Next Article in Journal
Joint Tumor Bud–MMP/TIMP Count at the Invasive Front Improves the Prognostic Evaluation of Invasive Breast Carcinoma
Next Article in Special Issue
MicroRNAs as Predictive Biomarkers of Resistance to Targeted Therapies in Gastrointestinal Tumors
Previous Article in Journal
Importance of the PD-1/PD-L1 Axis for Malignant Transformation and Risk Assessment of Oral Leukoplakia
Previous Article in Special Issue
Profiling 25 Bone Marrow microRNAs in Acute Leukemias and Secondary Nonleukemic Hematopoietic Conditions
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

LAT1 and ASCT2 Related microRNAs as Potential New Therapeutic Agents against Colorectal Cancer Progression

by
Francisca Dias
1,†,
Cristina Almeida
1,2,†,
Ana Luísa Teixeira
1,*,†,
Mariana Morais
1,3 and
Rui Medeiros
1,2,3,4,5
1
Molecular Oncology and Viral Pathology Group, IPO-Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Research Center—LAB2, E Bdg 1st Floor, Rua Dr António Bernardino de Almeida, 4200-072 Porto, Portugal
2
Research Department of the Portuguese League against Cancer Regional Nucleus of the North (LPCC-NRN), Estrada da Circunvalação 6657, 4200-177 Porto, Portugal
3
Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal
4
Faculty of Medicine, University of Porto (FMUP), Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
5
Biomedical Research Center (CEBIMED), Faculty of Health Sciences of Fernando Pessoa University (UFP), Praça 9 de Abril 349, 4249-004 Porto, Portugal
*
Author to whom correspondence should be addressed.
Authors contributed equally to this work.
Biomedicines 2021, 9(2), 195; https://doi.org/10.3390/biomedicines9020195
Submission received: 12 January 2021 / Revised: 3 February 2021 / Accepted: 12 February 2021 / Published: 16 February 2021
(This article belongs to the Special Issue MicroRNA in Solid Tumor and Hematological Diseases)

Abstract

:
The development and progression of colorectal cancer (CRC) have been associated with genetic and epigenetic alterations and more recently with changes in cell metabolism. Amino acid transporters are key players in tumor development, and it is described that tumor cells upregulate some AA transporters in order to support the increased amino acid (AA) intake to sustain the tumor additional needs for tumor growth and proliferation through the activation of several signaling pathways. LAT1 and ASCT2 are two AA transporters involved in the regulation of the mTOR pathway that has been reported as upregulated in CRC. Some attempts have been made in order to develop therapeutic approaches to target these AA transporters, however none have reached the clinical setting so far. MiRNA-based therapies have been gaining increasing attention from pharmaceutical companies and now several miRNA-based drugs are currently in clinical trials with promising results. In this review we combine a bioinformatic approach with a literature review in order to identify a miRNA profile with the potential to target both LAT1 and ASCT2 with potential to be used as a therapeutic approach against CRC.

1. Introduction

Colorectal cancer (CRC) is one of the most common cancers worldwide, with 1,849,518 new cases in 2018, being the third most common cancer [1]. Currently, CRC accounts for approximately 10% of all diagnosed cancers and it is the world’s second most deadly cancer [2]. CRC is the second most common neoplasia diagnosed in women, and the third in men, being the incidence and mortality approximately 25% lower in woman [2]. CRC development can be modulated by several factors, being the high alcohol consumption, overweigh, physical inactivity, tobacco smoking, diabetes mellitus, age, personal or family history of CRC well established risk factors [3,4]. Although the mortality rates have declined due to the improvement in diagnosis and treatment, CRC still represents one of the most lethal cancer types [3]. Furthermore, metastasis is also found in, approximately, 15–25% of CRC cases at the diagnosis, and increase to 50% during the course of the disease [2,5]. The advances in the pathophysiological and molecular CRC knowledge allowed the increase of the treatment options, but these new therapeutic approaches were proven to be more effective in patients with non-metastatic disease [2]. Thus, it is imperative to clarify the mechanisms involved in disease progression, aggressiveness and metastasis formation in order to improve the patients’ follow up and to identify new therapeutic approaches.
Recently, the literature showed that amino acid (AA) transporters, such as solute carrier transports (SLCs), are important players in tumor development, since tumor cells have an increased demand for AA to sustain their excessive proliferation rate [6]. In fact, it is described that tumor cells upregulate some AA transporters in order to support the increased AA demand and intake [7]. Moreover, SLCs are able to regulate the PI3K/Akt/mTORC1 signaling pathway, which is central in the regulation of CRC proliferation and aggressiveness and is also involved in metabolic reprograming [8,9]. In fact, there are some SLC dysfunctions associated with CRC, such as L-type amino acid transporter 1 (LAT1) and alanine-serine-cysteine transporter 2 (ASCT2) upregulation, that may have an impact on disease aggressiveness [9,10,11]. Given the growing evidence and interest in the impact of cancer metabolism in disease aggressiveness, it is imperative to further understand the regulatory mechanisms responsible for of LAT1 and ASCT2 modulation in CRC and study the potential of their inhibition as a therapeutic approach. Since these two AA transporters are frequently overexpressed in CRC cells, they have potential as drug targets because their inhibition or blockade could lead to cell cycle arrest and apoptosis [10,12,13,14].

1.1. Amino Acid Transporters Deregulation in CRC: The Impact of LAT1 and ASCT2

It has been nearly a century since the discovery that normal and tumor cells differ in energy metabolism, with tumor cells presenting a higher need of nutrients, being the AA bioavailability crucial to support cell proliferation and growth [15]. Amino acids can be classified into three groups: (1) essential AA (EAA), if the organism is not able to synthesize them and needs to acquire them from the diet; (2) non-essential AA, if they are synthesized in sufficient quantities by the organism or (3) conditional AA, if are usually nonessential, except in times of illness, trauma or stress were they become conditionally essential [16,17].
In addition to their need in protein synthesis, several amino acids have other roles in supporting cancer development. One example is glutamine, the most abundant AA that participates in energy production, redox homeostasis, macromolecular synthesis and cell signaling [18]. In fact, the commitment of glutamine in the these cell processes makes this AA conditionally essential in conditions characterized by a high proliferation rate, such as cancer, in which endogenous glutamine synthesis is not sufficient to satisfy the cell need [17].
Since AAs are hydrophilic, they need selective transport proteins in order to cross the plasma membrane of the cells. There are approximately two-dozen amino acid transporters in humans, and cancer cells must regulate one or more of these transporters to satisfy their nutrient demand [6]. LAT1 (SLC7A5) is a transmembrane transporter involved in the import of large and neutral AA such as leucine and phenylalanine, in exchange for intracellular AA, such as glutamine [10,13,19]. According to various studies, LAT1 is highly upregulated in multiple human cancers, including gastrointestinal cancers [10,19,20,21]. In fact, Hayase and coworkers found a higher expression of LAT1 in 72.4% of CRC cases when compared to colonic adenoma cases, concluding that LAT1 could be a marker for malignant lesions [10]. Furthermore, Zhang and colleagues also found an association of higher LAT1 expression levels to poorer outcomes and shorter survival in several types of cancer, including CRC [14]. The higher LAT1 expression in cancer cells shows the importance of this AA transporter in the maintenance of AA nutrition in cancer cells [6]. Studies conducted by Elorza and coworkers show that the upregulation of LAT1 is involved in the increase of mTORC1 activity through HIF2α activation, showing a relationship between the hypoxic microenvironment, HIF2α and LAT1 [22]. Furthermore, LAT1 mediates leucine uptake with high affinity, which is a key AA activator of the mTOR signaling pathway [23]. However, for mTOR activation, the functional LAT1 is coupled to ASCT2, another AA transporter involved in glutamine uptake [16].
The ASCT2 (SLC1A5) is expressed in most human tissues including the large intestine and CRC tumor cells, and is essentially responsible for the influx of glutamine inside the cells, inducing asparagine, serine and threonine efflux [24,25,26]. According to Liu and colleagues, ASCT2 expression levels can modulate the migration capacity of CRC cells, being the overexpression of this AA transportersassociated with a poorer patients’ prognosis [1,27]. In fact, ASCT2 is upregulated in several cancers, including triple-negative breast cancer, CRC, lung cancer, melanoma, neuroblastoma, glioblastoma and prostate cancer [12]. Some studies in glioblastomas and neuroblastoma support the involvement of the activation of c-Myc, n-Myc oncogenes in the inducing of ASCT2 expression [28,29].
Metabolic reprogramming is a well-known hallmark of cancer that has been gaining increasing attention in the last few years due to its importance in cancer cells viability and growth [30]. Cancer associated metabolic reprogramming influences intracellular and extracellular availability of metabolites that will result in alterations in gene expression, cellular differentiation and also in the tumor microenvironment [31]. Glutamine is considered to be a crucial nutrient for cancer proliferation due to its ability to donate its nitrogen and carbon to several growth-promoting pathways [32]. In 2012, Mootha and colleagues reported that tumor cells have a high necessity of glutamine uptake compared to other AA and, consequently, a glutamine starvation can interfere with tumor metabolism inhibiting tumor proliferation and progression [32]. More recently, Varshavi and colleagues, described a molecular association between CRC that present oncogenic KRAS mutation and glutamine metabolism, since these cells exhibit special metabolic phenotypes, including differences in glycolysis, glutamine utilization and AA metabolism [33]. Furthermore, glutamine is described as a signaling factor in the uptake of AA for the activation of mTORC1 [34]. Thus, the upregulation of AA transporters have an important role in the support of the high-level protein synthesis for continuous cancer growth and proliferation [10,35]. The mTOR pathway is well described as deregulated in CRC, and the availability of AA functions as a regulator of this pathway, since a high AA microenvironmental bioavailability induces mTOR activity and consequent biological processes, such as protein translation [36]. Some studies report a relationship between LAT1 and ASCT2, with a two-step mechanism of these AAT being able to regulate mTOR pathway [37,38,39]. Firstly, ASCT2 regulates the intracellular concentration of glutamine, and in turn LAT1 uses this intracellular glutamine as an efflux substrate, in order to regulate the uptake of extracellular leucine, which will lead to an activation of mTOR signaling and consequent induction of cell growth and proliferation [40,41] (Figure 1). Furthermore, according to Rajasinghe and coworkers, the inhibition of glutamine uptake in proliferating cells, through the inhibition of glutamine transporters LAT1 and ASCT2, results in the inhibition of cell proliferation and induces apoptosis, through the downregulation of the mTOR pathway [38]. Thus, the inhibition of LAT1 and ASCT2 expression levels could represent a promising therapeutic approach for CRC since it would reduce the AA intake, consequently causing mTOR pathway inhibition and compromising cancer cell proliferation.
The use of pharmacologic approaches against LAT1 and ASCT2 in cancers with overexpression of these two AA transporters seems be a promising strategy. In fact, over the last few years there was investment in the development of drugs against LAT1 and ASCT2 [26,38,42,43]. The design of drugs against these two AA transporters usually follows an approach based on substrate analogues, which act as competitive inhibitors [26]. In the case of ASCT2 there are also been developed monoclonal antibodies against its cell surface domains [44]. The pharmacological inhibitors against LAT1 and ASCT2 reported in CRC are listed on Table 1.
More recently, in a phase I study, Okano and coworkers observed that the JPH203 treatment was well tolerated by patients with CRC and biliary tract cancer (BTC). In fact, disease control was observed in two of the six CRC patients and in three of the five BTC patients [13]. Furthermore, a study from Toda and colleagues using two KRAS-mutated cells lines demonstrated a significant association between ASCT2 expression and KRAS mutation and, when the authors used siRNAs to silence KRAS, they observed a significant reduction of ASCT2 [48]. In addition to that, the authors also used specific inhibitors of Raf/MEK/ERK, and PI3K/Akt/mTOR pathways, and observed that both inhibitors presented the ability to reduce ASCT2 expression [48]. Moreover, studies using xenograft models demonstrated that the inhibition of ASCT2 expression is able to reduce the uptake of glutamine and inhibit tumor cell proliferation [49]. However, it is imperative to keep in mind that the block of AA transporters could be associated with the upregulation of compensatory and redundant pathways, being crucial an accurate overview of all network involved in the process [50]. In addition to that, there are some limitations in the use of pharmacological inhibitors due to the low affinity for the transporter and low selective capacity observed to cancer cells. Thus, these data highlight the need for a deeper understanding of other therapeutic approaches for the selective inhibition of LAT1 and ASCT2 in CRC.

