Next Article in Journal
Vitamin D: Can Gender Medicine Have a Role?
Previous Article in Journal
Laboratory Diagnosis of Antiphospholipid Syndrome in Anticoagulated Patients
Previous Article in Special Issue
Interaction of Drug-Sensitive and -Resistant Human Melanoma Cells with HUVEC Cells: A Label-Free Cell-Based Impedance Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Tumor Microenvironment Remodeling in Gastrointestinal Cancer: Role of miRNAs as Biomarkers of Tumor Invasion

1
Department of Experimental Medicine, University of Rome “Sapienza”, Viale Regina Elena 324, 00161 Rome, Italy
2
Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy
3
Departmental Faculty of Medicine and Surgery, Saint Camillus International University of Health and Medical Sciences, via di Sant’Alessandro 8, 00131 Rome, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Biomedicines 2023, 11(6), 1761; https://doi.org/10.3390/biomedicines11061761
Submission received: 10 May 2023 / Revised: 13 June 2023 / Accepted: 15 June 2023 / Published: 19 June 2023
(This article belongs to the Special Issue Recent Advances in Tumor Cell Migration)

Abstract

:
Gastrointestinal (GI) cancers are the most frequent neoplasm, responsible for half of all cancer-related deaths. Metastasis is the leading cause of death from GI cancer; thus, studying the processes that regulate cancer cell migration is of paramount importance for the development of new therapeutic strategies. In this review, we summarize the mechanisms adopted by cancer cells to promote cell migration and the subsequent metastasis formation by highlighting the key role that tumor microenvironment components play in deregulating cellular pathways involved in these processes. We, therefore, provide an overview of the role of different microRNAs in promoting tumor metastasis and their role as potential biomarkers for the prognosis, monitoring, and diagnosis of GI cancer patients. Finally, we relate the possible use of nutraceuticals as a new strategy for targeting numerous microRNAs and different pathways involved in GI tumor invasiveness.

1. Introduction

Gastrointestinal (GI) cancers are complex and heterogeneous diseases caused by the interaction of genetic and environmental factors. These types of tumors can affect several organs, such as the pancreas, liver, gallbladder, bile ducts, and colon. Because of the large cell mass and rapid turnover, GI cancers are among the most frequent neoplasms and are responsible for about half of all cancer-related deaths [1]. In fact, they account for 26% of global cancer incidence and 35% of all cancer-related deaths [2]. These neoplasms have different clinical features but share some similar characteristics; the most common is their poor prognosis meaning that most treatments can often be ineffective. Although much progress has been made in the early detection of colorectal cancer (CRC) through screening, the prognosis of other GI tumors tends to be unfavorable due to patients presenting late-stage diagnoses [3]. Current therapies involve adjuvant and neoadjuvant chemoradiotherapy, targeted therapy, or immunotherapy, which stimulate an antitumor immune response [3,4]. Despite these multidisciplinary approaches, a fair number of patients still develop distant metastasis and show resistance to therapy [5,6]. Indeed, metastasis is the leading cause of death from GI cancer, so it is of paramount importance to study the players and mechanisms that regulate tumor migration, invasion, and metastatic dissemination, in order to evaluate the possibility of using these substrates as new therapeutic targets.
In this review, we first provide an overview of the molecular basis of the tumor microenvironment (TME) in regulating cell migration, invasion, and metastasis formation in four GI tumors: gastric cancer (GC), cholangiocarcinoma (CCA), hepatocellular carcinoma (HCC), and CRC. We also describe the role of miRNAs as tumor invasivity biomarkers with a final focus on nutraceuticals in driving TME remodeling.

