hypoxia and EMT. Both of these processes are extremely important for development and for maintaining cellular homeostasis. Since these aspects of cell biology are dysregulated in human pathologies such as cardiovascular disease and cancer, understanding the role of hsa-miR-429 could be a starting point for novel therapeutic and diagnostic strategies. Nevertheless, understanding the complex and overlapping networks of hsa-miR-429 mRNA target genes that are master transcriptional factors for EMT and HIF pathways remains a difficult task. Performing mechanistic studies that would involve all of them simultaneously is currently beyond the current research possibilities.
Importantly, this problem can be partially solved by the proper identification of hsa-miR-429 direct targets and by analyzing the kinetics and priority of specific miRNA–mRNA interactions within signaling pathways, as well as the related changes in hsa-miR-429 levels and other miR-200 family members that share the same or very similar seed sequences. There are only a few reports, however, that utilize such an approach. Unfortunately, a plethora of proposed hsa-miR-429 targets were not rigorously verified and are often downstream targets of transcription factors that are themselves direct targets of hsa-miR-429. Furthermore, in terms of cancer cell lines, both miR-200 family members as well as the related signaling pathways can be affected by oncogenic transformation in a cancer/patient-specific manner, and generalizing the observation regarding hsa-miR-429 role requires direct verification in various cancer cell lines.
Taken together, despite hsa-miR-429 role (along with that of the other miR-200 family members) as an important regulator of both physiological and pathological processes, more rigorous and extensive studies are needed to decipher the network of has-miR-429 molecular targets and to estimate this miRNA family’s role on cellular homeostasis. Furthermore, since both EMT and the cellular response to hypoxia are important aspects of development, hsa-miR-429 may also be involved in the modulation of these processes. Nevertheless, only a few studies have considered the role of this miRNA in development [152,204,205], and therefore more research is needed to verify this regulatory role.

7. Conclusions

Although the role of miRNAs in the regulation of both physiological and pathological aspects of cell biology is undisputable, translation of these ncRNAs into the clinic remains difficult. As discussed here with the hsa-miR-429 example, the main limitations of the development of efficient miRNAs-related therapies and diagnostic approaches are not only the complex networks involving these noncoding RNAs, but also the difficulties in identifying false positives. Thus, to provide miRNA-related clinical benefits, more rigorous and comprehensive research is needed to increase our understanding of the mechanisms underlying their biological role in models that are as physiologically relevant.

Author Contributions

S.B., J.S., J.F.C. and R.B. wrote, read and revised the final version of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

Ago2 (Argonaute 2: AKT1 AKT serine/threonine kinase 1), ASCL2 (Achaete-Scute Family BHLH Transcription Factor 2), BMI1 (BMI1 proto-oncogene, polycomb ring finger), CD274 (CD274 molecule), CELF2 (CUGBP Elav-like family member 2), circADAM9 (circular RNA ADAM9_013), circEZH2 (circular RNA EZH2_027), circLIFR (circular RNA LIFR_002), circPTK2 (circular RNA PTK2_063), circPVT1 (circular RNA PVT1), CRKL (CRK like proto-oncogene adaptor protein), DOI (digital object identifier), E2F3 (E2F transcription factor 3), EMT (epithelial–mesenchymal transition), EPAS1 (endothelial PAS domain protein 1) (hypoxia inducible factor 2)), EZH2 (enhancer of zeste 2 polycomb repressive complex 2 subunit), FDR (false discovery rate), FOXK1 (forkhead box K1), FRS2 (fibroblast growth factor receptor substrate 2), HIF (hypoxia-inducible factor), HIF1A (hypoxia-inducible factor 1 subunit alpha), HIF3A (hypoxia-inducible factor 3 subunit alpha), HOXA9 (homeobox A9), IKBKB (inhibitor of nuclear factor kappa B kinase subunit beta), JAK1 (Janus kinase 1), LINC01234 (long intergenic non-protein coding RNA 1234), LINC01303 (long intergenic non-protein coding RNA 1303), lncRNA (long non-coding RNA), LRP4 (LDL receptor-related protein 4), MALAT1 (metastasis-associated lung adenocarcinoma transcript 1), MAPKAPK5-AS1 (MAPKAPK5 antisense RNA 1), miR (microRNA), mirDIP (microRNA Data Integration Portal), miRNA (microRNA), MSC-AS1 (MSC antisense RNA 1), mTOR (mammalian target of rapamycin), NGS (next-generation sequencing), NOTCH1 (notch receptor 1), OCLN (occludin), PAK6 (p21 (RAC1) activated kinase 6), PPARA (peroxisome proliferator activated receptor alpha), premiR (pre-microRNA), pre-miRNA (pre-microRNA), pri-miRNA (primary microRNA), PTEN (phosphatase and tensin homolog), qPCR (quantitative (real-time) polymerase chain reaction), RAB18 (RAB18, member RAS oncogene family), RAB23 (RAB23, member RAS oncogene family), RASSF8 (Ras association domain family member 8), RBBP4 (RB binding protein 4, chromatin remodeling factor), RIP (RNA immunoprecipitation), RISC (RNA-induced silencing complex), RNAi (RNA interference), RND3 (Rho family GTPase 3), RNF185-AS1 (RNF185 antisense RNA 1), SCAMP1 (secretory carrier membrane protein 1), SNHG6 (small nucleolar RNA host gene 6), ssRNA (single-stranded RNA), SYNJ1 (synaptojanin 1), TF (transcription factor), TGF-β transforming growth factor beta, TIMP2 TIMP metallopeptidase inhibitor 2, TJP1 (tight junction protein 1), TNF-α (tumor necrosis factor alpha, TRIB2 (tribbles pseudokinase 2), TRIM33 (tripartite motif containing 33), UTR (untranslated region), VEGF (vascular endothelial growth factor), XIST (X inactive specific transcript), ZEB1 (zinc finger E-box binding homeobox 1), ZEB2 (zinc finger E-box binding homeobox 2).

References

  1. Fire, A.; Xu, S.; Montgomery, M.K.; Kostas, S.A.; Driver, S.E.; Mello, C.C. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature 1998, 391, 806–811. [Google Scholar] [CrossRef] [PubMed]
  2. Bartel, D.P. Micrornas: Genomics, biogenesis, mechanism, and function. Cell 2004, 116, 281–297. [Google Scholar] [CrossRef] [PubMed]
  3. Elbashir, S.M.; Harborth, J.; Lendeckel, W.; Yalcin, A.; Weber, K.; Tuschl, T. Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature 2001, 411, 494–498. [Google Scholar] [CrossRef] [PubMed]
  4. Elbashir, S.M.; Lendeckel, W.; Tuschl, T. RNA interference is mediated by 21- and 22-nucleotide RNAs. Genes Dev. 2001, 15, 188–200. [Google Scholar] [CrossRef]
  5. Caplen, N.J.; Parrish, S.; Imani, F.; Fire, A.; Morgan, R.A. Specific inhibition of gene expression by small double-stranded RNAs in invertebrate and vertebrate systems. Proc. Natl. Acad. Sci. USA 2001, 98, 9742–9747. [Google Scholar] [CrossRef]
  6. 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]
  7. Lam, J.K.W.; Chow, M.Y.T.; Zhang, Y.; Leung, S.W.S. siRNA Versus miRNA as Therapeutics for Gene Silencing. Mol. Ther.—Nucleic Acids 2015, 4, e252. [Google Scholar] [CrossRef]
  8. Bartoszewska, S.; Cabaj, A.; Dabrowski, M.; Collawn, J.F.; Bartoszewski, R. MiR-34c-5p modulates X-box-binding protein 1 (XBP1) expression during the adaptive phase of the unfolded protein response. FASEB J. 2019, 33, 11541–11554. [Google Scholar] [CrossRef]
  9. Zhao, Z.; Lin, C.Y.; Cheng, K. SiRNA- and miRNA-based therapeutics for liver fibrosis. Transl. Res. J. Lab. Clin. Med. 2019, 214, 17–29. [Google Scholar] [CrossRef]
  10. Miroshnichenko, S.; Patutina, O. Enhanced inhibition of tumorigenesis using combinations of miRNA-targeted therapeutics. Front. Pharmacol. 2019, 10, 488. [Google Scholar] [CrossRef]