1.2. Applicability of microRNAs as Therapeutic Agents

Over the years, advances in genomic technologies have led to an identification of a variety of epigenetic alterations believed to be strongly involved in cancer initiation and progression [51]. In fact, several studies revealed that the altered metabolic pathways in cancer are tightly regulated by microRNAs (miRNAs) [52,53,54,55,56,57,58]. MiRNAs are a family of short non-coding RNAs with a length of approximately 19–25 nucleotides that post-transcriptionally regulate gene expression, with an important role in several biological pathways, including cell proliferation and differentiation [59,60]. MiRNAs can regulate the expression of more than 50% of protein-coding genes by binding to their target mRNA transcript and causing its degradation or translation repression [61]. Furthermore, the downstream targets of several miRNAs are directly or indirectly connected to metabolic alterations [52].
Regarding their applicability in the clinical setting, a growing number of evidence suggests a significant utility of miRNAs as biomarkers for pathogenic conditions, modulators of drug resistance and as therapeutic agents for medical intervention in almost all human health-related conditions [62,63,64,65]. The pleiotropic nature of miRNAs makes them particularly attractive, both as drugs or drug targets, for diseases with a multifactorial origin and no current effective treatments [66,67]. In addition to that, circulating miRNAs present several advantages compared to other circulating nucleic acids, such as: protection from RNAse degradation, high stability in circulation through the body, and resistance to adverse conditions such as temperature or pH alterations [68,69]. Regarding miRNA therapeutics applicability, there are reports demonstrating clinical utility of miRNA mimics and miRNA repressors and miRNAs loaded in something. In fact, there are two major types of miRNA-based therapies: miRNA suppression therapy, when the goal is the target mRNA upregulation and miRNA replacement therapy, when the goal is the target mRNA downregulation.
Overall, the current evidence suggests a viable future for miRNA drugs in diseases with no current effective treatments, such as CRC. Hence, the scope of this review is to gather and systematize the information available regarding the impact of LAT1 and ASCT2 related miRNAs in CRC development and establish a profile with potential application to be used as a therapeutic agent through in silico analysis combined with a literature review (Figure 2).

2. Materials and Methods

2.1. MiRNA Selection and Literature Review

In order to select miRNAs that target both LAT1 and ASCT2 we used miRTarBase (version 8.0), the largest known online database of validated miRNA:mRNA interactions [70]. According to miRTarBase there are 267 miRNAs that target LAT1 and 173 that target ASCT2 mRNAs. Since one miRNA has multiple targets and the same mRNA can be regulated by several miRNAs, we went to see if there were miRNAs that targeted both LAT1 and ASCT2. From the 440 miRNAs retrieved by miRTarBase, we observed that 33 targeted both LAT1 and ASCT2 (Figure 3).
After retrieving the miRNAs that target both LAT1 and ASCT2 from miRTarBase, a literature search in PubMed and Google Academic was conducted using the search terms “colorectal cancer” plus one of the following 33 microRNAs: “miR-122-5p”, “miR-1224-3p”, “miR-1260a”, “hsa-miR-1260b”, “hsa-1273g-3p”, hsa-miR-1273h-5p”, “hsa-miR-149-3p”, “hsa-miR-15b-5p”, “miR-16-5p”, “miR-193b-3p”, “miR-30b-3p”, “miR-3199”, “miR-3689a-3p”, “miR-3689b-3p”, “miR-3689c”, “miR-383-3p”, “miR-4690-5p”, “miR-4728-5p”, “miR-504-3p”, “miR-5693”, “miR-5698”, “miR-619-5p”, “miR-6499”, “miR-6778-3p”, “miR-6799-5p”, “miR-6780a-5p”, “miR-6785”, “miR-6799”, “miR-6821”, “miR-6883-5p”, “miR-6890-3p”, “miR-7106-5p” or “miR-7977”. The articles were selected by relevance of their findings, namely, a significant association between these miRNAs and colorectal cancer. Literature analysis includes scientific papers published in the last 6 years (between 2014 and 2020). The obtained scientific papers were manually curated in order to determine associations between the miRNAs and CRC, giving a total of 28 selected papers. The exclusion criteria for the collected papers were as follows: (1) no significant association between the miRNAs and CRC; (2) association of the miRNAs with a benign tumor and (3) individual papers that were already included in meta-analysis. For each study, information was extracted concerning the following characteristics: the name of the miRNA, type of sample where the miRNA was studied, miRNA expression levels (upregulated and downregulated) and their effect on CRC (e.g., prognosis, therapy response or pathways regulation).

2.2. In Silico Analysis

The Search Tool for the Retrieval of Interacting Genes (STRING) database is an online tool that is used to develop protein–protein interaction (PPI) networks [71]. We used the STRINGapp of the Cytoscape software (v3.7.X) to construct and visualize the protein interaction network of the selected target genes. Those with a combined score of >0.4 were selected as significant. The functional enrichment analysis of Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and Reactome pathways was made with the STRING enrichment analysis tool, with a false discovery rate (FDR) of p < 0.01. The enrichment results were filtered, and redundant terms were removed according to the Jaccard index.

3. Results

3.1. miRNAs that Target Both LAT1 and ASCT2 and their Impact on CRC

From the 33 candidate miRNAs, only 16 have already been described in CRC (Table 2). However, in terms of the miRNA: mRNA target interaction with LAT1 and ASCT2, none of the miRNAs have been yet validated for CRC.
Through the analysis of Table 1 we can observe that some of the miRNAs present opposite results regarding their expression levels, which may be related with the type of biological sample from which their expression levels are analyzed. Regarding their effects on CRC, the deregulation of miR-122-5p, miR-1273g-3p, miR-16-5p, miR-3199, miR-383-3p, miR-619-5p and miR-6883-5p was associated with the upregulation of important players of oncogenic pathways, such as TRIM29, CDC25A, PI3K/Akt, mTOR, VEGFA, MALAT1, SMAD4, STMN1, APRIL and CDK4, with an impact on cell proliferation, invasion and migration. In addition to that, miR-1260b, miR-149-3p and miR-15b-5p were reported as associated with resistance to 5′-FU treatment through the upregulation of PDCD4, PDK2 and XIAP, respectively. Moreover, only three miRNAs were associated with clinical endpoints. Higher plasmatic levels of hsa-miR-122-5p were associated with worse prognosis in metastatic patients and shorter RFS and OS in non-metastatic patients, while lower levels of CRC tissue hsa-miR-193b-3p and hsa-miR-619-5p were associated with shorter OS. Moreover, lower levels of CRC tissue hsa-miR-619-5p were also associated with shorter DFS, lymphovascular invasion and perineural invasion.