2. Gastrointestinal Cancers: A General Overview

2.1. Gastric Cancer

GC is one of the most common malignancies in the world, accounting for 5.6% of all cancers and 7.7% of all cancer-related deaths worldwide [7]. GC can develop in the stomach and spread throughout other organs, such as the small intestines, lymph nodes, liver, pancreas, and colon [8]. Most GCs are adenocarcinomas, which can be further subdivided into intestinal and diffuse types according to the Lauren classification [9,10] and are also associated with infectious agents, including the bacterium Helicobacter pylori and the Epstein–Barr virus [11,12]. Therapies are mainly based on chemotherapy, radiotherapy, and immunotherapy and are effective alternatives for patients who cannot undergo surgical resection [8,13,14]. However, a proportion of patients can develop resistance to therapy with subsequent metastasis formation. About 40% of patients with GC present metastasis, and only 5% of these patients have a 5-year survival [15]. The reasons are complex, and one of the most important issues is the potentiality of cancer cells to metastasize. The prognosis of GC patients with metastatic disease remains poor due to a lack of effective therapies and limited information regarding the mechanisms that regulate metastasis formation [16]. TME plays a key role in the initiation and formation of pre-metastatic niches, promoting the ability of cancer cells to proliferate, invade, and migrate [17]. Indeed, TME regulates multiple signaling pathways involved in migration and invasiveness, such as the Wnt/β-catenin pathway, nuclear factor kappaB (NF-κB), extracellular regulated kinase 1/2 (ERK1/2), transforming growth factor-β (TGF-β)/Smad, and phosphatidylinositol 3-kinase (PI3K)/Akt pathway [5,17].

2.2. Cholangiocarcinoma

CCA includes a heterogeneous group of malignancies arising along the biliary tree epithelium and represents an estimated 3–5% of all GI system malignancies [18,19]. According to their anatomical origin, CCAs are classified into three groups: intrahepatic (10% of all CCA), perihilar (50–60%), or distal (20–30%) [19]. They share similarities but also have important differences that can affect the pathogenesis and outcome. Although this anatomical classification is widely used, other factors such as tumor growth pattern and cell of origin (cholangiocytes, perishable glands, liver progenitor cells, or hepatocytes) provide alternative methods of classification that can better predict tumor behavior [20]. Patients with CCA are often asymptomatic in the early stages of the tumor, and, for most of them, diagnosis occurs at an advanced stage. Both late diagnosis and high chemoresistance of these tumors compromise possible curative treatment options and contribute to their poor prognosis [19]. Currently, the most effective treatment options consist of surgery or liver transplantation, although these are indicated in less than 30% of patients, and the possibility of tumor recurrence is high [21]. For unresectable cases, palliative treatment, consisting of a combination of different chemotherapeutic agents, such as gemcitabine and cisplatin, and the use of immune checkpoint inhibitors (ICIs), remains the only possible option, with overall survival of 12 months [21]. Similar to GC in CCA, TME plays a primary role in the ability of tumor cell invasion and subsequent metastasis formation. Indeed, the increase in TGF-β levels in CCA is responsible for the switch of E-cadherin to N-cadherin, which leads to a subsequent loss of cell–cell adhesion and promotion of tumor invasion [22,23]. In addition, cancer-associated fibroblasts (CAFs) in the adjacent stroma release factors responsible for invasion and metastasis formation through the E-to-N-cadherin conversion and activation of the PI3K-Akt pathway [22,24].

2.3. Hepatocellular Carcinoma

Hepatic carcinoma (HCC) is the most frequent neoplasm of the liver. It originates from the malignant transformation of hepatocytes and frequently evolves from chronic diseases such as hepatitis, fibrosis, and cirrhosis [25]. HCC is the third leading cause of cancer deaths worldwide, with a relative 5-year survival rate of approximately 18%. The similarity between incidence and mortality underlines the dismal prognosis associated with this disease [26]. When diagnosed at an early stage, it is curable with localized approaches, including surgical resection, chemotherapy, radiation therapy, or liver transplantation [27,28]. However, HCC is usually diagnosed in advanced stages when the tumor is unresectable, rendering these treatments ineffective. Hepatic tumor development is controlled by both extracellular factors and intracellular signaling pathways [29]. These pathways are found to be significantly altered and upregulated to promote tumorigenesis and cancer progression. TGF-β plays an important role in HCC tumor progression because it promotes epithelial–mesenchymal transition (EMT) and, thus, tumor cell motility [30]. It is worth noting that along with the role of TGF-β, other signaling pathways such as PI3K/Akt, ERK/c-Jun amino-terminal kinases (JNK), Wnt/β-catenin, and NF-κB are also closely involved in the migration and invasiveness of HCC cancer cells and metastasis formation [29].