  11. Bansal, P.; Kumar, A.; Chandna, S.; Arora, M.; Bansal, R. Targeting miRNA for therapeutics using a micronome based method for identification of miRNA-mRNA pairs and validation of key regulator miRNA. Methods Mol. Biol. 2018, 1823, 185–195. [Google Scholar] [PubMed]
  12. Li, D.J.; Sun, C.C. Editorial: Towards miRNA based therapeutics for lung cancer. Curr. Pharm. Des. 2018, 23, 5971–5972. [Google Scholar] [CrossRef] [PubMed]
  13. Wu, M.; Wang, G.; Tian, W.; Deng, Y.; Xu, Y. MiRNA-based therapeutics for lung cancer. Curr. Pharm. Des. 2018, 23, 5989–5996. [Google Scholar] [CrossRef]
  14. Wen, M.M. Getting miRNA Therapeutics into the Target Cells for Neurodegenerative Diseases: A Mini-Review. Front. Mol. Neurosci. 2016, 9, 129. [Google Scholar] [CrossRef] [PubMed]
  15. Zhu, K.; Liu, D.; Lai, H.; Li, J.; Wang, C. Developing miRNA therapeutics for cardiac repair in ischemic heart disease. J. Thorac. Dis. 2016, 8, E918–E927. [Google Scholar] [CrossRef]
  16. Kouri, F.M.; Ritner, C.; Stegh, A.H. miRNA-182 and the regulation of the glioblastoma phenotype—Toward miRNA-based precision therapeutics. Cell Cycle 2015, 14, 3794–3800. [Google Scholar] [CrossRef] [PubMed]
  17. Mishra, S.; Yadav, T.; Rani, V. Exploring miRNA based approaches in cancer diagnostics and therapeutics. Crit. Rev. Oncol. 2016, 98, 12–23. [Google Scholar] [CrossRef]
  18. Bernardo, B.C.; Ooi, J.Y.Y.; Lin, R.C.Y.; McMullen, J.R. miRNA therapeutics: A new class of drugs with potential therapeutic applications in the heart. Future Med. Chem. 2015, 7, 1771–1792. [Google Scholar] [CrossRef]
  19. Nouraee, N.; Mowla, S.J. miRNA therapeutics in cardiovascular diseases: Promises and problems. Front. Genet. 2015, 6, 232. [Google Scholar] [CrossRef]
  20. Kwekkeboom, R.F.J.; Lei, Z.; Doevendans, P.A.; Musters, R.J.P.; Sluijter, J.P.G. Targeted delivery of miRNA therapeutics for cardiovascular diseases: Opportunities and challenges. Clin. Sci. 2014, 127, 351–365. [Google Scholar] [CrossRef]
  21. Pereira, D.; Rodrigues, P.; Borralho, P.; Rodrigues, C. Delivering the promise of miRNA cancer therapeutics. Drug Discov. Today 2013, 18, 282–289. [Google Scholar] [CrossRef] [PubMed]
  22. Ford, L.P. Using synthetic miRNA mimics for diverting cell fate: A possibility of miRNA-based therapeutics? Leuk. Res. 2006, 30, 511–513. [Google Scholar] [CrossRef] [PubMed]
  23. Bartoszewska, S.; Kamysz, W.; Jakiela, B.; Sanak, M.; Króliczewski, J.; Bebok, Z.; Bartoszewski, R.; Collawn, J.F. miR-200b downregulates CFTR during hypoxia in human lung epithelial cells. Cell. Mol. Biol. Lett. 2017, 22, 23. [Google Scholar] [CrossRef]
  24. Kalinowski, L.; Janaszak-Jasiecka, A.; Siekierzycka, A.; Bartoszewska, S.; Woźniak, M.; Lejnowski, D.; Collawn, J.F.; Bartoszewski, R. Posttranscriptional and transcriptional regulation of endothelial nitric-oxide synthase during hypoxia: The role of microRNAs. Cell. Mol. Biol. Lett. 2016, 21, 16. [Google Scholar] [CrossRef]
  25. Kleinman, M.E.; Yamada, K.; Takeda, A.; Chandrasekaran, V.; Nozaki, M.; Baffi, J.Z.; Albuquerque, R.J.C.; Yamasaki, S.; Itaya, M.; Pan, Y.; et al. Sequence- and target-independent angiogenesis suppression by siRNA via TLR3. Nature 2008, 452, 591–597. [Google Scholar] [CrossRef] [PubMed]
  26. Wilkinson, T.; Vaishnaw, A.; Cehelsky, J.; Meyers, R.; Nochur, S.; Harrison, L.; Meeking, P.; Mann, A.; Moane, E.; Oxford, J.; et al. Viral Load Drives Disease in Humans Experimentally Infected with Respiratory Syncytial Virus. Am. J. Respir. Crit. Care Med. 2010, 182, 1305–1314. [Google Scholar] [CrossRef]
  27. DeVincenzo, J.; Lambkin-Williams, R.; Wilkinson, T.; Cehelsky, J.; Nochur, S.; Walsh, E.; Meyers, R.; Gollob, J.; Vaishnaw, A. A randomized, double-blind, placebo-controlled study of an RNAi-based therapy directed against respiratory syncytial virus. Proc. Natl. Acad. Sci. USA 2010, 107, 8800–8805. [Google Scholar] [CrossRef]
  28. Davis, M.E.; Zuckerman, J.E.; Choi, C.H.J.; Seligson, D.; Tolcher, A.; Alabi, C.A.; Yen, Y.; Heidel, J.D.; Ribas, A. Evidence of RNAi in humans from systemically administered siRNA via targeted nanoparticles. Nature 2010, 464, 1067–1070. [Google Scholar] [CrossRef]
  29. Zuckerman, J.E.; Davis, M.E. Clinical experiences with systemically administered siRNA-based therapeutics in cancer. Nature reviews. Drug Discov. 2015, 14, 843–856. [Google Scholar] [CrossRef]
  30. Ackley, K.L. Are we there yet? An update on oligonucleotide drug development. Chim. Oggi. 2016, 34, Xxxv–Xxxviii. [Google Scholar]
  31. Setten, R.L.; Rossi, J.J.; Han, S.-P. The current state and future directions of RNAi-based therapeutics. Nat. Rev. Drug Discov. 2019, 18, 421–446. [Google Scholar] [CrossRef] [PubMed]
  32. Gurtan, A.M.; Sharp, P.A. The Role of miRNAs in Regulating Gene Expression Networks. J. Mol. Biol. 2013, 425, 3582–3600. [Google Scholar] [CrossRef] [PubMed]
  33. Xu, H.; Zhang, Y.; Xie, S.J.; Xu, S.J.; Zhou, H.; Qu, L.H. Argonaute hits-clip decodes microRNA-mRNA interaction maps during heart development. Cardiology 2013, 126, 62. [Google Scholar]
  34. Chi, S.W.; Zang, J.B.; Mele, A.; Darnell, R.B. Argonaute hits-clip decodes microRNA-mRNA interaction maps. Nature 2009, 460, 479–486. [Google Scholar] [CrossRef] [PubMed]
  35. Olejniczak, M.; Kotowska-Zimmer, A.; Krzyzosiak, W. Stress-induced changes in miRNA biogenesis and functioning. Cell. Mol. Life Sci. 2018, 75, 177–191. [Google Scholar] [CrossRef]
  36. van Rooij, E.; Sutherland, L.B.; Qi, X.; Richardson, J.A.; Hill, J.; Olson, E.N. Control of Stress-Dependent Cardiac Growth and Gene Expression by a MicroRNA. Science 2007, 316, 575–579. [Google Scholar] [CrossRef]
  37. Leung, A.; Sharp, P.A. MicroRNA Functions in Stress Responses. Mol. Cell 2010, 40, 205–215. [Google Scholar] [CrossRef]
  38. Mendell, J.T.; Olson, E.N. MicroRNAs in Stress Signaling and Human Disease. Cell 2012, 148, 1172–1187. [Google Scholar] [CrossRef]
  39. Ge, X.-L.; Wang, J.-L.; Liu, X.; Zhang, J.; Liu, C.; Guo, L. Inhibition of miR-19a protects neurons against ischemic stroke through modulating glucose metabolism and neuronal apoptosis. Cell. Mol. Biol. Lett. 2019, 24, 37. [Google Scholar] [CrossRef]
  40. Lu, G.; Zhang, Y. MicroRNA-340-5p suppresses non-small cell lung cancer cell growth and metastasis by targeting ZNF503. Cell. Mol. Biol. Lett. 2019, 24, 34. [Google Scholar] [CrossRef]
  41. Fu, Y.; Lin, L.; Xia, L. MiR-107 function as a tumor suppressor gene in colorectal cancer by targeting transferrin receptor 1. Cell. Mol. Biol. Lett. 2019, 24, 31. [Google Scholar] [CrossRef] [PubMed]
  42. Liu, L.; Zuo, Y.; Xu, Y.; Zhang, Z.; Li, Y.; Pang, J. MiR-613 inhibits proliferation and invasion and induces apoptosis of rheumatoid arthritis synovial fibroblasts by direct down-regulation of DKK1. Cell. Mol. Biol. Lett. 2019, 24, 8. [Google Scholar] [CrossRef] [PubMed]
  43. Cai, Y.; Li, Y. Upregulation of miR-29b-3p protects cardiomyocytes from hypoxia-induced apoptosis by targeting TRAF5. Cell. Mol. Biol. Lett. 2019, 24, 27. [Google Scholar] [CrossRef] [PubMed]
  44. Li, S.; Sun, Z.; Chen, T.; Pan, J.; Shen, Y.; Chen, X.; Zhou, X.; Cheng, R.; Yang, Y. The role of miR-431-5p in regulating pulmonary surfactant expression in vitro. Cell. Mol. Biol. Lett. 2019, 24, 25. [Google Scholar] [CrossRef] [PubMed]
  45. Sun, L.; Lian, J.X.; Meng, S. MiR-125a-5p promotes osteoclastogenesis by targeting TNFRSF1B. Cell. Mol. Biol. Lett. 2019, 24, 23. [Google Scholar] [CrossRef] [PubMed]
  46. Zeng, Q.; Wang, Y.; Gao, J.; Yan, Z.; Li, Z.; Zou, X.; Li, Y.; Wang, J.; Guo, Y. miR-29b-3p regulated osteoblast differentiation via regulating IGF-1 secretion of mechanically stimulated osteocytes. Cell. Mol. Biol. Lett. 2019, 24, 11. [Google Scholar] [CrossRef]
  47. Yang, Y.; Bao, Y.; Yang, G.-K.; Wan, J.; Du, L.-J.; Ma, Z.-H. MiR-214 sensitizes human colon cancer cells to 5-FU by targeting Hsp27. Cell. Mol. Biol. Lett. 2019, 24, 22. [Google Scholar] [CrossRef]