3.2. Functional Annotation and Pathway Enrichment Analysis

Since the downregulation of a miRNA usually leads to the upregulation of its mRNA targets, we focused on the 11 miRNAs that have been reported as downregulated in CRC cells and tissues and therefore could be implicated in the upregulation of LAT1 and ASCT2 (miR-122-5p, miR-1260b, miR-149-3p, miR-15b-5p, miR-16-5p, miR-193b-3p, miR-3199, miR-383-3p, miR-619-5p, miR-6821-5p and miR-6883-5p) and did an in silico analysis to obtain a deeper knowledge of their impact on CRC. We used miRTarBase v8.0 to retrieve the mRNA targets of the selected miRNAs that were validated with strong evidence methods in order to do the functional annotation and enrichment analysis. From the 11 miRNAs studied, only miR-15b-5p, miR-16-5p, miR-122-5p, miR-149-3p, miR-1260b, miR-193b-3p and miR-383-3p presented mRNA targets validated with strong evidence methods (Western blot, qRT-PCR or luciferase assay), which are listed on Table 3.
In order to explore the biological impact of these miRNA profiles in CRC, we analyzed their 186 validated targets with the STRINGapp Protein Query from Cytoscape software. A total of 168 of the 186 coding genes were filtered into a protein–protein interaction (PPI) network with 168 nodes and 1284 edges that presented a significant enrichment (p = 1 × 10−16). We also applied a Markov clustering (MCL), which resulted in the clustering of the proteins into 11 clusters according to their STRING interaction score (Figure 4).
The functional enrichment analysis was made using an FDR threshold of p < 0.01, and the redundant terms were eliminated using a redundancy cutoff of 0.5, which resulted in a total of 892 enriched terms among the KEGG, Reactome and GO categories (Supplementary Tables S1–S5). The top 20 enriched terms for each category are represented on Figure 5. Among the functionally enriched terms in the KEGG and Reactome pathways we could find PI3K/Akt, MAPK, HIF-1, mTOR, VEGF and EGFR inhibitor resistant pathways, all of which are well established as involved in CRC development. Regarding the GO terms, if we focus on the molecular processes, we can observe that the two most enriched terms are the regulation of cell proliferation and the cellular response to organic substances, which may be related with the increase intake of nutrients as a consequence of the downregulation of this miRNA profile and consequent upregulation of AA transporters, such as LAT1 and ASCT2.

4. Discussion

CRC remains one of the most diagnosed cancers in the world, with a high metastatic potential and not enough therapeutic options. The previous underestimated metabolic alterations are now gaining more attention from the scientific community, and it is now known that metabolic cross-communication between tumor cells, immune cells, stromal cells and the gut microbiota are able to induce CRC proliferation, invasion and metastasis [98]. Among the metabolic alterations with potential to be targeted in order to develop new therapeutic approaches, the upregulation of AA transporters LAT1 and ASCT2 seems promising due to their impact in the regulation of the mTOR pathway. In addition to that, the liver is recognized as the most common metastatic site of CRC and cumulative evidences show that LAT1 and ASCT2 are overexpressed in hepatocellular carcinoma (HCC) and that these cells present a 10–20 fold increase in glutamine uptake, compared to normal hepatocytes [27,38,99,100,101]. Therefore, the definition of a new therapeutic approach involving the inhibition of these two AA transporters could be a promising strategy to control CRC proliferation and aggressiveness.
Recently, it has been suggested that modulation of miRNAs in cancer cells could be a potential tool for the improvement of cancer patients’ therapies. In fact, by suppressing oncogenic miRNAs or substituting deficient tumor suppressive miRNAs, we are able to control cancer cell growth and progression. The world’s first miRNA therapeutic, a short locked nucleic acid (LNA) antagonist for miR-122 named Miravirsen (produced by Roche/Santaris) was developed for the treatment of hepatitis C virus (HCV) infection [102]. Along with Miravirsen, all of the miRNA-based drugs are currently in clinical trials and none have yet reached the pharmaceutical breakthrough. However, acquisition of miRNA-based companies by famous pharmaceutical companies is sending a positive feedback on their potential [103]. Currently, there are several strategies used for miRNA-based therapies, which could include miRNA inhibition therapies that target oncomiRNAs, replacement therapies for tumor-suppressor miRNAs or miRNA-based delivery systems [104]. One example, of their applicability was the study performed by Callegari and coworkers that showed that the in vivo delivery of an anti-miR-221 caused a significant decrease in the size and number of tumor nodules, being established the promotor role of miR-221 in liver carcinogenesis [105]. On the other hand, Oshima and coworkers reported an effective delivery of miR-655-3p to CRC liver metastasis using nanoscale coordination polymers. The polymers used prolonged the miRNA distribution and miRNA-655-3p suppressed tumor growth when codelivered with oxaplatin, suggesting a synergistic effect of both therapeutic approaches [106].
Regarding the delivery mechanisms, miRNAs are delivered through the use of vectors that can be divided into two categories: viral vectors and nonviral vectors. The viral vectors used for miRNA delivery are mainly adenovirus vectors, adeno-associated virus vectors, retroviral vectors and lentivitus vectors. On the other hand, nonviral vectors include inorganic material-based delivery systems, lipid-based nanocarriers, polymeric vectors/dendrimer-based vectors, cell-derived membrane vesicles and 3D-Scaffold-based delivery systems [107]. The use of these delivery mechanisms improves targeting ability while protecting the miRNAs or miRNAs inhibitors from degradation. In fact, it was already demonstrated that, for cancer treatment, intratumoral injections of miRNA drugs directly into the tumor site are able to enhance target efficacy, specificity and minimize the side effects and there are also several ongoing clinical trials [62,108,109].
In the present review we combined a bioinformatic approach with a literature review to define a miRNA profile (miR-15b-5p, miR-16-5p, miR-122-5p, miR-1260b, miR-149-3p, miR-193b-3p and miR-383-3p) that has the potential to target both LAT1 and ASCT2 in CRC. The in silico approaches are very useful since they allow the simultaneous analysis of the interactions of hundreds of genes and, therefore, the creation of an integrative network that allows a deeper understanding of the biological processes regulated by them. Our in silico analysis result in a list of miRNAs that target both LAT1 and ASCT2 and the literature review allowed us to focus on the miRNAs that have already been studied in CRC and have been reported as downregulated. In addition to that, functional enrichment analysis showed that among the enriched terms derived from the miRNA profile targets, we can find PI3K/Akt, MAPK, HIF-1, mTOR, VEGF and EGFR inhibitor resistant pathways, all of which are well established as involved in CRC development.
The MAPK and PI3K/Akt signaling pathways are involved in cell proliferation and survival, and their deregulation confers proliferative advantages on cancer cells. In fact, KRAS, BRAF and PI3K mutations are frequent in CRC. Moreover, the increase of the PI3K/Akt pathway activation in CRC is also associated with the loss of the tumor suppressor PTEN, which is significantly associated with a worse prognosis [1]. In addition to that, according to Slatery and colleagues, approximately 41% of the genes of MAPK signaling are dysregulated in CRC [110]. These two signaling cascades can activate directly and indirectly the Ser/Thr protein kinase mTOR, respectively, being the mTOR involved in the regulation of cell proliferation and survival [111]. Similarly to other solid tumors, CRC is also characterized by a hypoxic microenvironment [112]. In fact, the cancer cells have the ability of adaptation to hypoxia through the regulation of the PI3K/AKT/mTOR pathway and by the transcription factors HIF-1α and HIF-2α, whose protein expression and transcriptional activity are also regulated by mTOR [113]. Furthermore, tumor hypoxia can also enhance cancer cells survival and proliferation through the upregulation of VEGF and its receptor VEGFR. VEGF promotes CRC growth through the stimulation of angiogenesis and its downstream signaling pathways are well characterized in cancer, with VEGF/VEGFR activation leading to the activation of MAPK/ERK, PI3K/Akt, PLC/PKC and other signaling pathways [114,115] .
Taking this information into consideration, we can conclude that the miRNA profile proposed in the present study plays an important role on CRC development and aggressiveness. However, despite promising, the results are still preliminary and require validation in CRC study models in order to assess the miRNA profile interaction with LAT1 and ASCT2 mRNAs, especially in terms of its inhibitory power. During the past few years, there has been a significant development of in vitro and in vivo preclinical research models, such as 3D cell culture of spheroids and organoids derived from several human tissues, which is helping in the translation of miRNAs into clinical practice [66,116,117]. In a recent study, Kawai and colleagues determined the culture conditions necessary to establish 3D cell culture models that mimic colon cancer heterogeneity [118]. In another study, Zoetemelk and colleagues established a robust, low-cost and reproducible short-term 3D colorectal cancer spheroids model to be used as a platform for screening the effect of combination therapies in CRC [119]. These enhanced research models are very useful for the study of miRNAs dynamics and for the development of the delivery systems for miRNA-based therapeutics [120,121]. Therefore, the next step should be focused on the delivery of the miRNA profile to CRC 3D culture models in order to see if it is sufficient to reverse the increased AA uptake caused by the increase of LAT1 and ASCT2 and inhibit cell proliferation.

Supplementary Materials

The following are available online at https://www.mdpi.com/2227-9059/9/2/195/s1, Table S1: GO Cellular Component enrichment results for terms with FDR p < 0.01, Table S2: GO Molecular Function enrichment results for terms with FDR p < 0.01, Table S3: GO Biological Process enrichment results for terms with FDR p < 0.01, Table S4: KEGG pathway enrichment results for terms with FDR p < 0.01, Table S5: Reactome pathways enrichment results for terms with FDR p < 0.01.