2.4. Colorectal Cancer

CRC is the second most common cancer in terms of mortality and the third type in frequency in both men and women [7,31]. About 50% of patients develop early metastasis and a poor prognosis because of resistance to chemotherapy [7,32]. This is due to the different molecular mechanisms responsible for CRC tumor progression. From a genomic standpoint, CRC is not a single disease but a heterogeneous group of malignancies arising within the colon. Indeed, the accumulation of genetic and epigenetic alterations deregulates several signaling pathways responsible for the activation of proliferative signaling, resistance to cell death, escape from immunosurveillance, deregulation of energy metabolism, induction of angiogenesis, and tumor invasion [33]. Approximately 15% of CRC cases display microsatellite instability secondary to a defective mismatch repair [34]. Based on this, CRC may have alternative therapeutic options relying on the administration of certain immunologic agents. Actually, immunotherapy is widely used for the treatment of CRC, along with preoperative radiation therapy, surgery, chemotherapy, and targeted therapy [35,36,37]. Additionally, in CRC, crosstalk between TME and tumor cells plays a key role in invasiveness, metastasis formation, and drug resistance. TME regulates a plethora of cellular signaling pathways that control multiple stages of tumor progression and invasion, such as mitogen-activated protein kinase (MAPKs), PI3K/Akt, NF-κB, JAK/STAT, Wnt/β-catenin, TGF-β, and Notch [38].

2.5. Gastrointestinal Stromal Tumor

Gastrointestinal stromal tumors (GISTs) are a subgroup of rare mesenchymal GI tumors that arise most frequently in the stomach ( ~ 60%) and small intestine ( ~ 25%) and less frequently in the rectum ( ~ 5%) and esophagus (<1%) [39,40]. Recently, the incidence has increased with 10–15 new cases per 100,000/year, mainly in older patients and rarely in younger patients [41,42]. GISTs develop from a particular type of cell called interstitial cells of Cajal (ICC). These are “pacemaker” cells responsible for the physiological contraction of the digestive tract and have specific characteristics according to their localization in the GI tract [43]. ICC and smooth muscle cells derive from the same precursors and express the tyrosine kinase receptor (KIT). While mature smooth muscle cells lose KIT expression, ICCs continue to express it [44]. In GISTs, diffuse hyperplasia of ICC is observed, which is considered a pre-neoplastic lesion. The pathogenesis of GISTs is determined by mutations in KIT or platelet-derived growth factor receptor A (PDGFRA) genes [45]. Surgery is the primary choice for resectable GISTs; however, the therapeutic treatment for unresectable metastatic GIST patients has been radically changed by the introduction of targeted therapy [46]. Administration of imatinib mesylate, together with other kinase inhibitors, has been associated with good improvement in clinical outcomes and survival [46]. Despite the many benefits, patients may develop resistance due to secondary mutations in KIT and PDGFRA that lead to the deregulation of crucial signaling pathways involved in cell proliferation and migration, such as RAS/RAF/MAPK and PI3K/Akt/mTOR [47].

3. Tumor Microenvironment in Promoting Cancer Cells Migration and Invasion

Over the past decade, several studies have shown that cancer growth is determined not only by tumor cells but also by the TME [48]. Indeed, it consists of a complex network of malignant and non-malignant cells that potentiate cancer progression and metastasis and modulate responses to therapy [48,49]. Specifically, the TME is composed of various elements, including tumor cells, immune cells such as T cells, B cells, dendritic cells (DC), myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages (TAMs), CAFs, and blood and lymphatic vessels [50,51]. Intercellular communication is driven by a complex and dynamic network of cytokines, chemokines, growth factors, and inflammatory and matrix-remodeling enzymes, all of which can promote tumor progression and invasion (Figure 1) [52]. In particular, the TME of GI tumors is characterized by the presence of CAFs, which promote tumor growth, angiogenesis, invasion, and metastasis, along with extracellular matrix (ECM) remodeling and even chemoresistance [53]. In addition, TME is defined by the presence of an immunosuppressive immune infiltrate mainly composed of M2 macrophages, MDSCs, and T regulatory cells (Tregs), and finally by the presence of soluble factors such as TGF-β, which has a primary role in promoting tumor migration and metastases (Figure 1) [54].