  48. Ma, F.; Lin, P.; Chen, Q.; Lu, X.; Zhang, Y.E.; Wu, C.-I. Direct measurement of pervasive weak repression by microRNAs and their role at the network level. BMC Genom. 2018, 19, 362. [Google Scholar] [CrossRef]
  49. Vidigal, J.A.; Ventura, A. The biological functions of miRNAs: Lessons from in vivo studies. Trends Cell Biol. 2015, 25, 137–147. [Google Scholar] [CrossRef]
  50. Moszynska, A.; Jaskiewicz, M.; Serocki, M.; Cabaj, A.; Crossman, D.K.; Bartoszewska, S.; Gebert, M.; Dabrowski, M.; Collawn, J.F.; Bartoszewski, R. The hypoxia-induced changes in miRNA-mRNA in RNA-induced silencing complexes and HIF-2 induced miRNAs in human endothelial cells. FASEB J. 2022, 36, e22412. [Google Scholar] [CrossRef]
  51. Serocki, M.; Bartoszewska, S.; Janaszak-Jasiecka, A.; Ochocka, R.J.; Collawn, J.F.; Bartoszewski, R. miRNAs regulate the HIF switch during hypoxia: A novel therapeutic target. Angiogenesis 2018, 21, 183–202. [Google Scholar] [CrossRef] [PubMed]
  52. Guo, C.M.; Liu, S.Q.; Sun, M.Z. miR-429 as biomarker for diagnosis, treatment and prognosis of cancers and its potential action mechanisms: A systematic literature review. Neoplasma 2020, 67, 215–228. [Google Scholar] [CrossRef] [PubMed]
  53. Cavallari, I.; Ciccarese, F.; Sharova, E.; Urso, L.; Raimondi, V.; Silic-Benussi, M.; D’agostino, D.M.; Ciminale, V. The miR-200 Family of microRNAs: Fine Tuners of Epithelial-Mesenchymal Transition and Circulating Cancer Biomarkers. Cancers 2021, 13, 5874. [Google Scholar] [CrossRef]
  54. Klicka, K.; Grzywa, T.M.; Mielniczuk, A.; Klinke, A.; Włodarski, P.K. The role of miR-200 family in the regulation of hallmarks of cancer. Front. Oncol. 2022, 12, 965231. [Google Scholar] [CrossRef] [PubMed]
  55. Feng, B.; Wang, R.; Chen, L.-B. Review of MiR-200b and cancer chemosensitivity. Biomed. Pharmacother. 2012, 66, 397–402. [Google Scholar] [CrossRef]
  56. Huang, G.-L.; Sun, J.; Lu, Y.; Liu, Y.; Cao, H.; Zhang, H.; Calin, G.A. MiR-200 family and cancer: From a meta-analysis view. Mol. Asp. Med. 2019, 70, 57–71. [Google Scholar] [CrossRef] [PubMed]
  57. Diaz-Riascos, Z.V.; Ginesta, M.M.; Fabregat, J.; Serrano, T.; Busquets, J.; Buscail, L.; Cordelier, P.; Capellá, G. Expression and Role of MicroRNAs from the miR-200 Family in the Tumor Formation and Metastatic Propensity of Pancreatic Cancer. Mol. Ther.—Nucleic Acids 2019, 17, 491–503. [Google Scholar] [CrossRef]
  58. Senfter, D.; Madlener, S.; Krupitza, G.; Mader, R.M. The microRNA-200 family: Still much to discover. Biomol. Concepts 2016, 7, 311–319. [Google Scholar] [CrossRef]
  59. Hauschild, A.-C.; Pastrello, C.; Ekaputeri, G.K.A.; Bethune-Waddell, D.; Abovsky, M.; Ahmed, Z.; Kotlyar, M.; Lu, R.; Jurisica, I. MirDIP 5.2: Tissue context annotation and novel microRNA curation. Nucleic Acids Res. 2023, 51, D217–D225. [Google Scholar] [CrossRef]
  60. Tokar, T.; Pastrello, C.; Rossos, A.E.M.; Abovsky, M.; Hauschild, A.C.; Tsay, M.; Lu, R.; Jurisica, I. Mirdip 4.1-integrative database of human microRNA target predictions. Nucleic Acids Res. 2018, 46, D360–D370. [Google Scholar] [CrossRef]
  61. Huang, H.Y.; Lin, Y.C.; Cui, S.; Huang, Y.; Tang, Y.; Xu, J.; Bao, J.; Li, Y.; Wen, J.; Zuo, H.; et al. Mirtarbase update 2022: An informative resource for experimentally validated miRNA-target interactions. Nucleic Acids Res. 2022, 50, D222–D230. [Google Scholar] [CrossRef] [PubMed]
  62. Antes, G.; von Elm, E. The prisma statement—What should be reported about systematic reviews? Dtsch. Med. Wochenschr. 2009, 134, 1619. [Google Scholar] [CrossRef] [PubMed]
  63. Moher, D.; Liberati, A.; Tetzlaff, J.; Altman, D.G.; The PRISMA Group. Preferred Reporting Items for Systematic Reviews and Meta-Analyses: The PRISMA Statement. J. Clin. Epidemiol. 2009, 62, 1006–1012. [Google Scholar] [CrossRef] [PubMed]
  64. Xie, Z.; Bailey, A.; Kuleshov, M.V.; Clarke, D.J.B.; Evangelista, J.E.; Jenkins, S.L.; Lachmann, A.; Wojciechowicz, M.L.; Kropiwnicki, E.; Jagodnik, K.M.; et al. Gene Set Knowledge Discovery with Enrichr. Curr. Protoc. 2021, 1, e90. [Google Scholar] [CrossRef]
  65. Liao, Y.; Wang, J.; Jaehnig, E.J.; Shi, Z.; Zhang, B. WebGestalt 2019: Gene Set Analysis Toolkit with Revamped UIs and APIs. Nucleic Acids Res. 2019, 47, W199–W205. [Google Scholar] [CrossRef]
  66. Zhang, S.; Zhu, F. Comment on: “MicroRNA Mimics or Inhibitors as Antiviral Therapeutic Approaches Against COVID-19”. Drugs 2021, 81, 1691–1692. [Google Scholar] [CrossRef]
  67. Bartoszewski, R.; Sikorski, A.F. Editorial focus: Understanding off-target effects as the key to successful RNAi therapy. Cell. Mol. Biol. Lett. 2019, 24, 69. [Google Scholar] [CrossRef]
  68. Wang, H.; Jiang, Y.; Peng, H.; Chen, Y.; Zhu, P.; Huang, Y. Recent progress in microRNA delivery for cancer therapy by non-viral synthetic vectors. Adv. Drug Deliv. Rev. 2015, 81, 142–160. [Google Scholar] [CrossRef]
  69. Terasawa, K.; Shimizu, K.; Tsujimoto, G. Synthetic pre-miRNA-based shRNA as potent RNAi triggers. J. Nucleic Acids 2011, 2011, 131579. [Google Scholar] [CrossRef]
  70. Zhang, Y.; Wang, Z.; Gemeinhart, R.A. Progress in microRNA delivery. J. Control. Release 2013, 172, 962–974. [Google Scholar] [CrossRef]
  71. Bartoszewski, R.; Sikorski, A.F. Editorial focus: Entering into the non-coding RNA era. Cell. Mol. Biol. Lett. 2018, 23, 45. [Google Scholar] [CrossRef] [PubMed]
  72. Jin, Y.; Chen, Z.; Liu, X.; Zhou, X. Evaluating the microRNA targeting sites by luciferase reporter gene assay. Methods Mol. Biol. 2013, 936, 117–127. [Google Scholar]
  73. Diener, C.; Hart, M.; Fecher-Trost, C.; Knittel, J.; Rheinheimer, S.; Meyer, M.R.; Mayer, J.; Flockerzi, V.; Keller, A.; Meese, E. Outside the limit: Questioning the distance restrictions for cooperative miRNA binding sites. Cell. Mol. Biol. Lett. 2023, 28, 8. [Google Scholar] [CrossRef] [PubMed]
  74. Gebert, M.; Bartoszewska, S.; Janaszak-Jasiecka, A.; Moszyńska, A.; Cabaj, A.; Króliczewski, J.; Madanecki, P.; Ochocka, R.J.; Crossman, D.K.; Collawn, J.F.; et al. PIWI proteins contribute to apoptosis during the UPR in human airway epithelial cells. Sci. Rep. 2018, 8, 16431. [Google Scholar] [CrossRef] [PubMed]
  75. Gebert, M.; Sobolewska, A.; Bartoszewska, S.; Cabaj, A.; Crossman, D.K.; Króliczewski, J.; Madanecki, P.; Dąbrowski, M.; Collawn, J.F.; Bartoszewski, R. Genome-wide mRNA profiling identifies X-box-binding protein 1 (XBP1) as an IRE1 and PUMA repressor. Cell. Mol. Life Sci. 2021, 78, 7061–7080. [Google Scholar] [CrossRef] [PubMed]
  76. Bartoszewski, R.; Gebert, M.; Janaszak-Jasiecka, A.; Cabaj, A.; Króliczewski, J.; Bartoszewska, S.; Sobolewska, A.; Crossman, D.K.; Ochocka, R.; Kamysz, W.; et al. Genome-wide mRNA profiling identifies RCAN1 and GADD45A as regulators of the transitional switch from survival to apoptosis during ER stress. FEBS J. 2020, 287, 2923–2947. [Google Scholar] [CrossRef]
  77. Stenvang, J.; Petri, A.; Lindow, M.; Obad, S.; Kauppinen, S. Inhibition of microRNA function by antimiR oligonucleotides. Silence 2012, 3, 1. [Google Scholar] [CrossRef]
  78. Esau, C.C. Inhibition of microRNA with antisense oligonucleotides. Methods 2008, 44, 55–60. [Google Scholar] [CrossRef]
  79. Davis, S.; Propp, S.; Freier, S.M.; Jones, L.E.; Serra, M.J.; Kinberger, G.; Bhat, B.; Swayze, E.E.; Bennett, C.F.; Esau, C. Potent inhibition of microRNA in vivo without degradation. Nucleic Acids Res. 2009, 37, 70–77. [Google Scholar] [CrossRef]
  80. Elmén, J.; Lindow, M.; Schütz, S.; Lawrence, M.; Petri, A.; Obad, S.; Lindholm, M.; Hedtjärn, M.; Hansen, H.F.; Berger, U.; et al. LNA-mediated microRNA silencing in non-human primates. Nature 2008, 452, 896–899. [Google Scholar] [CrossRef]