Author Contributions

Conceptualization, F.D.; methodology, F.D., C.A. and A.L.T.; software, F.D.; investigation, F.D., A.L.T.; data curation, F.D.; writing—original draft preparation, F.D., C.A. and A.L.T.; writing—review and editing, M.M., F.D., A.L.T.; supervision, R.M.; project administration, R.M.; funding acquisition, R.M., A.L.T. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the European Regional Development Fund (ERDF), grant number NORTE-01-0247-FEDER-033399.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. Mármol, I.; Sánchez-De-Diego, C.; Dieste, A.P.; Cerrada, E.; Yoldi, M.J.R. Colorectal carcinoma: A general overview and future perspectives in colorectal cancer. Int. J. Mol. Sci. 2017, 18, 197. [Google Scholar] [CrossRef] [Green Version]
  2. Dekker, E.; Tanis, P.J.; Vleugels, J.L.A.; Kasi, P.M.; Wallace, M.B. Colorectal cancer. Lancet 2019, 394, 1467–1480. [Google Scholar] [CrossRef]
  3. Ahmed, M. Colon cancer: A clinician’s perspective in 2019. Gastroenterol. Res. 2020, 13, 1–10. [Google Scholar] [CrossRef] [PubMed]
  4. Karimi, N.; Feizi, M.A.H.; Safaralizadeh, R.; Hashemzadeh, S.; Baradaran, B.; Shokouhi, B.; Teimourian, S. Serum overexpression of miR-301a and miR-23a in patients with colorectal cancer. J. Chin. Med Assoc. 2019, 82, 215–220. [Google Scholar] [CrossRef]
  5. Oh, B.Y.; Hong, H.K.; Lee, W.Y.; Cho, Y.B. Animal models of colorectal cancer with liver metastasis. Cancer Lett. 2017, 387, 114–120. [Google Scholar] [CrossRef]
  6. Bhutia, Y.D.; Babu, E.; Ramachandran, S.; Ganapathy, V. Amino acid transporters in cancer and their relevance to “glutamine addiction”: Novel targets for the design of a new class of anticancer drugs. Cancer Res. 2015, 75, 1782–1788. [Google Scholar] [CrossRef] [Green Version]
  7. Bröer, S. Amino acid transporters as targets for cancer therapy: Why, where, when, and how. Int. J. Mol. Sci. 2020, 21, 6156. [Google Scholar] [CrossRef]
  8. Bai, X.; Moraes, T.F.; Reithmeier, R.A.F. Structural biology of solute carrier (SLC) membrane transport proteins. Mol. Membr. Biol. 2017, 34, 1–32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Xie, J.; Zhu, X.Y.; Liu, L.M.; Meng, Z.Q. Solute carrier transporters: Potential targets for digestive system neoplasms. Cancer Manag. Res. 2018, 10, 153–166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Hayase, S.; Kumamoto, K.; Saito, K.; Kofunato, Y.; Sato, Y.; Okayama, H.; Miyamoto, K.; Ohki, S.; Takenoshita, S. L-type amino acid transporter 1 expression is upregulated and associated with cellular proliferation in colorectal cancer. Oncol. Lett. 2017, 14, 7410–7416. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Nyquist, M.D.; Prasad, B.; Mostaghel, E.A. Harnessing solute carrier transporters for precision oncology. Molecules 2017, 22, 539. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Bhutia, Y.D.; Ganapathy, V. Glutamine transporters in mammalian cells and their functions in physiology and cancer. Biochim. Biophys. Acta 1863, 2531–2539. [Google Scholar] [CrossRef] [PubMed]
  13. Scalise, M.; Pochini, L.; Console, L.; Losso, M.A.; Indiveri, C. The human SLC1A5 (ASCT2) amino acid transporter: From function to structure and role in cell biology. Front. Cell Dev. Biol. 2018, 6, 96. [Google Scholar] [CrossRef]
  14. Zhang, C.; Xu, J.; Xue, S.; Ye, J. Prognostic Value of L-type amino acid transporter 1 (LAT1) in various cancers: A meta-analysis. Mol. Diagn. Ther. 2020, 24, 523–536. [Google Scholar] [CrossRef]
  15. Ganapathy, V.; Thangaraju, M.; Prasad, P.D. Nutrient transporters in cancer: Relevance to Warburg hypothesis and beyond. Pharmacol. Ther. 2009, 121, 29–40. [Google Scholar] [CrossRef] [PubMed]
  16. Choi, B.-H.; Coloff, J.L. The diverse functions of non-essential amino acids in cancer. Cancers 2019, 11, 675. [Google Scholar] [CrossRef] [Green Version]
  17. Scalise, M.; Console, L.; Rovella, F.; Galluccio, M.; Pochini, L.; Indiveri, C. Membrane transporters for amino acids as players of cancer metabolic rewiring. Cells 2020, 9, 2028. [Google Scholar] [CrossRef] [PubMed]
  18. Hensley, C.T.; Wasti, A.T.; DeBerardinis, R.J. Glutamine and cancer: Cell biology, physiology, and clinical opportunities. J. Clin. Investig. 2013, 123, 3678–3684. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Singh, N.; Ecker, G.F. Insights into the structure, function, and ligand discovery of the large neutral amino acid transporter 1, LAT1. Int. J. Mol. Sci. 2018, 19, 1278. [Google Scholar] [CrossRef] [Green Version]
  20. Häfliger, P.; Charles, R.-P. The L-type amino acid transporter LAT1—An emerging target in Cancer. Int. J. Mol. Sci. 2019, 20, 2428. [Google Scholar] [CrossRef] [Green Version]
  21. Ogawa, H.; Kaira, K.; Motegi, Y.; Yokobori, T.; Takada, T.; Katoh, R.; Osone, K.; Takahashi, R.; Katayama, C.; Oyama, T.; et al. Role of amino acid transporter expression as a prognostic marker in patients with surgically resected colorectal cancer. Anticancer Res. 2019, 39, 2535–2543. [Google Scholar] [CrossRef] [PubMed]
  22. Elorza, A.; Soro-Arnáiz, I.; Meléndez-Rodríguez, F.; Rodríguez-Vaello, V.; Marsboom, G.; De Cárcer, G.; Acosta-Iborra, B.; Albacete-Albacete, L.; Ordóñez, A.; Serrano-Oviedo, L.; et al. HIF2α acts as an mTORC1 activator through the amino acid carrier SLC7A5. Mol. Cell 2012, 48, 681–691. [Google Scholar] [CrossRef] [Green Version]
  23. Taylor, P.M. Role of amino acid transporters in amino acid sensing. Am. J. Clin. Nutr. 2014, 99, 223S–230S. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Liu, Y.; Zhao, T.; Li, Z.; Wang, L.; Yuan, S.; Sun, L. The role of ASCT2 in cancer: A review. Eur. J. Pharmacol. 2018, 837, 81–87. [Google Scholar] [CrossRef]
  25. Cormerais, Y.; Massard, P.A.; Vucetic, M.; Giuliano, S.; Tambutté, E.; Durivault, J.; Vial, V.; Endou, H.; Wempe, M.F.; Parks, S.K.; et al. The glutamine transporter ASCT2 (SLC1A5) promotes tumor growth independently of the amino acid transporter LAT1 (SLC7A5). J. Biol. Chem. 2018, 293, 2877–2887. [Google Scholar] [CrossRef] [Green Version]
  26. Jiang, H.; Zhang, N.; Tang, T.; Feng, F.; Sun, H.; Qu, W. Target the human alanine/serine/cysteine transporter 2(ASCT2): Achievement and future for novel cancer therapy. Pharmacol. Res. 2020, 158, 104844. [Google Scholar] [CrossRef]
  27. Scalise, M.; Pochini, L.; Galluccio, M.; Console, L.; Indiveri, C. Glutamine transport and mitochondrial metabolism in cancer cell growth. Front. Oncol. 2017, 7, 306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Wise, D.R.; DeBerardinis, R.J.; Mancuso, A.; Sayed, N.; Zhang, X.-Y.; Pfeiffer, H.K.; Nissim, I.; Daikhin, E.; Yudkoff, M.; McMahon, S.B.; et al. Myc regulates a transcriptional program that stimulates mitochondrial glutaminolysis and leads to glutamine addiction. Proc. Natl. Acad. Sci. USA 2008, 105, 18782–18787. [Google Scholar] [CrossRef] [Green Version]
  29. Ren, P.; Yue, M.; Xiao, D.; Xiu, R.; Gan, L.; Liu, H.; Qing, G. ATF4 and N-Myc coordinate glutamine metabolism in MYCN-amplified neuroblastoma cells through ASCT2 activation. J. Pathol. 2015, 235, 90–100. [Google Scholar] [CrossRef]
  30. Hanahan, D.; Weinberg, R.A. Hallmarks of Cancer: The Next Generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
  31. Pavlova, N.N.; Thompson, C.B. The emerging hallmarks of cancer metabolism. Cell Metab. 2016, 23, 27–47. [Google Scholar] [CrossRef] [Green Version]
  32. Liberti, M.V.; Locasale, J.W. The Warburg Effect: How does it benefit cancer cells? Trends Biochem. Sci. 2016, 41, 211–218. [Google Scholar] [CrossRef] [Green Version]