3.1. Cancer-Associated Fibroblasts

CAFs constitute a dominant stromal component of the TME of GI cancers and contribute in many ways to tumor progression and resistance to therapy [53,55,56]. They originate predominantly from tissue-resident fibroblasts that are activated in response to signals from cancer cells and the TME [57]. In GI tumors, particularly in CRC and HCC, the presence of activated CAFs lead to increased expression of activated markers, such as α-smooth muscle actin (α-SMA) and fibroblast activation protein (FAP), and the production of large amounts of glycoproteins, including tenascin-C and collagen maturation enzymes, which are responsible for ECM remodeling [58,59]. Alterations in the biomechanical properties of the ECM can trigger cancer cell migration [53,60]. CAFs can generate gaps in stromal and basement membrane components that are connected through cell–cell junctions to promote EMT and mediate collective migration of tumor cells through matrix metalloproteases (MMP)-dependent or -independent mechanisms [61]. In addition, CAFs produce interleukin-(IL)-6 that induces the activation of the JAK2/STAT3 axis in GC and promotes EMT, increasing migration and invasion [62].
Activated CAFs can even produce abundant soluble molecules, including basic fibroblast growth factor (bFGF), members of the vascular endothelial growth factor (VEGF) family, platelet-derived growth factor (PDGF), ligands of epidermal growth factor receptor (EGFR), interleukins, and TGF-β [63,64]. The production of these soluble factors promotes cancer invasion and metastasis formation [63]. Further studies have shown that CXCL12 and its receptor CXCR4 derived from CAFs can promote cell invasion in GC and CRC tumors [65]. Activation of the CXCL12/CXCR4 axis in the TME, thus, provides paracrine signaling that mediates integrin β1 clustering on the surface of tumor cells, promoting tumor EMT [65]. In addition, CAFs recruit and polarize immune cells such as macrophages, neutrophils, T lymphocytes, and DCs toward a pro-tumorigenic phenotype by secreting various cytokines, chemokines, and other effector molecules such as IL-6, IL-8, CXCL12, CCL2, TGF-β, SDF-1, VEGF, and indoleamine-pyrrole2,3-dioxygenase (IDO) [66,67,68,69].
Finally, the accumulation of activated CAFs correlates with resistance to therapy. For example, the increase in FAP protein can cause resistance to chemotherapy, radiotherapy, and immune checkpoint blockade [55,70]. Furthermore, in experimental models of CRC, chemotherapy-stimulated CAFs increase the secretion of specific cytokines, such as IL-17A, which promotes chemoresistance through the NF-κB pathway and increases tumor invasion and growth in vivo. Of note, an increase in IL-17 was also observed in patients with therapy-resistant CRC [55,71].