  81. Torres, A.G.; Fabani, M.M.; Vigorito, E.; Gait, M.J. MicroRNA fate upon targeting with anti-miRNA oligonucleotides as revealed by an improved Northern-blot-based method for miRNA detection. RNA 2011, 17, 933–943. [Google Scholar] [CrossRef]
  82. Krützfeldt, J.; Kuwajima, S.; Braich, R.; Rajeev, K.G.; Pena, J.; Tuschl, T.; Manoharan, M.; Stoffel, M. Specificity, duplex degradation and subcellular localization of antagomirs. Nucleic Acids Res. 2007, 35, 2885–2892. [Google Scholar] [CrossRef] [PubMed]
  83. Jaskiewicz, M.; Moszynska, A.; Gebert, M.; Collawn, J.F.; Bartoszewski, R. EPAS1 resistance to miRNA-based regulation contributes to prolonged expression of hif-2 during hypoxia in human endothelial cells. Gene 2023, 868, 147376. [Google Scholar] [CrossRef] [PubMed]
  84. Staton, A.A.; Giraldez, A.J. Use of target protector morpholinos to analyze the physiological roles of specific miRNA-mRNA pairs in vivo. Nat. Protoc. 2011, 6, 2035–2049. [Google Scholar] [CrossRef] [PubMed]
  85. Summerton, J.E. Morpholino, siRNA, and S-DNA Compared: Impact of Structure and Mechanism of Action on Off-Target Effects and Sequence Specificity. Curr. Top. Med. Chem. 2007, 7, 651–660. [Google Scholar] [CrossRef] [PubMed]
  86. Hudziak, R.M.; Barofsky, E.; Barofsky, D.F.; Weller, D.L.; Huang, S.-B.; Weller, D.D. Resistance of Morpholino Phosphorodiamidate Oligomers to Enzymatic Degradation. Antisense Nucleic Acid Drug Dev. 1996, 6, 267–272. [Google Scholar] [CrossRef]
  87. Summerton, J. Morpholino antisense oligomers: The case for an RNase H-independent structural type. Biochim. Biophys. Acta 1999, 1489, 141–158. [Google Scholar] [CrossRef]
  88. Gagliardi, M.; Matarazzo, M.R. RIP: RNA Immunoprecipitation. Methods Mol. Biol. 2016, 1480, 73–86. [Google Scholar] [CrossRef]
  89. Jayaseelan, S.; Doyle, F.; Tenenbaum, S.A. Profiling post-transcriptionally networked mRNA subsets using RIP-Chip and RIP-Seq. Methods 2014, 67, 13–19. [Google Scholar] [CrossRef]
  90. Liu, Y.; Beyer, A.; Aebersold, R. On the Dependency of Cellular Protein Levels on mRNA Abundance. Cell 2016, 165, 535–550. [Google Scholar] [CrossRef]
  91. Gregory, P.A.; Bert, A.G.; Paterson, E.L.; Barry, S.C.; Tsykin, A.; Farshid, G.; Vadas, M.A.; Khew-Goodall, Y.; Goodall, G.J. The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nat. Cell Biol. 2008, 10, 593–601. [Google Scholar] [CrossRef] [PubMed]
  92. Liu, X.; Liu, Y.; Wu, S.; Shi, X.; Li, L.; Zhao, J.; Xu, H. Tumor-Suppressing Effects of miR-429 on Human Osteosarcoma. Cell Biochem. Biophys. 2014, 70, 215–224. [Google Scholar] [CrossRef] [PubMed]
  93. Shi, M.; Chen, X.; Li, H.; Zheng, L. Delta-tocotrienol suppresses the migration and angiogenesis of trophoblasts in preeclampsia and promotes their apoptosis via miR-429/ ZEB1 axis. Bioengineered 2021, 12, 1861–1873. [Google Scholar] [CrossRef] [PubMed]
  94. Peng, Z.; Ouyang, X.; Wang, Y.; Fan, Q. MAPKAPK5-AS1 drives the progression of hepatocellular carcinoma via regulating mir-429/zeb1 axis. BMC Mol. Cell. Biol. 2022, 23, 21. [Google Scholar] [CrossRef]
  95. Wang, Y.; Dong, X.; Hu, B.; Wang, X.J.; Wang, Q.; Wang, W.L. The effects of microRNA-429 on inhibition of cervical cancer cells through targeting zeb1 and crkl. Biomed. Pharmacother. 2016, 80, 311–321. [Google Scholar] [CrossRef]
  96. Tian, Y.; Pan, Q.; Shang, Y.; Zhu, R.; Ye, J.; Liu, Y.; Zhong, X.; Li, S.; He, Y.; Chen, L.; et al. MicroRNA-200 (miR-200) cluster regulation by achaete scute-like 2 (ASCL2): Impact on the epithelial-mesenchymal transition in colon cancer cells. J. Biol. Chem. 2014, 289, 36101–36115. [Google Scholar] [CrossRef]
  97. Shen, J.; Hong, L.; Yu, D.; Cao, T.; Zhou, Z.; He, S. LncRNA XIST promotes pancreatic cancer migration, invasion and EMT by sponging miR-429 to modulate ZEB1 expression. Int. J. Biochem. Cell Biol. 2019, 113, 17–26. [Google Scholar] [CrossRef]
  98. Sun, B.; Zheng, X.; Ye, W.; Zhao, P.; Ma, G. LncRNA linc01303 promotes the progression of oral squamous cell carcinomas via the mir-429/ZEB1/EMT axis. J. Oncol. 2021, 2021, 7974012. [Google Scholar] [CrossRef]
  99. Liu, X.; Zhao, W.; Wang, X. Inhibition of long non-coding RNA MALAT1 elevates microRNA-429 to suppress the progression of hypopharyngeal squamous cell carcinoma by reducing ZEB1. Life Sci. 2020, 262, 118480. [Google Scholar] [CrossRef]
  100. Liu, D.; Zhang, M.; Song, Y.; Yang, N. RNF185 antisense RNA 1 (RNF185-AS1) promotes proliferation, migration, and invasion in papillary thyroid carcinoma. Anti-Cancer Drugs 2022, 33, 595–606. [Google Scholar] [CrossRef]
  101. Bartoszewska, S.; Kochan, K.; Piotrowski, A.; Kamysz, W.; Ochocka, R.J.; Collawn, J.F.; Bartoszewski, R. The hypoxia-inducible miR-429 regulates hypoxia-inducible factor-1alpha expression in human endothelial cells through a negative feedback loop. FASEB J. 2015, 29, 1467–1479. [Google Scholar] [CrossRef] [PubMed]
  102. Ge, L.; Wang, Y.; Cao, Y.; Li, G.; Sun, R.; Teng, P.; Wang, Y.; Bi, Y.; Guo, Z.; Yuan, Y.; et al. MiR-429 improved the hypoxia tolerance of human amniotic cells by targeting HIF-1α. Biotechnol. Lett. 2018, 40, 1477–1486. [Google Scholar] [CrossRef] [PubMed]
  103. Janaszak-Jasiecka, A.; Bartoszewska, S.; Kochan, K.; Piotrowski, A.; Kalinowski, L.; Kamysz, W.; Ochocka, R.J.; Bartoszewski, R.; Collawn, J.F. miR-429 regulates the transition between Hypoxia-Inducible Factor (HIF)1A and HIF3A expression in human endothelial cells. Sci. Rep. 2016, 6, 22775. [Google Scholar] [CrossRef] [PubMed]
  104. Sun, L.; Wang, L.; Luan, S.; Jiang, Y.; Wang, Q. MiR-429 inhibits osteosarcoma progression by targeting HOXA9 through suppressing WNT/beta-catenin signaling pathway. Oncol. Lett. 2020, 20, 2447–2455. [Google Scholar] [CrossRef] [PubMed]
  105. Xue, H.; Tian, G.-Y. MiR-429 regulates the metastasis and EMT of HCC cells through targeting RAB23. Arch. Biochem. Biophys. 2018, 637, 48–55. [Google Scholar] [CrossRef]
  106. Chen, L.; Xue, Y.; Zheng, J.; Liu, X.; Liu, J.; Chen, J.; Li, Z.; Xi, Z.; Teng, H.; Wang, P.; et al. MiR-429 Regulated by Endothelial Monocyte Activating Polypeptide-II (EMAP-II) Influences Blood-Tumor Barrier Permeability by Inhibiting the Expressions of ZO-1, Occludin and Claudin-5. Front. Mol. Neurosci. 2018, 11, 35. [Google Scholar] [CrossRef]
  107. Li, X.; Chen, H.; Wang, S.; Dai, J.; Yan, L.; Wang, J.; Sun, Y. Tacrolimus induces fibroblasts apoptosis and reduces epidural fibrosis by regulating miR-429 and its target of RhoE. Biochem. Biophys. Res. Commun. 2017, 490, 1197–1204. [Google Scholar] [CrossRef]
  108. Su, Z.; Jiang, G.; Chen, J.; Liu, X.; Zhao, H.; Fang, Z.; He, Y.; Jiang, X.; Xu, G. Erratum: MicroRNA-429 inhibits cancer cell proliferation and migration by targeting AKT1 in renal cell carcinoma. Mol. Clin. Oncol. 2020, 13, 92. [Google Scholar]
  109. Tian, X.; Wei, Z.; Wang, J.; Liu, P.; Qin, Y.; Zhong, M. MicroRNA-429 inhibits the migration and invasion of colon cancer cells by targeting pak6/cofilin signaling. Oncol. Rep. 2015, 34, 707–714. [Google Scholar] [CrossRef]
  110. Qiu, M.; Liang, Z.; Chen, L.; Tan, G.; Wang, K.; Liu, L.; Liu, J.; Chen, H. MicroRNA-429 suppresses cell proliferation, epithelial-mesenchymal transition, and metastasis by direct targeting of BMI1 and E2F3 in renal cell carcinoma. Urol. Oncol. 2015, 33, 332.e9–332.e18. [Google Scholar] [CrossRef]
  111. Xu, H.; Jin, L.; Chen, Y.; Li, J. Downregulation of microRNA-429 protects cardiomyocytes against hypoxia-induced apoptosis by increasing Notch1 expression. Int. J. Mol. Med. 2016, 37, 1677–1685. [Google Scholar] [CrossRef] [PubMed]
  112. You, X.; Liu, F.; Zhang, T.; Li, Y.; Ye, L.; Zhang, X. Hepatitis B virus X protein upregulates oncogene Rab18 to result in the dysregulation of lipogenesis and proliferation of hepatoma cells. Carcinogenesis 2013, 34, 1644–1652. [Google Scholar] [CrossRef] [PubMed]