  33. Wu, H.; Wei, M.; Jiang, X.; Tan, J.; Xu, W.; Fan, X.; Zhang, R.; Ding, C.; Zhao, F.; Shao, X.; et al. lncRNA PVT1 promotes tumorigenesis of colorectal cancer by stabilizing miR-16-5p and interacting with the VEGFA/VEGFR1/AKT axis. Mol. Ther. Nucleic Acids 2020, 20, 438–450. [Google Scholar] [CrossRef]
  34. Ward, P.S.; Thompson, C.B. Metabolic reprogramming: A cancer hallmark even Warburg did not anticipate. Cancer Cell 2012, 21, 297–308. [Google Scholar] [CrossRef] [Green Version]
  35. Yanagida, O.; Kanai, Y.; Chairoungdua, A.; Kim, D.K.; Segawa, H.; Nii, T.; Cha, S.H.; Matsuo, H.; Fukushima, J.-I.; Fukasawa, Y.; et al. Human L-type amino acid transporter 1 (LAT1): Characterization of function and expression in tumor cell lines. Biochim. Biophys. Acta 2001, 1514, 291–302. [Google Scholar] [CrossRef] [Green Version]
  36. Bar-Peled, L.; Sabatini, D.M. Regulation of mTORC1 by amino acids. Trends Cell Biol. 2014, 24, 400–406. [Google Scholar] [CrossRef] [PubMed]
  37. Toda, K.; Nishikawa, G.; Iwamoto, M.; Itatani, Y.; Takahashi, R.; Sakai, Y.; Kawada, K. Clinical role of ASCT2 (SLC1A5) in KRAS-mutated colorectal cancer. Int. J. Mol. Sci. 2017, 18, 1632. [Google Scholar] [CrossRef] [Green Version]
  38. Rajasinghe, L.D.; Hutchings, M.; Gupta, S.V. Delta-tocotrienol modulates glutamine dependence by inhibiting ASCT2 and LAT1 transporters in non-small cell lung cancer (NSCLC) cells: A metabolomic approach. Metabolites 2019, 9, 50. [Google Scholar] [CrossRef] [Green Version]
  39. Alves, M.J.F.; Uno, M.; da Silva, R.; Shinjo, S.M.; Marie, S.K.N. The expression of the aminoacid transporters ASCT2 (SLC1A5) and LAT1 (SLC7A5) in astrocytomas. Med. Express 2016, 3. [Google Scholar] [CrossRef] [Green Version]
  40. Yazawa, T.; Shimizu, K.; Kaira, K.; Nagashima, T.; Ohtaki, Y.; Atsumi, J.; Obayashi, K.; Nagamori, S.; Kanai, Y.; Oyama, T.; et al. Clinical significance of coexpression of L-type amino acid transporter 1 (LAT1) and ASC amino acid transporter 2 (ASCT2) in lung adenocarcinoma. Am. J. Transl. Res. 2015, 7, 1126–1139. [Google Scholar]
  41. Nicklin, P.; Bergman, P.; Zhang, B.; Triantafellow, E.; Wang, H.; Nyfeler, B.; Yang, H.; Hild, M.; Kung, C.; Wilson, C.; et al. Bidirectional transport of amino acids regulates mTOR and autophagy. Cell 2009, 136, 521–534. [Google Scholar] [CrossRef] [Green Version]
  42. Foster, A.C.; Rangel-Diaz, N.; Yang, J.-Y.; Penjwini, M.; Viswanath, V.; Li, Y.-X.; Staubli, U. Phenylglycine analogs are inhibitors of the neutral amino acid transporters ASCT1 and ASCT2 and enhance NMDA receptor-mediated LTP in rat visual cortex slices. Neuropharmacology 2017, 126, 70–83. [Google Scholar] [CrossRef] [PubMed]
  43. Hayashi, K.; Anzai, N. Novel therapeutic approaches targeting L-type amino acid transporters for cancer treatment. World J. Gastrointest. Oncol. 2017, 9, 21–29. [Google Scholar] [CrossRef] [PubMed]
  44. Suzuki, M.; Toki, H.; Furuya, A.; Ando, H. Establishment of monoclonal antibodies against cell surface domains of ASCT2/SLC1A5 and their inhibition of glutamine-dependent tumor cell growth. Biochem. Biophys. Res. Commun. 2017, 482, 651–657. [Google Scholar] [CrossRef]
  45. Muto, Y.; Furihata, T.; Kaneko, M.; Higuchi, K.; Okunushi, K.; Morio, H.; Reien, Y.; Uesato, M.; Matsubara, H.; Anzai, N. Different response profiles of gastrointestinal cancer cells to an L-type amino acid transporter inhibitor, JPH203. Anticancer. Res. 2019, 39, 159–165. [Google Scholar] [CrossRef]
  46. Okunushi, K.; Furihata, T.; Morio, H.; Muto, Y.; Higuchi, K.; Kaneko, M.; Otsuka, Y.; Ohno, Y.; Watanabe, Y.; Reien, Y.; et al. JPH203, a newly developed anti-cancer drug, shows a preincubation inhibitory effect on L-type amino acid transporter 1 function. J. Pharmacol. Sci. 2020, 144, 16–22. [Google Scholar] [CrossRef]
  47. Schulte, M.L.; Fu, A.; Zhao, P.; Allie, F.; Geng, L.; Smith, S.T.; Kondo, J.; Coffey, R.J.; Johnson, M.O.; Rathmell, J.C.; et al. Pharmacological blockade of ASCT2-dependent glutamine transport leads to antitumor efficacy in preclinical models. Nat. Med. 2018, 24, 194–202. [Google Scholar] [CrossRef] [PubMed]
  48. Namikawa, M.; Kakizaki, S.; Kaira, K.; Tojima, H.; Yamazaki, Y.; Horiguchi, N.; Satoru, K.; Oriuchi, N.; Tominaga, H.; Sunose, Y.; et al. Expression of amino acid transporters (LAT1, ASCT2 and xCT) as clinical significance in hepatocellular carcinoma. Hepatol. Res. 2014, 45, 1014–1022. [Google Scholar] [CrossRef]
  49. Song, W.; Li, D.; Tao, L.; Luo, Q.; Chen, L. Solute carrier transporters: The metabolic gatekeepers of immune cells. Acta Pharm. Sin. B 2020, 10, 61–78. [Google Scholar] [CrossRef]
  50. Bröer, S. Amino acid transporters as disease modifiers and drug targets. SLAS Discov. 2018, 23, 303–320. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Danese, E.; Montagnana, M. Epigenetics of colorectal cancer: Emerging circulating diagnostic and prognostic biomarkers. Ann. Transl. Med. 2017, 5, 279. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Hatziapostolou, M.; Polytarchou, C.; Iliopoulos, D. miRNAs link metabolic reprogramming to oncogenesis. Trends Endocrinol. Metab. 2013, 24, 361–373. [Google Scholar] [CrossRef] [Green Version]
  53. Shu, S.; Yang, Y.; Allen, C.L.; Maguire, O.; Minderman, H.; Sen, A.; Ciesielski, M.J.; Collins, K.A.; Bush, P.J.; Singh, P.; et al. Metabolic reprogramming of stromal fibroblasts by melanoma exosome microRNA favours a premetastatic microenvironment. Sci. Rep. 2018, 8, 12905. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Sun, Y.; Oravecz-Wilson, K.; Bridges, S.; McEachin, R.; Wu, J.; Kim, S.H.; Taylor, A.; Zajac, C.; Fujiwara, H.; Peltier, D.C.; et al. miR-142 controls metabolic reprogramming that regulates dendritic cell activation. J. Clin. Investig. 2019, 129, 2029–2042. [Google Scholar] [CrossRef]
  55. Yan, W.; Wu, X.; Zhou, W.; Fong, M.Y.; Cao, M.; Liu, J.; Liu, X.; Chen, C.H.; Fadare, O.; Pizzo, D.P.; et al. Cancer-cell-secreted exosomal miR-105 promotes tumour growth through the MYC-dependent metabolic reprogramming of stromal cells. Nat. Cell Biol. 2018, 20, 597–609. [Google Scholar] [CrossRef] [Green Version]
  56. Nogueira, I.; Dias, F.; Morais, M.; Teixeira, A.L.; Medeiros, R. Everolimus resistance in clear cell renal cell carcinoma: miRNA-101 and HIF-2α as molecular triggers? Futur. Oncol. 2019, 15, 2361–2370. [Google Scholar] [CrossRef] [PubMed]
  57. Nogueira, I.; Dias, F.; Teixeira, A.L.; Medeiros, R. miRNAs as potential regulators of mTOR pathway in renal cell carcinoma. Pharmacogenomics 2018, 19, 249–261. [Google Scholar] [CrossRef]
  58. Morais, M.; Dias, F.; Teixeira, A.L.; Medeiros, R. MicroRNAs and altered metabolism of clear cell renal cell carcinoma: Potential role as aerobic glycolysis biomarkers. Biochim. Biophys. Acta. Gen. Subj. 2017, 1861, 2175–2185. [Google Scholar] [CrossRef] [PubMed]
  59. Acunzo, M.; Romano, G.; Wernicke, D.; Croce, C.M. MicroRNA and cancer—A brief overview. Adv. Biol. Regul. 2015, 57, 1–9. [Google Scholar] [CrossRef] [PubMed]
  60. Khan, S.; Ayub, H.; Khan, T.; Wahid, F. MicroRNA biogenesis, gene silencing mechanisms and role in breast, ovarian and prostate cancer. Biochimie 2019, 167, 12–24. [Google Scholar] [CrossRef]
  61. Tovar-Camargo, O.A.; Toden, S.; Goel, A. Exosomal microRNA biomarkers: Emerging frontiers in colorectal and other human cancers. Expert Rev. Mol. Diagn. 2016, 16, 553–567. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Hanna, J.; Hossain, G.S.; Kocerha, J. The potential for MicroRNA therapeutics and clinical research. Front. Genet. 2019, 10, 478. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Dias, F.; Teixeira, A.L.; Nogueira, I.; Morais, M.; Maia, J.; Bodo, C.; Ferreira, M.; Silva, A.; Vilhena, M.; Lobo, J.; et al. Extracellular vesicles enriched in hsa-miR-301a-3p and hsa-miR-1293 dynamics in clear cell renal cell carcinoma patients: Potential biomarkers of metastatic disease. Cancers 2020, 12, 1450. [Google Scholar] [CrossRef] [PubMed]