3.2. Cytokines and Chemokines

Cytokines and chemokines are inflammatory mediators secreted by several components of the TME. Their action in promoting cancer development includes an antagonizing antitumor immune response, recruiting tumor-supportive stromal cells and immune-suppressive cells, inducing angiogenesis and metastasis, and altering the responses to therapeutic agents [72].
Several cytokines may contribute to tumor progression, particularly IL-1β, TNFα, and IL-6 play key roles in promoting and enhancing EMT. IL-6, secreted by CAFs, promotes STAT3 activation and, thus, tumor invasion and metastasis formation [73]. Similar to IL-6, IL-23 also acts by increasing the expression level of STAT3 and inducing EMT-mediated metastasis [74].
In addition to cytokines, several chemokines are also responsible for promoting tumor invasiveness. CXCL1 promotes the proliferation and migration of colon cancer cells and has a facilitating effect on tumor angiogenesis [75]. Increased CXCL1 promotes TAM2 migration by inhibiting the recall of CD4+ and CD8+ T cells at tumor sites [76]. CXCL5, in the GC TME, facilitates metastasis by promoting the invasion and migration of tumor cells through the activation of the ERK signaling pathway in cancer cells [77,78]. The CXCL12–CXCR4 axis is particularly important in tumor development to participate in CRC, pancreatic cancer, and HCC metastasis [79,80]. The binding of CXCL12 to its receptor CXCR4 on CRC cells causes pro-metastatic signaling through decreasing E-cadherin and inducing ICAM-1 expression [79]. CXCL12–CXCR4 also induces the Wnt/β-catenin pathway with increased MMP-2, MMP-9, and plasminogen activator and consecutive metastatic initiation in CRC [81]. As mentioned above, CAFs are responsible for the production of CXCL12; several studies have claimed that CXCL12 increases the proliferation, migration, and invasion of CRC cells through the induction of M2 polarization [65]. Another chemokine associated with the development of CRC and its liver metastasis is CXCL8, which promotes angiogenesis, proliferation, invasion, migration, and survival of tumor cells through the induction of EMT [82]. TGF-β promotes the generation of an immunosuppressive environment through the recall of TAM2, MDSC, and Tregs at the tumor site via activation of the Smad transcription factor [83,84,85,86]. TGF-β is a dominant effector in all GI cancers and mediates the conversion of fibroblasts to CAFs, promoting cell migration and tumor invasiveness through the induction of EMT [64,85]. In these tumors, TGF-β may have an antitumoral function in the early stage of tumor development by inducing apoptosis; on the contrary, in more advanced stages, it can support tumor progression by improving cell survival, EMT, migration, invasion, and metastasis [85]. In addition, TGF-β induces MMP-8 gene expression through PI3K/Akt/Rac1 signaling in HCC cells that promotes tumor cell EMT and malignant progression [87]. Finally, in addition to TGF-β, insulin-like growth factor-1 (IGF-1) can also promote cancer proliferation and survival, inducing EMT that contributes to tumor migration, invasiveness, and metastasis [88].

3.3. Immune Infiltrating Cells

The immune cell populations within the TME play a significant role in patient prognosis and response to treatment [89]. The presence of a good immune infiltrate, such as tumor-infiltrating lymphocytes (TILs), correlates with a good prognosis in a large spectrum of solid tumors, including GI cancers [90,91]. However, pro-tumor immune populations, including TAM2, MDSCs, neutrophil N2, and Tregs, are highly present in these tumors [92,93,94]. Each one contributes to tumor aggressiveness through the secretion of inflammatory cytokines and chemokines, key effector molecules such as MMPs, prostaglandin E2 (PGE2), and TGF-β. The production of these molecules in the TME, as already described, is responsible for EMT and the promotion of tumor migration and invasiveness [95,96].
TAMs play an important role in tumor progression by promoting pro-angiogenic and immunosuppressive signaling [97]. Through the production of chemokines such as CCL17, CCL22, and CCL24, they lead to the recruitment of T helper 2 cells, Tregs, eosinophils, and basophils, inducing an immunosuppressive environment [98,99]. In addition, TAM2s produce inflammatory cytokines and TGF-β, which are associated with a more invasive tumor profile [99]. For these reasons, in GI tumors, especially in CRC, GIST, and HCC, they have a pro-tumor and pro-metastatic action, are associated with poor prognosis, and correlate with worse overall survival [100,101].
MDSCs play an important role in suppressing the immune response through a series of secretory factors such as arginases, nitrites, reactive oxygen species (ROS), immunosuppressive cytokines, and the expansion of immunosuppressive cells such as Tregs [102]. In GI tumors, MDSCs are strongly involved in the regulation of the immune system and act to dampen its response to tumors, promoting the escape of tumor cells from immunosurveillance and increasing both metastasis and recurrence [102]. For this reason, they are associated with poor prognosis and low survival [103]. MDSCs induce immunosuppressive effects through the production of several cytokines, such as IL-6, IL-10, PGE2, and TGF-β [104,105,106]. In addition, MDSCs promote tumor progression through the induction of MMP-9 and TGF-β, which are responsible for the establishment of a more invasive tumor phenotype [105].
Tumor-associated neutrophils (TANs) play a controversial role in tumor progression. Although they are known to have antitumor activity, many studies have shown that the presence of TANs is associated with the promotion of tumor metastatic potential and poor prognosis in many tumor types [107,108]. Especially, type 2 TANs have immunosuppressive action characterized by the production of chemokines, some having a role in cancer cell migration, such as CCL2, CCL3, CCL4, CCL8, CXCL8, and CXCL16 [109]. In HCC, TAN2 promotes tumor progression and metastasis by inducing the recruitment of TAM2 and Tregs into the TME [109].
Tregs are essential for the maintenance of immunological homeostasis and self-tolerance. In tumors, they may have extensive suppressive activity, secreting immunomodulatory cytokines and cytolytic molecules that allow them to regulate immune responses [110]. In GI tumors, high numbers of Tregs are often associated with poor prognosis and low survival rates [111,112]. It has been extensively studied that Tregs generate a strong pro-inflammatory environment in GI tumors through the secretion of inflammatory cytokines such as IL-12, TGF-β, and TNF-α promoting an immunosuppressive TME and, therefore, supporting tumor invasiveness and metastasis formation [113,114].