  113. Lv, J.; Guo, T.; Qu, X.; Che, X.; Li, C.; Wang, S.; Gong, J.; Wu, P.; Liu, Y.; Liu, Y.; et al. PD-L1 Under Regulation of miR-429 Influences the Sensitivity of Gastric Cancer Cells to TRAIL by Binding of EGFR. Front. Oncol. 2020, 10, 1067. [Google Scholar] [CrossRef] [PubMed]
  114. Wang, Y.; Liu, J.; Yao, Q.; Wang, Y.; Liu, Z.; Zhang, L. LncRNA SNHG6 promotes wilms’ tumor progression through regulating mir-429/frs2 axis. Cancer Biother. Radiopharm. 2021. [Google Scholar] [CrossRef] [PubMed]
  115. Wang, L.; Tong, X.; Zhou, Z.; Wang, S.; Lei, Z.; Zhang, T.; Liu, Z.; Zeng, Y.; Li, C.; Zhao, J.; et al. Circular RNA hsa_circ_0008305 (circptk2) inhibits tgf-beta-induced epithelial-mesenchymal transition and metastasis by controlling tif1gamma in non-small cell lung cancer. Mol. Cancer 2018, 17, 140. [Google Scholar] [CrossRef]
  116. Wang, J.; Lai, X.; Peng, X. CircLIFR Inhibits Non-small Cell Lung Cancer Progression by Acting as a miR-429 Sponge to Enhance CELF2 Expression. Biochem. Genet. 2023, 61, 725–741. [Google Scholar] [CrossRef]
  117. Shen, F.; Zheng, H.; Zhou, L.; Li, W.; Xu, X. Overexpression of MALAT1 contributes to cervical cancer progression by acting as a sponge of miR-429. J. Cell. Physiol. 2019, 234, 11219–11226. [Google Scholar] [CrossRef]
  118. Bi, M.; Zheng, L.; Chen, L.; He, J.; Yuan, C.; Ma, P.; Zhao, Y.; Hu, F.; Tang, W.; Sheng, M. Ln RNA linc01234 promotes triple-negative breast cancer progression through regulating the miR-429/SYNJ1 axis. Am. J. Transl. Res. 2021, 13, 11399–11412. [Google Scholar]
  119. Luo, N. LncRNA MSC-AS1/miRNA-429 axis mediates growth and metastasis of nasopharyngeal carcinoma via JAK1/STAT3 signaling pathway. Comput. Math. Methods Med. 2022, 2022, 1447207. [Google Scholar] [CrossRef]
  120. Cao, L.; Zhou, X.; Ding, X.; Gao, D. Knockdown of circ-pvt1 inhibits the progression of lung adenocarcinoma and enhances the sensitivity to cisplatin via the miR-429/FOXK1 signaling axis. Mol. Med. Rep. 2021, 24, 684. [Google Scholar] [CrossRef]
  121. Meng, D.-F.; Shao, H.; Feng, C.-B. LINC00894 Enhances the Progression of Breast Cancer by Sponging miR-429 to Regulate ZEB1 Expression. OncoTargets Ther. 2021, 14, 3395–3407. [Google Scholar] [CrossRef] [PubMed]
  122. Chen, Z.; Chen, J.; Wa, Q.; He, M.; Wang, X.; Zhou, J.; Cen, Y. Knockdown of circ_0084043 suppresses the development of human melanoma cells through mir-429/tribbles homolog 2 axis and Wnt/beta-catenin pathway. Life Sci. 2020, 243, 117323. [Google Scholar] [CrossRef] [PubMed]
  123. Sun, Y.; Hou, Z.; Luo, B.; Li, C.; Liu, J.; Liu, J.; Tang, J.; Yao, G. Circular RNA circRNA_0082835 promotes progression and lymphatic metastasis of primary melanoma by sponging microRNA miRNA-429. Bioengineered 2021, 12, 4159–4173. [Google Scholar] [CrossRef] [PubMed]
  124. Zhou, X.; Lu, H.; Li, F.; Hao, X.; Han, L.; Dong, Q.; Chen, X. MicroRNA-429 inhibits neuroblastoma cell proliferation, migration and invasion via the NF-κB pathway. Cell. Mol. Biol. Lett. 2020, 25, 5. [Google Scholar] [CrossRef]
  125. Kawasaki, H.; Amano, H. Anti-inflammatory role of microRNA-429 in human gingival epithelial cells-inhibition of Il-8 production through direct binding to ikkbeta mRNA. Mol. Med. Rep. 2021, 24, 581. [Google Scholar] [CrossRef]
  126. Zhang, Z.; Wen, H.; Peng, B.; Weng, J.; Zeng, F. Hfd-induced TRAF6 upregulation promotes liver cholesterol accumulation and fatty liver development via ezh2-mediated miR-429/pparalpha axis. Molecular therapy. Nucleic Acids 2021, 24, 711–727. [Google Scholar] [CrossRef]
  127. Cheng, Z.; Li, Z.; Ma, K.; Li, X.; Tian, N.; Duan, J.; Xiao, X.; Wang, Y. Long Non-coding RNA XIST Promotes Glioma Tumorigenicity and Angiogenesis by Acting as a Molecular Sponge of miR-429. J. Cancer 2017, 8, 4106–4116. [Google Scholar] [CrossRef]
  128. Lang, Y.; Xu, S.; Ma, J.; Wu, J.; Jin, S.; Cao, S.; Yu, Y. MicroRNA-429 induces tumorigenesis of human non-small cell lung cancer cells and targets multiple tumor suppressor genes. Biochem. Biophys. Res. Commun. 2014, 450, 154–159. [Google Scholar] [CrossRef]
  129. Tang, J.; Li, L.; Huang, W.; Sui, C.; Yang, Y.; Lin, X.; Hou, G.; Chen, X.; Fu, J.; Yuan, S.; et al. MiR-429 increases the metastatic capability of HCC via regulating classic Wnt pathway rather than epithelial–mesenchymal transition. Cancer Lett. 2015, 364, 33–43. [Google Scholar] [CrossRef]
  130. Wang, J.; Wang, C.; Li, Q.; Guo, C.; Sun, W.; Zhao, D.; Jiang, S.; Hao, L.; Tian, Y.; Liu, S.; et al. MiR-429-CRKL axis regulates clear cell renal cell carcinoma malignant progression through sOS1/MEK/ERK/MMP2/MMP9 pathway. Biomed. Pharmacother. 2020, 127, 110215. [Google Scholar] [CrossRef]
  131. Guo, C.; Zhao, D.; Zhang, Q.; Liu, S.; Sun, M.-Z. miR-429 suppresses tumor migration and invasion by targeting CRKL in hepatocellular carcinoma via inhibiting Raf/MEK/ERK pathway and epithelial-mesenchymal transition. Sci. Rep. 2018, 8, 2375. [Google Scholar] [CrossRef] [PubMed]
  132. Li, L.; Tang, J.; Zhang, B.; Yang, W.; LiuGao, M.; Wang, R.; Tan, Y.; Fan, J.; Chang, Y.; Fu, J.; et al. Epigenetic modification of MiR-429 promotes liver tumour-initiating cell properties by targeting Rb binding protein 4. Gut 2015, 64, 156–167. [Google Scholar] [CrossRef]
  133. Colquhoun, D. An investigation of the false discovery rate and the misinterpretation of p-values. R Soc. Open Sci. 2014, 1, 140216. [Google Scholar] [CrossRef] [PubMed]
  134. Ioannidis, J.P.A. Why Most Published Research Findings Are False. PLoS Med. 2005, 2, e124. [Google Scholar] [CrossRef] [PubMed]
  135. Huang, Y.; Hong, W.; Wei, X. The molecular mechanisms and therapeutic strategies of EMT in tumor progression and metastasis. J. Hematol. Oncol. 2022, 15, 129. [Google Scholar] [CrossRef]
  136. Micalizzi, D.S.; Farabaugh, S.M.; Ford, H.L. Epithelial-Mesenchymal Transition in Cancer: Parallels Between Normal Development and Tumor Progression. J. Mammary Gland Biol. Neoplasia 2010, 15, 117–134. [Google Scholar] [CrossRef]
  137. Yeung, K.T.; Yang, J. Epithelial-mesenchymal transition in tumor metastasis. Mol. Oncol. 2017, 11, 28–39. [Google Scholar] [CrossRef]
  138. Brabletz, S.; Brabletz, T. The ZEB/miR-200 feedback loop--a motor of cellular plasticity in development and cancer? EMBO Rep. 2010, 11, 670–677. [Google Scholar] [CrossRef]
  139. Burk, U.; Schubert, J.; Wellner, U.; Schmalhofer, O.; Vincan, E.; Spaderna, S.; Brabletz, T. A reciprocal repression between ZEB1 and members of the miR-200 family promotes EMT and invasion in cancer cells. EMBO Rep. 2008, 9, 582–589. [Google Scholar] [CrossRef]
  140. Bracken, C.P.; Gregory, P.A.; Kolesnikoff, N.; Bert, A.G.; Wang, J.; Shannon, M.F.; Goodall, G.J. A Double-Negative Feedback Loop between ZEB1-SIP1 and the microRNA-200 Family Regulates Epithelial-Mesenchymal Transition. Cancer Res 2008, 68, 7846–7854. [Google Scholar] [CrossRef]
  141. Bartoszewski, R.; Serocki, M.; Janaszak-Jasiecka, A.; Bartoszewska, S.; Kochan-Jamrozy, K.; Piotrowski, A.; Króliczewski, J.; Collawn, J.F. miR-200b downregulates Kruppel Like Factor 2 (KLF2) during acute hypoxia in human endothelial cells. Eur. J. Cell Biol. 2017, 96, 758–766. [Google Scholar] [CrossRef] [PubMed]
  142. Cardenas, H.; Zhao, J.; Vieth, E.; Nephew, K.P.; Matei, D. EZH2 inhibition promotes epithelial-to-mesenchymal transition in ovarian cancer cells. Oncotarget 2016, 7, 84453–84467. [Google Scholar] [CrossRef] [PubMed]
  143. Li, Y.D.; Lv, Z.; Zhu, W.F. RBBP4 promotes colon cancer malignant progression via regulating Wnt/beta-catenin pathway. World J. Gastroenterol. 2020, 26, 5328–5342. [Google Scholar] [CrossRef]
  144. Shao, S.; Zhao, X.; Zhang, X.; Luo, M.; Zuo, X.; Huang, S.; Wang, Y.; Gu, S.; Zhao, X. Notch1 signaling regulates the epithelial–mesenchymal transition and invasion of breast cancer in a Slug-dependent manner. Mol. Cancer 2015, 14, 28. [Google Scholar] [CrossRef] [PubMed]