  64. Adem, B.F.; Bastos, N.R.A.; Dias, F.; Teixeira, A.L.; Medeiros, R. miRNAs: Mediators of ErbB family targeted therapy resistance. Pharmacogenomics 2016, 17, 1175–1187. [Google Scholar] [CrossRef]
  65. Mollaei, H.; Safaralizadeh, R.; Rostami, Z. MicroRNA replacement therapy in cancer. J. Cell Physiol. 2019, 234, 12369–12384. [Google Scholar] [CrossRef]
  66. Chakraborty, C.; Sharma, A.R.; Sharma, G.; Doss, C.G.P.; Lee, S.-S. Therapeutic miRNA and siRNA: Moving from Bench to Clinic as Next Generation Medicine. Mol. Ther. Nucleic Acids 2017, 8, 132–143. [Google Scholar] [CrossRef] [Green Version]
  67. Van Rooij, E.; Purcell, A.L.; Levin, A.A. Developing MicroRNA therapeutics. Circ. Res. 2012, 110, 496–507. [Google Scholar] [CrossRef] [PubMed]
  68. Glinge, C.; Clauss, S.; Boddum, K.; Jabbari, R.; Jabbari, J.; Risgaard, B.; Tomsits, P.; Hildebrand, B.; Kääb, S.; Wakili, R.; et al. Stability of circulating blood-based MicroRNAs—Pre-analytic methodological considerations. PLoS ONE 2017, 12, e0167969. [Google Scholar] [CrossRef]
  69. Sohel, M.H. Extracellular/circulating MicroRNAs: Release mechanisms, functions and challenges. Achiev. Life Sci. 2016, 10, 175–186. [Google Scholar] [CrossRef] [Green Version]
  70. Huang, H.Y.; Lin, Y.C.; Li, J.; Huang, K.Y.; Shrestha, S.; Hong, H.C.; Tang, Y.; Chen, Y.G.; Jin, C.N.; Yu, Y.; et al. miRTarBase 2020: Updates to the experimentally validated microRNA-target interaction database. Nucleic Acids Res. 2020, 48, D148–D154. [Google Scholar] [CrossRef] [Green Version]
  71. Szklarczyk, D.; Gable, A.L.; Lyon, D.; Junge, A.; Wyder, S.; Huerta-Cepas, J.; Simonovic, M.; Doncheva, N.T.; Morris, J.H.; Bork, P.; et al. STRING v11: Protein–protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 2019, 47, D607–D613. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Yin, W.; Xu, J.; Li, C.; Dai, X.; Wu, T.; Wen, J. Circular RNA circ_0007142 facilitates colorectal cancer progression by modulating CDC25A expression via miR-122-5p. OncoTargets Ther. 2020, 13, 3689–3701. [Google Scholar] [CrossRef]
  73. Asadi, M.; Talesh, S.T.; Gjerstorff, M.F.; Shanehbandi, D.; Baradaran, B.; Hashemzadeh, S.; Zafari, V. Identification of miRNAs correlating with stage and progression of colorectal cancer. Color. Cancer 2019, 8, CRC06. [Google Scholar] [CrossRef] [Green Version]
  74. Liu, J.; Li, H.; Sun, L.; Shen, S.; Zhou, Q.; Yuan, Y.; Xing, C. Epigenetic alternations of MicroRNAs and DNA methylation contribute to liver metastasis of colorectal cancer. Dig. Dis. Sci. 2019, 64, 1523–1534. [Google Scholar] [CrossRef] [PubMed]
  75. Qu, A.; Yang, Y.; Zhang, X.; Wang, W.; Liu, Y.; Zheng, G.; Du, L.; Wang, C. Development of a preoperative prediction nomogram for lymph node metastasis in colorectal cancer based on a novel serum miRNA signature and CT scans. EBioMedicine 2018, 37, 125–133. [Google Scholar] [CrossRef] [Green Version]
  76. Maierthaler, M.; Benner, A.; Hoffmeister, M.; Surowy, H.; Jansen, L.; Knebel, P.; Chang-Claude, J.; Brenner, H.; Burwinkel, B. Plasma miR-122 and miR-200 family are prognostic markers in colorectal cancer. Int. J. Cancer 2017, 140, 176–187. [Google Scholar] [CrossRef] [PubMed]
  77. Lin, M.; Duan, B.; Hu, J.; Yu, H.; Sheng, H.; Gao, H.; Huang, J. Decreased expression of miR-193a-3p is associated with poor prognosis in colorectal cancer. Oncol. Lett. 2017, 14, 1061–1067. [Google Scholar] [CrossRef] [Green Version]
  78. Wang, Y.-N.; Chen, Z.-H.; Chen, W.-C. Novel circulating microRNAs expression profile in colon cancer: A pilot study. Eur. J. Med. Res. 2017, 22, 1–11. [Google Scholar] [CrossRef] [Green Version]
  79. Zhao, J.; Cao, J.; Zhou, L.; Du, Y.; Zhang, X.; Yang, B.; Gao, Y.; Wang, Y.; Ma, N.; Yang, W. MiR-1260b inhibitor enhances the chemosensitivity of colorectal cancer cells to fluorouracil by targeting PDCD4/IGF. Oncol. Lett. 2018, 16, 5131–5139. [Google Scholar] [CrossRef] [Green Version]
  80. Zhang, J.; Luo, X.; Li, H.; Deng, L.; Wang, Y. Genome-wide uncovering of STAT3-mediated miRNA expression profiles in colorectal cancer cell lines. BioMed Res. Int. 2014, 2014, 1–11. [Google Scholar] [CrossRef]
  81. Slattery, M.L.; Herrick, J.S.; Pellatt, D.F.; Stevens, J.R.; Mullany, L.E.; Wolff, E.; Hoffman, M.D.; Samowitz, W.S.; Wolff, R.K. MicroRNA profiles in colorectal carcinomas, adenomas and normal colonic mucosa: Variations in miRNA expression and disease progression. Carcinogenesis 2016, 37, 245–261. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Zhang, Y.; Li, M.; Ding, Y.; Fan, Z.; Zhang, J.; Zhang, H.; Jiang, B.; Zhu, Y. Serum MicroRNA profile in patients with colon adenomas or cancer. BMC Med Genom. 2017, 10, 23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Cha, D.J.; Franklin, J.L.; Dou, Y.; Liu, Q.; Higginbotham, J.N.; Beckler, M.D.; Weaver, A.M.; Vickers, K.C.; Prasad, N.; Levy, S.; et al. KRAS-dependent sorting of miRNA to exosomes. eLife 2015, 4, e07197. [Google Scholar] [CrossRef]
  84. Li, M.; Qian, X.; Zhu, M.; Li, A.; Fang, M.; Zhu, Y.; Zhang, J. miR-1273g-3p promotes proliferation, migration and invasion of LoVo cells via cannabinoid receptor 1 through activation of ERBB4/PIK3R3/mTOR/S6K2 signaling pathway. Mol. Med. Rep. 2018, 17, 4619–4626. [Google Scholar] [CrossRef] [Green Version]
  85. Du, B.; Wu, D.; Yang, X.; Wang, T.; Shi, X.; Lv, Y.; Zhou, Z.; Liu, Q.; Zhang, W. The expression and significance of microRNA in different stages of colorectal cancer. Medicine 2018, 97, e9635. [Google Scholar] [CrossRef] [PubMed]
  86. Liang, Y.; Hou, L.; Li, L.; Li, L.; Zhu, L.; Wang, Y.; Huang, X.; Hou, Y.; Zhu, D.; Zou, H.; et al. Dichloroacetate restores colorectal cancer chemosensitivity through the p53/miR-149-3p/PDK2-mediated glucose metabolic pathway. Oncogene 2020, 39, 469–485. [Google Scholar] [CrossRef] [PubMed]
  87. Zhao, C.; Zhao, Q.; Zhang, C.; Wang, G.; Yao, Y.; Huang, X.; Zhan, F.; Zhu, Y.; Shi, J.; Chen, J.; et al. miR-15b-5p resensitizes colon cancer cells to 5-fluorouracil by promoting apoptosis via the NF-κB/XIAP axis. Sci. Rep. 2017, 7, 4194. [Google Scholar] [CrossRef] [PubMed]
  88. Gasparello, J.; Gambari, L.; Papi, C.; Rozzi, A.; Manicardi, A.; Corradini, R.; Gambari, R.; Finotti, A. High levels of apoptosis are induced in the human colon cancer HT-29 cell line by co-administration of sulforaphane and a peptide nucleic acid targeting miR-15b-5p. Nucleic Acid Ther. 2020, 30, 164–174. [Google Scholar] [CrossRef] [PubMed]
  89. Guo, F.; Luo, Y.; Mu, Y.-F.; Qin, S.-L.; Qi, Y.; Qiu, Y.-E.; Zhong, M. miR-193b directly targets STMN1 and inhibits the malignant phenotype in colorectal cancer. Am. J. Cancer Res. 2016, 6, 2463–2475. [Google Scholar]
  90. Kara, M.; Yumrutas, O.; Ozcan, O.; Celik, O.I.; Bozgeyik, E.; Bozgeyik, I.; Tasdemir, S. Differential expressions of cancer-associated genes and their regulatory miRNAs in colorectal carcinoma. Gene 2015, 567, 81–86. [Google Scholar] [CrossRef]
  91. Yan, W.; Liu, Z.; Yang, W.; Wu, G. miRNA expression profiles in Smad4-positive and Smad4-negative SW620 human colon cancer cells detected by next-generation small RNA sequencing. Cancer Manag. Res. 2018, 10, 5479–5490. [Google Scholar] [CrossRef] [Green Version]