4. miRNA in Gastrointestinal Cancer

MicroRNAs (miRNAs) are small, endogenous, non-coding RNAs, 17–25 nucleotides long that regulate gene-expression post-transcriptionally by recognizing complementary sites in the 3′untraslated region (UTR) of the target messenger RNA (mRNA). A single miRNA is able to target hundreds of mRNAs and influence the expression of many genes. A disruption of the miRNA-mediated gene expression control may lead to environmental stresses usually implicated in the development and progression of human cancer, such as starvation, hypoxia, oxidative stress, and DNA damage [115]. As a matter of fact, miRNAs are involved in cancer initiation, progression, and metastasis. Indeed, many miRNAs are found to be up- or down-regulated in cancer samples when compared to normal tissue (Table 1). miRNAs can modulate the expression of mechanisms such as toll-like receptors, Wnt/β-catenin, Hedgehog, and Jak/Stat signaling pathways [116]. Of note, they can act as oncogenes or tumor suppressors through various mechanisms [117].

5. miRNA and Tumor Microenvironment

As previously described, TME is a key factor in the progression and proliferation of tumor cells and drug resistance [139]. Recently, attention was given to the role of miRNAs in the modulation of different types of cells in the TME, such as immune cells and CAFs [139,140,141]. Aberrant expression of miRNAs leads to the reprogramming of cells in CAFs in HCC and GC. In detail, miR-21 converts hepatic stellate cells in activated CAFs by regulating TGF-β signaling and inducing phosphatase and tensin homolog (PTEN) down-regulation and consequent up-regulation of the PI3K/Akt signaling pathway in HCC [127,142]. miR-1247-3p converts normal fibroblast in CAFs promoting HCC progression [143]. miR-27a acts as an oncogene inducing the reprogramming of normal fibroblast in CAFs promoting cancer proliferation and metastasis in GC [130]. Moreover, miR-106b directly targets the PTEN gene in CAFs promoting proliferation, migration, and invasion of GC cells [144].
CAFs-derived miR-493-5p plays a role in the progression and cell growth in CCA. In fact, miR-493-5p expression levels are higher in extracellular vesicles derived from CAFs than the ones from normal fibroblasts [141]. miRNAs secreted by CAFs also play a role in drug resistance. For example, GC cells’ resistance to cisplatin treatment is due to the expression of miR-522 derived from CAFs [145]. miRNAs secreted by CAFs also play an important role in CRC. Jiang et al. highlighted the correlation between the expression of exosomal miR-181b-3p derived from CAFs and CRC development and progression [146]. A recent study by Liu et al. demonstrated that exosomal miR-29a targets and reduces proteins expressed on the vascular endothelial cells in CRC, facilitating metastasis [147].
Like CAFs, the activation of TAMs also plays a role in the modulation of TME. In GC, exosomal miR-21 derived from TAMs leads to resistance to cisplatin through suppression of apoptosis and activation of the PI3K/Akt signaling pathway in cancer cells [148]. Other miRNAs promote cancer progression and metastasis regulating immune cell responses, such as miR-192-5p, which modulates tumor-infiltrating Tregs in GC activity [149].