  145. Li, C.-W.; Xia, W.; Lim, S.-O.; Hsu, J.L.; Huo, L.; Wu, Y.; Li, L.-Y.; Lai, C.-C.; Chang, S.-S.; Hsu, Y.-H.; et al. AKT1 Inhibits Epithelial-to-Mesenchymal Transition in Breast Cancer through Phosphorylation-Dependent Twist1 Degradation. Cancer Res 2016, 76, 1451–1462. [Google Scholar] [CrossRef]
  146. Kyuno, D.; Yamaguchi, H.; Ito, T.; Kono, T.; Kimura, Y.; Imamura, M.; Konno, T.; Hirata, K.; Sawada, N.; Kojima, T. Targeting tight junctions during epithelial to mesenchymal transition in human pancreatic cancer. World J. Gastroenterol. 2014, 20, 10813–10824. [Google Scholar] [CrossRef]
  147. Lee, S.H.; Paek, A.R.; Yoon, K.; Kim, S.H.; Lee, S.Y.; You, H.J. Tight junction protein 1 is regulated by transforming growth factor-beta and contributes to cell motility in nsclc cells. BMB Rep. 2015, 48, 115–120. [Google Scholar] [CrossRef]
  148. Zhu, J.; Huang, Z.; Zhang, M.; Wang, W.; Liang, H.; Zeng, J.; Wu, K.; Wang, X.; Hsieh, J.; Guo, P.; et al. HIF-1α promotes ZEB1 expression and EMT in a human bladder cancer lung metastasis animal model. Oncol. Lett. 2018, 15, 3482–3489. [Google Scholar] [CrossRef]
  149. Yoshimoto, S.; Tanaka, F.; Morita, H.; Hiraki, A.; Hashimoto, S. Hypoxia-induced HIF-1alpha and ZEB1 are critical for the malignant transformation of ameloblastoma via TGF-beta-dependent emt. Cancer Med. 2019, 8, 7822–7832. [Google Scholar] [CrossRef]
  150. Shih, C.-H.; Chuang, L.-L.; Tsai, M.-H.; Chen, L.-H.; Chuang, E.Y.; Lu, T.-P.; Lai, L.-C. Hypoxia-Induced MALAT1 Promotes the Proliferation and Migration of Breast Cancer Cells by Sponging MiR-3064-5p. Front. Oncol. 2021, 11, 658151. [Google Scholar] [CrossRef]
  151. Zheng, X.; Linke, S.; Dias, J.M.; Zheng, X.; Gradin, K.; Wallis, T.P.; Hamilton, B.R.; Gustafsson, M.; Ruas, J.L.; Wilkins, S.; et al. Interaction with factor inhibiting HIF-1 defines an additional mode of cross-coupling between the Notch and hypoxia signaling pathways. Proc. Natl. Acad. Sci. USA 2008, 105, 3368–3373. [Google Scholar] [CrossRef] [PubMed]
  152. Cheng, Y.-L.; Park, J.-S.; Manzanero, S.; Choi, Y.; Baik, S.-H.; Okun, E.; Gelderblom, M.; Fann, D.Y.-W.; Magnus, T.; Launikonis, B.S.; et al. Evidence that collaboration between HIF-1α and Notch-1 promotes neuronal cell death in ischemic stroke. Neurobiol. Dis. 2014, 62, 286–295. [Google Scholar] [CrossRef] [PubMed]
  153. Zhou, X.-L.; Wan, L.; Xu, Q.-R.; Zhao, Y.; Liu, J.-C. Notch signaling activation contributes to cardioprotection provided by ischemic preconditioning and postconditioning. J. Transl. Med. 2013, 11, 251. [Google Scholar] [CrossRef] [PubMed]
  154. Sallé-Lefort, S.; Miard, S.; Nolin, M.-A.; Boivin, L.; Paré, M.-È.; Debigaré, R.; Picard, F. Hypoxia upregulates Malat1 expression through a CaMKK/AMPK/HIF-1α axis. Int. J. Oncol. 2016, 49, 1731–1736. [Google Scholar] [CrossRef]
  155. Wang, H.; Li, Z.; Gao, J.; Liao, Q. Circular RNA circptk2 regulates oxygen-glucose deprivation-activated microglia-induced hippocampal neuronal apoptosis via miR-29b-sOCS-1-JAK2/STAT3-IL-1beta signaling. Int. J. Biol. Macromol. 2019, 129, 488–496. [Google Scholar] [CrossRef]
  156. Zhang, M.-X.; Zhang, L.-Z.; Fu, L.-M.; Yao, H.-H.; Tan, L.; Feng, Z.-H.; Li, J.-Y.; Lu, J.; Pan, Y.-H.; Shu, G.-N.; et al. Positive feedback regulation of lncRNA PVT1 and HIF2α contributes to clear cell renal cell carcinoma tumorigenesis and metastasis. Oncogene 2021, 40, 5639–5650. [Google Scholar] [CrossRef]
  157. Wang, J.; Dong, Z.; Sheng, Z.; Cai, Y. Hypoxia-induced PVT1 promotes lung cancer chemoresistance to cisplatin by autophagy via PVT1/miR-140-3p/ATG5 axis. Cell Death Discov. 2022, 8, 104. [Google Scholar] [CrossRef]
  158. Lin, C.; Zhang, Y.; Chen, Y.; Bai, Y.; Zhang, Y. Long noncoding RNA LINC01234 promotes serine hydroxymethyltransferase 2 expression and proliferation by competitively binding miR-642a-5p in colon cancer. Cell Death Dis. 2019, 10, 137. [Google Scholar] [CrossRef]
  159. Wang, L.; Sun, L.; Liu, R.; Mo, H.; Niu, Y.; Chen, T.; Wang, Y.; Han, S.; Tu, K.; Liu, Q. Long non-coding RNA MAPKAPK5-AS1/PLAGL2/HIF-1alpha signaling loop promotes hepatocellular carcinoma progression. J. Exp. Clin. Cancer Res. 2021, 40, 72. [Google Scholar] [CrossRef]
  160. Hu, C.; Bai, X.; Liu, C.; Hu, Z. Long noncoding RNA XIST participates hypoxia-induced angiogenesis in human brain microvascular endothelial cells through regulating miR-485/SOX7 axis. Am. J. Transl. Res. 2019, 11, 6487–6497. [Google Scholar]
  161. Xie, J. Long Noncoding RNA XIST Regulates Myocardial Infarction via miR-486-5p/SIRT1 Axis. Appl. Biochem. Biotechnol. 2023, 195, 725–734. [Google Scholar] [CrossRef]
  162. Li, Y.; Zhao, L.; Li, X.-F. Hypoxia and the Tumor Microenvironment. Technol. Cancer Res. Treat. 2021, 20, 15330338211036304. [Google Scholar] [CrossRef] [PubMed]
  163. Balamurugan, K. HIF-1 at the crossroads of hypoxia, inflammation, and cancer. Int. J. Cancer 2016, 138, 1058–1066. [Google Scholar] [CrossRef] [PubMed]
  164. Nejad, A.E.; Najafgholian, S.; Rostami, A.; Sistani, A.; Shojaeifar, S.; Esparvarinha, M.; Nedaeinia, R.; Javanmard, S.H.; Taherian, M.; Ahmadlou, M.; et al. The role of hypoxia in the tumor microenvironment and development of cancer stem cell: A novel approach to developing treatment. Cancer Cell Int. 2021, 21, 62. [Google Scholar] [CrossRef] [PubMed]
  165. Majumder, S.; Jacob, S.T. Emerging Role of MicroRNAs in Drug-Resistant Breast Cancer. Gene Expr. 2011, 15, 141–151. [Google Scholar] [CrossRef] [PubMed]
  166. Zhang, Y.; Wang, J. MicroRNAs are important regulators of drug resistance in colorectal cancer. Biol. Chem. 2017, 398, 929–938. [Google Scholar] [CrossRef]
  167. Baxter, D.E.; Allinson, L.M.; Al Amri, W.S.; Poulter, J.A.; Pramanik, A.; Thorne, J.L.; Verghese, E.T.; Hughes, T.A. MiR-195 and Its Target SEMA6D Regulate Chemoresponse in Breast Cancer. Cancers 2021, 13, 5979. [Google Scholar] [CrossRef]
  168. Nakajima, G.; Hayashi, K.; Xi, Y.; Kudo, K.; Uchida, K.; Takasaki, K.; Yamamoto, M.; Ju, J. Non-coding microRNAs hsa-let-7g and hsa-miR-181b are associated with chemoresponse to s-1 in colon cancer. Cancer Genom. Proteom. 2006, 3, 317–324. [Google Scholar]
  169. Bogutz, A.B.; Oh-McGinnis, R.; Jacob, K.J.; Ho-Lau, R.; Gu, T.; Gertsenstein, M.; Nagy, A.; Lefebvre, L. Transcription factor ASCL2 is required for development of the glycogen trophoblast cell lineage. PLoS Genet. 2018, 14, e1007587. [Google Scholar] [CrossRef]
  170. Ziello, J.E.; Jovin, I.S.; Huang, Y. Hypoxia-Inducible Factor (HIF)-1 Regulatory Pathway and its Potential for Therapeutic Intervention in Malignancy and Ischemia. Yale J. Biol. Med. 2007, 80, 51–60. [Google Scholar]
  171. Semenza, G.L. Hypoxia-Inducible Factor 1 (HIF-1) Pathway. Sci. STKE 2007, 2007, cm8. [Google Scholar] [CrossRef] [PubMed]
  172. Salceda, S.; Caro, J. Hypoxia-inducible factor 1α (HIF-1α) protein is rapidly degraded by the ubiquitin-proteasome system under normoxic conditions: Its stabilization by hypoxia depends on redox-induced changes*. J. Biol. Chem. 1997, 272, 22642–22647. [Google Scholar] [CrossRef] [PubMed]
  173. Jaśkiewicz, M.; Moszyńska, A.; Króliczewski, J.; Cabaj, A.; Bartoszewska, S.; Charzyńska, A.; Gebert, M.; Dąbrowski, M.; Collawn, J.F.; Bartoszewski, R. The transition from HIF-1 to HIF-2 during prolonged hypoxia results from reactivation of PHDs and HIF1A mRNA instability. Cell. Mol. Biol. Lett. 2022, 27, 109. [Google Scholar] [CrossRef] [PubMed]
  174. Semenza, G.L. Hypoxia-Inducible Factors in Physiology and Medicine. Cell 2012, 148, 399–408. [Google Scholar] [CrossRef]
  175. Li, C.; Jackson, R.M. Reactive species mechanisms of cellular hypoxia-reoxygenation injury. Am. J. Physiol. Physiol. 2002, 282, C227–C241. [Google Scholar] [CrossRef]