  92. Cui, Y.; Chen, L.; Yao, H.; Zhang, J.; Ding, K. Upregulation of microRNA-383 inhibits the proliferation, migration and invasion of colon cancer cells. Oncol. Lett. 2017, 15, 1184–1190. [Google Scholar] [CrossRef] [Green Version]
  93. Ghanbari, R.; Mosakhani, N.; Sarhadi, V.K.; Armengol, G.; Nouraee, N.; Mohammadkhani, A.; Khorrami, S.; Arefian, E.; Paryan, M.; Malekzadeh, R.; et al. Simultaneous underexpression of let-7a-5p and let-7f-5p microRNAs in plasma and stool samples from early stage colorectal carcinoma. Biomark. Cancer 2015, 7, BIC.S25252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Mullany, L.E.; Herrick, J.S.; Wolff, R.K.; Stevens, J.R.; Slattery, M.L. Association of cigarette smoking and microRNA expression in rectal cancer: Insight into tumor phenotype. Cancer Epidemiol. 2016, 45, 98–107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Qiu, G.; Zhang, X.-B.; Zhang, S.-Q.; Liu, P.-L.; Wu, W.; Zhang, J.-Y.; Dai, S.-R. Dysregulation of MALAT1 and miR-619-5p as a prognostic indicator in advanced colorectal carcinoma. Oncol. Lett. 2016, 12, 5036–5042. [Google Scholar] [CrossRef] [Green Version]
  96. Zhou, J.-M.; Hu, S.-Q.; Jiang, H.; Chen, Y.-L.; Feng, J.-H.; Chen, Z.-Q.; Wen, K.-M. OCT4B1 Promoted EMT and regulated the self-renewal of CSCs in CRC: Effects associated with the balance of miR-8064/PLK1. Mol. Ther. Oncolytics 2019, 15, 7–20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Lulla, A.R.; Slifker, M.J.; Zhou, Y.; Lev, A.; Einarson, M.B.; Dicker, D.T.; El-Deiry, W.S. miR-6883 family miRNAs target CDK4/6 to induce G(1) phase cell-cycle arrest in colon cancer cells. Cancer Res. 2017, 77, 6902–6913. [Google Scholar] [CrossRef] [Green Version]
  98. Brown, R.E.; Short, S.P.; Williams, C.S. Colorectal cancer and metabolism. Curr. Color. Cancer Rep. 2018, 14, 226–241. [Google Scholar] [CrossRef]
  99. Verrey, F. System L: Heteromeric exchangers of large, neutral amino acids involved in directional transport. Pflügers Arch. 2003, 445, 529–533. [Google Scholar] [CrossRef]
  100. Fuchs, B.C.; Bode, B.P. Amino acid transporters ASCT2 and LAT1 in cancer: Partners in crime? Semin. Cancer Biol. 2005, 15, 254–266. [Google Scholar] [CrossRef]
  101. Hur, K.; Toiyama, Y.; Okugawa, Y.; Ide, S.; Imaoka, H.; Boland, C.R.; Goel, A. Circulating microRNA-203 predicts prognosis and metastasis in human colorectal cancer. Gut 2017, 66, 654–665. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Gebert, L.F.R.; Rebhan, M.A.E.; Crivelli, S.E.M.; Denzler, R.; Stoffel, M.; Hall, J. Miravirsen (SPC3649) can inhibit the biogenesis of miR-122. Nucleic Acids Res. 2014, 42, 609–621. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Bonneau, E.; Neveu, B.; Kostantin, E.; Tsongalis, G.J.; De Guire, V. How close are miRNAs from clinical practice? A perspective on the diagnostic and therapeutic market. EJIFCC 2019, 30, 114–127. [Google Scholar] [PubMed]
  104. Sharad, S.; Kapur, S. MiRNA-based therapeutics in oncology, realities and challenges (2018). In Antisense Therapy; Balacescu, O., Visan, S., Baldasici, O., Balacescu, L., Vlad, C., Achimas-Cadariu, P., Eds.; IntechOpen: London, UK.
  105. Callegari, E.; Elamin, B.K.; Giannone, F.; Milazzo, M.; Altavilla, G.; Fornari, F.; Giacomelli, L.; D’Abundo, L.; Ferracin, M.; Bassi, C.; et al. Liver tumorigenicity promoted by microRNA-221 in a mouse transgenic model. Hepatology 2012, 56, 1025–1033. [Google Scholar] [CrossRef] [PubMed]
  106. Oshima, G.; Guo, N.; He, C.; Stack, M.E.; Poon, C.; Uppal, A.; Wightman, S.C.; Parekh, A.; Skowron, K.B.; Posner, M.C.; et al. In vivo delivery and therapeutic effects of a MicroRNA on colorectal liver metastases. Mol. Ther. 2017, 25, 1588–1595. [Google Scholar] [CrossRef] [Green Version]
  107. Fu, Y.; Chen, J.; Huang, Z. Recent progress in microRNA-based delivery systems for the treatment of human disease. ExRNA 2019, 1, 24. [Google Scholar] [CrossRef] [Green Version]
  108. Chen, Y.; Gao, D.-Y.; Huang, L. In vivo delivery of miRNAs for cancer therapy: Challenges and strategies. Adv. Drug Deliv. Rev. 2015, 81, 128–141. [Google Scholar] [CrossRef] [Green Version]
  109. Mercatelli, N.; Coppola, V.; Bonci, D.; Miele, F.; Costantini, A.; Guadagnoli, M.; Bonanno, E.; Muto, G.; Frajese, G.V.; De Maria, R.; et al. The inhibition of the highly expressed Mir-221 and Mir-222 impairs the growth of prostate carcinoma xenografts in mice. PLoS ONE 2008, 3, e4029. [Google Scholar] [CrossRef] [PubMed]
  110. Slattery, M.L.; Mullany, L.E.; Sakoda, L.C.; Wolff, R.K.; Samowitz, W.S.; Herrick, J.S. The MAPK-signaling pathway in colorectal cancer: Dysregulated genes and their association with MicroRNAs. Cancer Inform. 2018, 17, 1176935118766522. [Google Scholar] [CrossRef] [Green Version]
  111. Reita, D.; Bour, C.; BenBrika, R.; Groh, A.; Pencreach, E.; Guérin, E.; Guenot, D. Synergistic anti-tumor effect of mTOR inhibitors with irinotecan on colon cancer cells. Cancers 2019, 11, 1581. [Google Scholar] [CrossRef] [Green Version]
  112. Mi, Y.; Mu, L.; Huang, K.; Hu, Y.; Yan, C.; Zhao, H.; Ma, C.; Li, X.; Tao, D.; Qin, J. Hypoxic colorectal cancer cells promote metastasis of normoxic cancer cells depending on IL-8/p65 signaling pathway. Cell Death Dis. 2020, 11, 1–11. [Google Scholar] [CrossRef] [PubMed]
  113. Agani, F.; Jiang, B.H. Oxygen-independent regulation of HIF-1: Novel involvement of PI3K/AKT/mTOR pathway in cancer. Curr. Cancer Drug Targets 2013, 13, 245–251. [Google Scholar] [CrossRef]
  114. Ntellas, P.; Mavroeidis, L.; Gkoura, S.; Gazouli, I.; Amylidi, A.-L.; Papadaki, A.; Zarkavelis, G.; Mauri, D.; Karpathiou, G.; Kolettas, E.; et al. Old player-new tricks: Non angiogenic effects of the VEGF/VEGFR pathway in cancer. Cancers 2020, 12, 3145. [Google Scholar] [CrossRef]
  115. Ahluwalia, A.; Jones, M.K.; Szabo, S.; Tarnawski, A.S. Aberrant, ectopic expression of VEGF and VEGF receptors 1 and 2 in malignant colonic epithelial cells. Implications for these cells growth via an autocrine mechanism. Biochem. Biophys. Res. Commun. 2013, 437, 515–520. [Google Scholar] [CrossRef] [Green Version]
  116. Boo, L.; Ho, W.Y.; Ali, N.M.; Yeap, S.K.; Ky, H.; Chan, K.G.; Yin, W.F.; Satharasinghe, D.A.; Liew, W.C.; Tan, S.W.; et al. MiRNA transcriptome profiling of spheroid-enriched cells with cancer stem cell properties in human breast MCF-7 cell line. Int. J. Biol. Sci. 2016, 12, 427–445. [Google Scholar] [CrossRef]
  117. Park, H.; Hwang, S.; Jeong, J.-Y.; Jung, S.G.; Choi, M.C.; Joo, W.D.; Song, S.H.; Lee, C.; An, H.J. Integrative analysis of transcription factors and microRNAs in ovarian cancer cell spheroids. J. Ovarian Res. 2020, 13, 1–10. [Google Scholar] [CrossRef] [PubMed]
  118. Kawai, S.; Yamazaki, M.; Shibuya, K.; Yamazaki, M.; Fujii, E.; Nakano, K.; Suzuki, M. Three-dimensional culture models mimic colon cancer heterogeneity induced by different microenvironments. Sci. Rep. 2020, 10, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Zoetemelk, M.; Rausch, M.; Colin, D.J.; Dormond, O.; Nowak-Sliwinska, P. Short-term 3D culture systems of various complexity for treatment optimization of colorectal carcinoma. Sci. Rep. 2019, 9, 7103. [Google Scholar] [CrossRef] [PubMed]
  120. Zhang, L.; Yang, X.; Lv, Y.; Xin, X.; Qin, C.; Han, X.; Yang, L.; He, W.; Yin, L. Cytosolic co-delivery of miRNA-34a and docetaxel with core-shell nanocarriers via caveolae-mediated pathway for the treatment of metastatic breast cancer. Sci. Rep. 2017, 7, 46186. [Google Scholar] [CrossRef] [Green Version]
  121. Morgan, R.G.; Chambers, A.C.; Legge, D.N.; Coles, S.J.; Greenhough, A.; Williams, A.C. Optimized delivery of siRNA into 3D tumor spheroid cultures in situ. Sci. Rep. 2018, 8, 1–10. [Google Scholar] [CrossRef]