6. miRNAs as Potential Biomarkers in Gastrointestinal Cancers

As mentioned above, miRNA levels are altered in different pathological processes of several tumors, including GI cancers. This deregulation may act as a specific tumor signature and could be useful in differential diagnosis and in correlation with specific clinical characteristics. miRNA expression levels could be associated with a better or worse prognosis. For example, miR-451 is associated with a worse prognosis in GC and CRC [150]. Therefore, the idea of using miRNAs as potential biomarkers for GI tumor screening, prognosis, diagnosis, and disease monitoring is increasingly dominant [151,152]. Profiling of miRNAs has advantages over mRNA and protein, such as their stability in body fluids and human formalin-fixed paraffin-embedded (FFPE) tissues, their small size, and their regulatory function on different target molecules and pathways [153].
Moreover, miRNA expression profiles could be analyzed with miRNA microarray platforms, quantitative reverse transcription-polymerase chain reaction (qRT-PCR), or Next-Generation Sequencing (NGS) approaches (such as RNA-seq analysis) [151].

6.1. miRNAs as a Biomarker in Gastric Cancer

Usually, oncogenic miRNAs are up-regulated in GC, while tumor-suppressor miRNAs are down-regulated [151]. An alteration of miRNA expression can induce changes in cell proliferation, cell cycle progression, apoptosis, cell migration, and invasion that may lead to GC (Table 2). Several studies demonstrated that miRNAs are able to promote migration and invasion of cancer cells. Hu et al. studied the over-expression of miR-532 in GC tissues and cells compared to normal stomach tissue and surrounding non-cancer tissue [154]. Wound healing and transwell assay demonstrated that miR-532 induces cell migration and invasion in the GC by targeting Nkd1 and inhibiting the Wnt/β-catenin pathway [154]. Numerous miRNAs affect cell migration and invasion in GC, such as miR-215, targeting FOXO1 and up-regulating activated leukocyte cell adhesion molecule (ALCAM) [151] or miR-186, which modulates Twist1 (Table 2) [155,156]. miR-192-5p expression facilitates cell proliferation, the EMT process, and cell invasion in GC by regulating Wnt, TGF-β, and PI3K/Akt signaling pathways [157,158]. Shayimu et al. studied the role of miR-922 on normal gastric epithelial and GC cell lines. This study highlighted the capability of miR-922 to induce cell invasion and migration by promoting MMP-2 and MMP-9 while suppressing SOCS1 expression [138]. Moreover, miR-922 is able to block apoptosis and promote cell proliferation in GC cell lines [138]. The capability of miR-210 to induce, alone or in synergy with long non-coding RNA MIR210HG, cell migration and metastasis was also studied [137]. In particular, c-myc activates miR-210 and MIR210HG, resulting in the induction of cell migration by promoting MMPs [137]. As noted by Zhang et al., miR-21 has an important role in tumor invasion and metastasis in GC. It is up-regulated in GC, and its main target is RECK, a protein involved in the modulation of MMP-2, MMP-9, and MMP-14 expression [159]. Moreover, miR-21 modulates PTEN and Programmed Cell Death 4 (PDCD4) expression, supporting invasion, migration, EMT, and metastasis. These processes are also promoted by the modulation of several EMT proteins, such as vimentin, SNAIL, ACTA2, and TWIST1 [160]. Other studies reported that miR-196a/b over-expression increases radixin (RDX) protein levels and down-regulates MAX dimerization protein 1 (MDX1), promoting cell migration, invasion, and metastasis [161,162]. Other miRNAs, such as miR-370, interfere with the TGF-β signaling pathway, involved in cell migration, and with the Ubiquinol–cytochrome c reductase core protein 2 (UQCRC2) axis regulating EMT [163,164]. On the other side, decreased expression of tumor suppressor miRNAs, such as miR-218 and let-7, promote tumor invasiveness in GC by eliminating repression of the Robo1 pathway and increasing high mobility group AT-hook 2 (HGMB2) expression, respectively [165,166]. In addition, miR-200 family members increase EMT by reducing the expression of E-cadherin repressors [167]. The down-regulation of miR-335 and miR-153 promotes invasion and metastasis processes targeting BCL-w and SNAIL [168,169]. Furthermore, some miRNAs, such as miR-9, miR-10b, and miR-223, have a dual role in GC progression, acting as both tumor promoters and suppressors according to their target genes [151]. Table 2 lists the principal miRNAs that are deregulated in GC.