  176. Talks, K.L.; Turley, H.; Gatter, K.C.; Maxwell, P.H.; Pugh, C.W.; Ratcliffe, P.J.; Harris, A.L. The expression and distribution of the hypoxia-inducible factors HIF-1alpha and HIF-2alpha in normal human tissues, cancers, and tumor-associated macrophages. Am. J. Pathol. 2000, 157, 411–421. [Google Scholar] [CrossRef]
  177. Tian, H.; McKnight, S.L.; Russell, D.W. Endothelial PAS domain protein 1 (EPAS1), a transcription factor selectively expressed in endothelial cells. Genes Dev. 1997, 11, 72–82. [Google Scholar] [CrossRef]
  178. Flamme, I.; Fröhlich, T.; von Reutern, M.; Kappel, A.; Damert, A.; Risau, W. HRF, a putative basic helix-loop-helix-PAS-domain transcription factor is closely related to hypoxia-inducible factor-1α and developmentally expressed in blood vessels. Mech. Dev. 1997, 63, 51–60. [Google Scholar] [CrossRef]
  179. Koh, M.Y.; Powis, G. Passing the baton: The HIF switch. Trends Biochem. Sci. 2012, 37, 364–372. [Google Scholar] [CrossRef]
  180. Bartoszewski, R.; Moszynska, A.; Serocki, M.; Cabaj, A.; Polten, A.; Ochocka, R.; Dell’ Italia, L.; Bartoszewska, S.; Kroliczewski, J.; Dabrowski, M.; et al. Primary endothelial cell-specific regulation of hypoxia-inducible factor (HIF)-1 and HIF-2 and their target gene expression profiles during hypoxia. FASEB J. 2019, 33, 7929–7941. [Google Scholar] [CrossRef]
  181. Carmeliet, P.; Dor, Y.; Herbert, J.M.; Fukumura, D.; Brusselmans, K.; Dewerchin, M.; Neeman, M.; Bono, F.; Abramovitch, R.; Maxwell, P.; et al. Role of HIF-1alpha in hypoxia-mediated apoptosis, cell proliferation and tumour angiogenesis. Nature 1998, 394, 485–490. [Google Scholar] [CrossRef] [PubMed]
  182. Hashimoto, T.; Shibasaki, F. Hypoxia-Inducible Factor as an Angiogenic Master Switch. Front. Pediatr. 2015, 3, 33. [Google Scholar] [CrossRef] [PubMed]
  183. Ryan, H.E.; Lo, J.; Johnson, R.S. HIF-1alpha is required for solid tumor formation and embryonic vascularization. EMBO J. 1998, 17, 3005–3015. [Google Scholar] [CrossRef] [PubMed]
  184. Packer, M. Mutual Antagonism of Hypoxia-Inducible Factor Isoforms in Cardiac, Vascular, and Renal Disorders. JACC Basic Transl. Sci. 2020, 5, 961–968. [Google Scholar] [CrossRef] [PubMed]
  185. Tolonen, J.-P.; Heikkilä, M.; Malinen, M.; Lee, H.-M.; Palvimo, J.J.; Wei, G.-H.; Myllyharju, J. A long hypoxia-inducible factor 3 isoform 2 is a transcription activator that regulates erythropoietin. Cell. Mol. Life Sci. 2020, 77, 3627–3642. [Google Scholar] [CrossRef] [PubMed]
  186. Jaskiewicz, M.; Moszynska, A.; Serocki, M.; Kroliczewski, J.; Bartoszewska, S.; Collawn, J.F.; Bartoszewski, R. Hypoxia-inducible factor (HIF)-3a2 serves as an endothelial cell fate executor during chronic hypoxia. EXCLI J. 2022, 21, 454–469. [Google Scholar]
  187. Ravenna, L.; Salvatori, L.; Russo, M.A. HIF3α: The little we know. FEBS J. 2016, 283, 993–1003. [Google Scholar] [CrossRef]
  188. Pasanen, A.; Heikkilä, M.; Rautavuoma, K.; Hirsilä, M.; Kivirikko, K.I.; Myllyharju, J. Hypoxia-inducible factor (HIF)-3α is subject to extensive alternative splicing in human tissues and cancer cells and is regulated by HIF-1 but not HIF-2. Int. J. Biochem. Cell Biol. 2010, 42, 1189–1200. [Google Scholar] [CrossRef]
  189. Maynard, M.A.; Evans, A.J.; Hosomi, T.; Hara, S.; Jewett, M.A.; Ohh, M. Human HIF-3alpha4 is a dominant-negative regulator of HIF-1 and is down-regulated in renal cell carcinoma. FASEB J. 2005, 19, 1396–1406. [Google Scholar] [CrossRef]
  190. Bartoszewska, S.; Collawn, J.F.; Bartoszewski, R. The Role of the Hypoxia-Related Unfolded Protein Response (UPR) in the Tumor Microenvironment. Cancers 2022, 14, 4870. [Google Scholar] [CrossRef]
  191. Harris, A.L. Hypoxia--a key regulatory factor in tumour growth. Nat. Rev. Cancer 2002, 2, 38–47. [Google Scholar] [CrossRef] [PubMed]
  192. Pore, N.; Jiang, Z.; Shu, H.-K.; Bernhard, E.; Kao, G.D.; Maity, A. Akt1 Activation Can Augment Hypoxia-Inducible Factor-1α Expression by Increasing Protein Translation through a Mammalian Target of Rapamycin–Independent Pathway. Mol. Cancer Res. 2006, 4, 471–479. [Google Scholar] [CrossRef] [PubMed]
  193. Emerling, B.M.; Weinberg, F.; Liu, J.-L.; Mak, T.W.; Chandel, N.S. PTEN regulates p300-dependent hypoxia-inducible factor 1 transcriptional activity through Forkhead transcription factor 3a (FOXO3a). Proc. Natl. Acad. Sci. USA 2008, 105, 2622–2627. [Google Scholar] [CrossRef] [PubMed]
  194. Zundel, W.; Schindler, C.; Haas-Kogan, D.; Koong, A.; Kaper, F.; Chen, E.; Gottschalk, A.R.; Ryan, H.E.; Johnson, R.S.; Jefferson, A.B.; et al. Loss of PTEN facilitates HIF-1-mediated gene expression. Genes Dev. 2000, 14, 391–396. [Google Scholar] [CrossRef] [PubMed]
  195. Narravula, S.; Colgan, S.P. Hypoxia-Inducible Factor 1-Mediated Inhibition of Peroxisome Proliferator-Activated Receptor α Expression During Hypoxia. J. Immunol. 2001, 166, 7543–7548. [Google Scholar] [CrossRef] [PubMed]
  196. Cole, M.A.; Abd Jamil, A.H.; Heather, L.C.; Murray, A.J.; Sutton, E.R.; Slingo, M.; Sebag-Montefiore, L.; Tan, S.C.; Aksentijevic, D.; Gildea, O.S.; et al. On the pivotal role of pparalpha in adaptation of the heart to hypoxia and why fat in the diet increases hypoxic injury. FASEB J. 2016, 30, 2684–2697. [Google Scholar] [CrossRef]
  197. Ramakrishnan, S.; Anand, V.; Roy, S. Vascular Endothelial Growth Factor Signaling in Hypoxia and Inflammation. J. Neuroimmune Pharmacol. 2014, 9, 142–160. [Google Scholar] [CrossRef]
  198. Park, T. Crk and CrkL as Therapeutic Targets for Cancer Treatment. Cells 2021, 10, 739. [Google Scholar] [CrossRef]
  199. Chen, P.Y.; Qin, L.; Zhuang, Z.W.; Tellides, G.; Lax, I.; Schlessinger, J.; Simons, M. The docking protein Frs2alpha is a critical regulator of Vegf receptors signaling. Proc. Natl. Acad. Sci. USA 2014, 111, 5514–5519. [Google Scholar] [CrossRef]
  200. Stetler-Stevenson, W.G.; Seo, D.-W. TIMP-2: An endogenous inhibitor of angiogenesis. Trends Mol. Med. 2005, 11, 97–103. [Google Scholar] [CrossRef]
  201. Xue, C.; Xie, J.; Zhao, D.; Lin, S.; Zhou, T.; Shi, S.; Shao, X.; Lin, Y.; Zhu, B.; Cai, X. The JAK/STAT3 signalling pathway regulated angiogenesis in an endothelial cell/adipose-derived stromal cell co-culture, 3D gel model. Cell Prolif. 2017, 50, e12307. [Google Scholar] [CrossRef] [PubMed]
  202. Agostini, M.; Ganini, C.; Candi, E.; Melino, G. The role of noncoding RNAs in epithelial cancer. Cell Death Discov. 2020, 6, 13. [Google Scholar] [CrossRef] [PubMed]
  203. Zhou, Q.; Liu, L.; Zhou, J.; Chen, Y.; Xie, D.; Yao, Y.; Cui, D. Novel Insights into MALAT1 Function as a MicroRNA Sponge in NSCLC. Front. Oncol. 2021, 11, 758653. [Google Scholar] [CrossRef]
  204. Choi, P.S.; Zakhary, L.; Choi, W.-Y.; Caron, S.; Alvarez-Saavedra, E.; Miska, E.A.; McManus, M.; Harfe, B.; Giraldez, A.J.; Horvitz, R.H.; et al. Members of the miRNA-200 Family Regulate Olfactory Neurogenesis. Neuron 2008, 57, 41–55. [Google Scholar] [CrossRef] [PubMed]
  205. Sun, Y.; Ding, S.; Fan, Y.; Shen, F.; Dong, Q.; Zhao, B.; Pan, Y.; Wan, J. MiR-429 inhibits the angiogenesis of human brain microvascular endothelial cells through Snai2-mediated Gsk-3beta/beta-catenin pathway. Comput. Math. Methods Med. 2021, 2021, 6753926. [Google Scholar] [CrossRef]
Figure 1. Genetic organization of the miR-200 family in humans. (A) The chromosome loci of the two clusters of the miR-200 family with their seed sequences are presented in boxes. The seed sequences differ by one nucleotide (indicated in red). (B) Venn diagram representing the distribution overlap of the top 1% of predicted targets (p-value < 0.01, mirDIP webserver) of the miR-200 family members. (C) Venn diagram representing the distribution overlap of experimentally confirmed targets (mirTarbase webserver) of the miR-200 family members.