Figure 1. Representation of the interplay between ASCT2, LAT1 and mTOR pathway in colorectal cancer (CRC). This image was created using BioRender.
Figure 1. Representation of the interplay between ASCT2, LAT1 and mTOR pathway in colorectal cancer (CRC). This image was created using BioRender.
Biomedicines 09 00195 g001
Figure 2. Schematic overview of the applicability of this review in the future development of miRNA-based therapies against LAT1 and ASCT2 in CRC. This image was created using BioRender.
Figure 2. Schematic overview of the applicability of this review in the future development of miRNA-based therapies against LAT1 and ASCT2 in CRC. This image was created using BioRender.
Biomedicines 09 00195 g002
Figure 3. Venn diagram of the validated miRNAs that target ASCT2 and LAT1 obtained using Venny 2.1. (http://bioinfogp.cnb.csic.es/tools/venny, accessed on 21 December 2020) and the detailed list of the 33 miRNAs.
Figure 3. Venn diagram of the validated miRNAs that target ASCT2 and LAT1 obtained using Venny 2.1. (http://bioinfogp.cnb.csic.es/tools/venny, accessed on 21 December 2020) and the detailed list of the 33 miRNAs.
Biomedicines 09 00195 g003
Figure 4. Protein–protein interaction (PPI) network. The proteins were clustered using the clusterMaker app from Cytoscape with an inflation value of 2.5 and a cutoff edge of 0.5. All the singletons were removed.
Figure 4. Protein–protein interaction (PPI) network. The proteins were clustered using the clusterMaker app from Cytoscape with an inflation value of 2.5 and a cutoff edge of 0.5. All the singletons were removed.
Biomedicines 09 00195 g004
Figure 5. Kyoto Encyclopedia of Genes and Genomes (KEGG), Reactome and Gene Ontology (GO) analysis of the 186 selected target genes. The functional enrichment analysis was made using the STRINGapp from Cytoscape.
Figure 5. Kyoto Encyclopedia of Genes and Genomes (KEGG), Reactome and Gene Ontology (GO) analysis of the 186 selected target genes. The functional enrichment analysis was made using the STRINGapp from Cytoscape.
Biomedicines 09 00195 g005
Table 1. Pharmacological inhibitors of LAT1 and ASCT2 reported in CRC.
Table 1. Pharmacological inhibitors of LAT1 and ASCT2 reported in CRC.
AA TransporterInhibitorInhibitor TypeReference
LAT1JHP203Tyrosine analog[45,46]
ASCT2MAb KM4008Monoclinal antibodies against cell surface domains[44]
MAb KM4012
MAb KM4018
V-9302Competitive antagonist[47]
Table 2. Selected miRNAs’ impact on CRC.
Table 2. Selected miRNAs’ impact on CRC.
miRNAExpressionSample TypeEffectReference
Hsa-miR-122-5pDownCRC Tissue and cellsIncrease in cell proliferation, migration and invasion through the upregulation of CDC25AYin 2020 [72]
DownCRC TissuesUpregulation of the PI3K/Akt pathway through upregulation of TRIM29Asadi 2019 [73]
UpCRC liver metastatic tissuesNot describedLiu 2019 [74]
UpSerum and HT-29 and SW480 cell linesLymph node metastasis biomarker and cell migration inducerQu 2018 [75]
UpCRC PlasmaWorse prognosis in metastatic patients and shorter RFS and OS in non-metastatic patientsMaiertheler 2017 [76]
Hsa-miR-1224-3pUpCRC TissuesUpregulated in E cadherin positive tissuesLin 2017 [77]
Hsa-miR-1260aDownCRC SerumNot describedWang 2017 [78]
Hsa-miR-1260bUpHCT116 cellsChemoresistance to 5-FU through upregulation of PDCD4Zhao 2018 [79]
DownSW480 cellsDownregulated by STAT3-siRNAZhang 2014 [80]
UpCarcinoma vs adenoma (tissue)Not describedSlattery 2016 [81]
DownCRC SerumNot describedZhang 2017 [82]
UpDKO-1 cellsEnriched in KRAS mutant cellsCha 2015 [83]
Hsa-miR-1273g-3pUpLoVo cellsProliferation, migration and invasion through activation of ERBB4/PIK3R3/mTOR/S6K2 pathwayLi 2018 [84]
Hsa-miR-1273h-5pUpCRC tissuesNot describedDu 2018 [85]
Hsa-miR-149-3pDownHCT-8 and HCT-116 cellsChemoresistance to 5-FU through upregulation of PDK2Liang 2020 [86]
Hsa-miR-15b-5pDownCRC tissues and cell linesChemoresistance to 5-FU through upregulation of XIAPZhao 2017 [87]
UpHT-29 cell lineCell growth and inhibition of the proapoptotic pathwayGasparello 2020 [88]
DownKRAS mutated CRC tissues vs wild type CRC tissuesNot describedMilanesi 2020 (82)
Hsa-miR-16-5pDownCRC tissues and cell linesUpregulation of VEGFAWu 2020 [33]
Hsa-miR-193b-3pDownCRC tissues vs adjacent normal tissuesShorter OS of CRC patients and upregulation of STMN1Guo 2016 [89]
UpCRC tissuesDownregulation of RAD51Kara 2015 [90]
Hsa-miR-3199DownSW620 cell lineUpregulation of SMAD4Yan 2018 [91]
Hsa-miR-383-3pDownCRC tissues and HT-29 and LoVo cell linesUpregulation of APRILCui 2018 [92]
Hsa-miR-4690-5pDownCRC StoolNot describedGhanbari 2015 [93]
UpCRC tissuesUpregulated in CIMP high/MSI CRC tissuesMullany 2016 [94]
Hsa-miR-619-5pDownCRC tissues vs adjacent normal tissuesUpregulation of MALAT1, lymphovascular invasion perineural invasion, shorter DFS and shorter OSQiu 2016 [95]
Hsa-miR-6821-5pDownSW480 CSCs vs SW480 wild-typeNot describedZhou 2019 [96]
UpCRC tissuesNot describedDu 2018 [85]
Hsa-miR-6883-5pDownTCGA dataset and Cell linesUpregulation of CDK4 and CDK6 and cell growth stimulusLulla 2017 [97]
Table 3. Validated targets of miR-15b-5p, miR-16-5p, miR-122-5p, miR-1260b, miR-149-3p, miR-193b-3p and miR-383-3p.
Table 3. Validated targets of miR-15b-5p, miR-16-5p, miR-122-5p, miR-1260b, miR-149-3p, miR-193b-3p and miR-383-3p.
miRNATarget mRNA
miR-16-5pZYX, YAP1, WNT4, WNT3A, WEE1, VEGFA, UNG, UCA1, TPPP3, TP53, SOX6, SOX5, SOCS3, SLC6A4, RPS6KB1, RICTOR, RECK, RAF1, PURA, PTGS2, PRDM4, PPM1D, PIM1, OPRM1, NCSTN, NCOR2, MYB, MTOR, METTL13, MAP7, KRAS, KDR, IL12B, IGF1R, IFNG, HMGA2, HMGA1, HGF, HDGF, GLS2, FGFR1, FGF2, CLDN2, CHUK, CHEK1, CDS2, CDK6, CCNE1, CCND3, CCND2, CCND1, CAPRIN1, CADM1, BRCA1, BMI1, BIRC5, BDNF, BCL2, BACE1, AXIN2, ARL2, ARHGDIA, APP, AKT3, ADORA2A, ACVR2A
miR-15b-5pWEE1, VEGFA, TRIM29, TRIM14, TGFB1, TBR1, SOCS3, SMURF1, SMAD2, RECK, RAB1A, PURA, PPM1D, PEBP4, OIP5, MTSS1, MMP9, KDR, INSR, IFNG, HNF1A, FUT2, FOXO1, EIF4A1, CHEK1, CCNE1, CCND3, CCND1, BCL2, BAX, AXIN2, AKT3, AGO2
miR-122-5pZNF395, XPO6, WNT1, VEGFC, UBAP2, TRIB1, TPD52L2, TBX19, SRF, SPRY2, SOCS1, SLC7A11, SLC7A1, RHOA, RAC1, RAB6B, RAB11FIP1, PTPN1, PRKRA, PRKAB1, PKM, PEG10, PDK4, P4HA1, NUMBL, NT5C3A, NOD2, NFATC2IP, NCAM1, MEF2D, MECP2, MAPK11, LPIN1, IL1A, IGF1R, HMOX1, GYS1, GALNT10, G6PC3, FUT8, FUNDC2, FOXP1, FOXJ3, FAM117B, ENTPD4, EGLN3, EGFR, DUSP2, DSTYK, CYP7A1, CTDNEP1, CREB1, CLIC4, CDK4, CCNG1, BCL2L2, BAX, AXL, ATP1A2, AP3M2, ANXA11, ANK2, ALDOA, AKT3, ADAM17, ADAM10, AACS
miR-1260bSMAD4, SFRP1, DKK2
miR-193b-3pYWHAZ, SMAD3, SHMT2, RAD51, PRAP1, PLAU, NF1, MYB, MCL1, MAX, KRAS, KIT, ETS1, ESR1, CCND1, AKR1C2
miR-383-3pPRPF31
miR-149-3pWNT1, MYBL2, GPC1, FGFR1, E2F1, AKT1
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Dias, F.; Almeida, C.; Teixeira, A.L.; Morais, M.; Medeiros, R. LAT1 and ASCT2 Related microRNAs as Potential New Therapeutic Agents against Colorectal Cancer Progression. Biomedicines 2021, 9, 195. https://doi.org/10.3390/biomedicines9020195

AMA Style

Dias F, Almeida C, Teixeira AL, Morais M, Medeiros R. LAT1 and ASCT2 Related microRNAs as Potential New Therapeutic Agents against Colorectal Cancer Progression. Biomedicines. 2021; 9(2):195. https://doi.org/10.3390/biomedicines9020195

Chicago/Turabian Style

Dias, Francisca, Cristina Almeida, Ana Luísa Teixeira, Mariana Morais, and Rui Medeiros. 2021. "LAT1 and ASCT2 Related microRNAs as Potential New Therapeutic Agents against Colorectal Cancer Progression" Biomedicines 9, no. 2: 195. https://doi.org/10.3390/biomedicines9020195

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