Figure 1. Genetic organization of the miR-200 family in humans. (A) The chromosome loci of the two clusters of the miR-200 family with their seed sequences are presented in boxes. The seed sequences differ by one nucleotide (indicated in red). (B) Venn diagram representing the distribution overlap of the top 1% of predicted targets (p-value < 0.01, mirDIP webserver) of the miR-200 family members. (C) Venn diagram representing the distribution overlap of experimentally confirmed targets (mirTarbase webserver) of the miR-200 family members.
Cancers 15 02903 g001
Figure 2. Flowchart of the literature search and identification of targets discussed in this study.
Figure 2. Flowchart of the literature search and identification of targets discussed in this study.
Cancers 15 02903 g002
Figure 3. Schematic comparison of different experimental approaches to verify miRNA–mRNA target direct binding.
Figure 3. Schematic comparison of different experimental approaches to verify miRNA–mRNA target direct binding.
Cancers 15 02903 g003
Figure 4. hsa-miR-429 is an important modular of EMT. hsa-miR-429 creates a negative feedback loop with ZEB1/ZEB2 transcription factors to reversibly direct cells to EMT. Furthermore, other hsa-miR-429 targets as well as transcriptional and posttranscriptional regulators of this miRNA expression modulate the extent of EMT signaling (see text for more details).
Figure 4. hsa-miR-429 is an important modular of EMT. hsa-miR-429 creates a negative feedback loop with ZEB1/ZEB2 transcription factors to reversibly direct cells to EMT. Furthermore, other hsa-miR-429 targets as well as transcriptional and posttranscriptional regulators of this miRNA expression modulate the extent of EMT signaling (see text for more details).
Cancers 15 02903 g004
Figure 5. hsa-miR-429 is an important mediator of the adaptive response to hypoxia and angiogenesis. (A) hsa-miR-429 creates a negative feedback loop with HIF-1α transcription factor to tune the extent of HIF-1-mediated reprograming of gene expression in response to hypoxia. Furthermore, other hsa-miR-429 targets, as well as transcriptional and posttranscriptional regulators of this miRNA, tune the extent of the adaptive response to hypoxia, including angiogenesis (see text for more details). (B) hsa-miR-429 contributes to the switch from HIF-1 to HIF-2 and HIF-3. During acute hypoxia, the negative feedback loop between HIF-1 and miR-429 controls HIF-1 levels, prevents HIF-3 signaling and reduces HIF1A levels. This promotes the transition from HIF-1 to HIF-2. Furthermore, reduced levels of hsa-miR-429 during chronic hypoxia support HIF-3 accumulation (see text for more detail).
Figure 5. hsa-miR-429 is an important mediator of the adaptive response to hypoxia and angiogenesis. (A) hsa-miR-429 creates a negative feedback loop with HIF-1α transcription factor to tune the extent of HIF-1-mediated reprograming of gene expression in response to hypoxia. Furthermore, other hsa-miR-429 targets, as well as transcriptional and posttranscriptional regulators of this miRNA, tune the extent of the adaptive response to hypoxia, including angiogenesis (see text for more details). (B) hsa-miR-429 contributes to the switch from HIF-1 to HIF-2 and HIF-3. During acute hypoxia, the negative feedback loop between HIF-1 and miR-429 controls HIF-1 levels, prevents HIF-3 signaling and reduces HIF1A levels. This promotes the transition from HIF-1 to HIF-2. Furthermore, reduced levels of hsa-miR-429 during chronic hypoxia support HIF-3 accumulation (see text for more detail).
Cancers 15 02903 g005
Table 1. Experimentally verified hsa-miR-429 targets. (+) and (−) indicate the lack or presence of experimental approaches towards miRNA direct binding validation; (*) indicates an miRNA sponge. If Digital Object Identifier (DOI) was not available, the Pub Med ID (PMID) was used.
Table 1. Experimentally verified hsa-miR-429 targets. (+) and (−) indicate the lack or presence of experimental approaches towards miRNA direct binding validation; (*) indicates an miRNA sponge. If Digital Object Identifier (DOI) was not available, the Pub Med ID (PMID) was used.
DOITargetsSponge (*)Transcriptional RegulatorMimicInhibitorLuciferase ReporterTarget ProtectorRIPqPCRWBAnimal ModelmiR-200 Family VerificationReference
ttAV10.1038/ncb1722ZEB1
ZEB2
ZEB1+++++++[91]
10.1007/s12013-014-9885-8ZEB1+++++++[92]
10.1080/21655979.2021.1923238ZEB1++++++[93]
10.1186/s12860-022-00420-xZEB1
MAPKAPK5-AS1 *
+++++++[94]
Q31016/j.biopha.2016.03.035ZEB1
CRKL
++++++[95]
10.1074/jbc.M114.598383ZEB1
ZEB2
ASCL2+++++[96]
10.1016/j.biocel.2019.05.021ZEB1
XIST *
++++++[97]
10.1155/2021/7974012ZEB1
LINC01303 *
++++++++[98]
10.1016/j.lfs.2020.118480ZEB1
MALAT1 *
+++++++[99]
10.1097/CAD.0000000000001295LRP4
RNF185-AS1 *
++++++++[100]
10.1096/fj.14-267054HIF1AHIF-1++++++[101]
10.1007/s10529-018-2604-6HIF1A+++++[102]
10.1038/srep22775HIF3AHIF-1++++++[103]
10.3892/ol.2020.11766HOXA9++++++[104]
10.1016/j.abb.2017.11.011RAB23++++++[105]
10.3389/fnmol.2018.00035TJP1
OCLN
+++++[106]
10.1016/j.bbrc.2017.06.181RohE++++++[107]
10.3892/mco.2019.1940AKT1++++++[108]
10.3892/or.2015.4039PAK6+++++[109]
10.1016/j.urolonc.2015.03.016BMI1
E2F3
+++++[110]
10.3892/ijmm.2016.2558NOTCH1+++++[111]
10.1093/carcin/bgt089RAB18HBx++++++[112]
10.3389/fonc.2020.01067CD274+++++[113]
10.1089/cbr.2020.3705FRS2
SNHG6 *
++++++++[114]
10.1186/s12943-018-0889-7TRIM33
circPTK2 *
++++++++[115]
10.1007/s10528-022-10285-6CELF2
circLIFR *
++++++++[116]
10.1002/jcp.27772MALAT1 *+++++++[117]
PMID: 34786067SYNJ1
LINC01234 *
++++++++[118]
10.1155/2022/1447207 JAK1
MSC-AS1 *
+++++++[119]
10.3892/mmr.2021.12323FOXK1
circPVT1 *
++++++[120]
10.2147/OTT.S277284SCAMP1+++++++[121]
10.1016/j.lfs.2020.117323TRIB2
circ_0084043 *
++++++++[122]
10.1080/21655979.2021.1953822EZH2 circRNA_0082835 *+++++++[123]
10.1186/s11658-020-0202-9 IKKB++++++[124]
10.3892/mmr.2021.12220IKKB+++++[125]
10.1016/j.omtn.2021.01.026 PPARAEZH2++++++[126]
10.7150/jca.21024XIST *+++++++[127]
10.1016/j.bbrc.2014.05.084PTEN
RASSF8
TIMP2
+++++[128]
10.1016/j.canlet.2015.04.023PTEN+++++++[129]
10.1016/j.biopha.2020.110215CRKL+++++[130]
10.1038/s41598-018-20258-8CRKL+++++[131]
10.1136/gutjnl-2013-305715 RBBP4++++++[132]
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

Bartoszewska, S.; Sławski, J.; Collawn, J.F.; Bartoszewski, R. HIF-1-Induced hsa-miR-429: Understanding Its Direct Targets as the Key to Developing Cancer Diagnostics and Therapies. Cancers 2023, 15, 2903. https://doi.org/10.3390/cancers15112903

AMA Style

Bartoszewska S, Sławski J, Collawn JF, Bartoszewski R. HIF-1-Induced hsa-miR-429: Understanding Its Direct Targets as the Key to Developing Cancer Diagnostics and Therapies. Cancers. 2023; 15(11):2903. https://doi.org/10.3390/cancers15112903

Chicago/Turabian Style

Bartoszewska, Sylwia, Jakub Sławski, James F. Collawn, and Rafal Bartoszewski. 2023. "HIF-1-Induced hsa-miR-429: Understanding Its Direct Targets as the Key to Developing Cancer Diagnostics and Therapies" Cancers 15, no. 11: 2903. https://doi.org/10.3390/cancers15112903

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