6.2. miRNAs as Biomarker in Cholangiocarcinoma

The aberrant expression of several miRNAs (Table 3) modulates the expression of genes involved in the processes of invasion and cell proliferation in CCA. The up-regulation of miR-10a-5p induces proliferation in CCA cell lines by targeting and modifying PTEN and phospho-Akt (ser473) expression [125]. Zhang et al. also demonstrated that the aberrant expression of miR-30a-5p can inhibit CCA cell apoptosis and promote CCA progression by targeting the SOCS3 gene [189]. In 2018, Wan et al. showed that CCA tissues presented an elevated expression of miR-383 compared to normal hepatic tissue samples [190]. Wan et al. demonstrated that the over-expression of miR-383 negatively regulates the interferon regulatory factor-1 (IRF1), involved in the cell cycle, inhibiting its tumor suppressor role and inducing cell proliferation and migration [190]. It was demonstrated that several miRNAs were identified as prognostic and diagnostic biomarkers in CCA. For instance, Kishimoto et al. demonstrated miR-21 over-expression, thus indicating that it may be used as a biomarker able to distinguish cancer patients from healthy ones. Notably, miR-21 targets NAD(+)-linked 15-hydroxyprostaglandin dehydrogenase (15-PGDH/HPGD), promoting cell growth and up-regulates EMT-related KLF4, N-cadherin, vimentin, Akt and ERK1/2, inducing EMT and invasion [191,192,193]. In addition, the over-expression of miR-27a promotes cell proliferation, migration, and invasion. Effectively, high levels of miR-27a are associated with lymph node metastasis and a poor prognosis in CCA patients [194]. Many studies reported an abnormal expression of miR-29b in several human cancers. miR-29b was down-regulated and associated with poor overall survival in CCA cells and tissues [195]. Proliferation tests, together with flow cytometry analysis, demonstrated that miR-29b influences both cell cycle and apoptosis in CCA cell lines [195].
Sheng et al. reported the up-regulation of Yes-associated protein 1 (YAP1), a transcriptional coactivator of the tumor-suppressive Hippo pathway, and implicated in CCA pathogenesis. Moreover, it was discovered that YAP is the target of miR-16, which results in suppressed CCA determining proliferation, invasion, and metastasis [196].
However, it was observed that seven miRNAs, miR-21, miR-26, miR-106a, miR-150, miR-192, and miR-194, could be employed for differential diagnosis to distinguish patients with CCA from controls [197]. Furthermore, it was discovered that the 4-miRNA CCA signature (miR-30a, miR-200c, miR-141, and miR-425) could differentiate CCA from other GI tumors [198] (Table 4).
Table 3. Aberrantly expressed miRNAs in Cholangiocarcinoma, with their specific targets.
Table 3. Aberrantly expressed miRNAs in Cholangiocarcinoma, with their specific targets.
miRNATargetEffectsRefs.
let-7cEZH2Promotes cell migration and invasion
Metastasis
[122]
miR-10a-5pPTENInduces cell proliferation[125]
miR-16YAP1Induces cell proliferation
Promotes cell invasion
Metastasis
[196]
miR-21KLF4
N-Cadherin
VIMENTIN
AKT
ERK1/2
15-PGDH
Induces cell proliferation and EMT
Promotes cell invasion
Metastasis
[191,192,193]
miR-23DNM3Induces cell proliferation[199]
miR-27aD1 CYCLIN
E-Cadherin
KRAS
YAP
Induces cell proliferation
Promotes cell migration and invasion
Metastasis
[194]
miR-29bDNMT3BInfluences cell cycle and apoptosis[195]
miR-30a-5pSOCS3Inhibits apoptosis
Induces cell proliferation
[189]
miR-96MTSS1Induces cell proliferation
Metastasis
[200]
miR-137WNT2BRegulates apoptosis
Induces cell migration and invasion
[201]
miR-181b-5pPARK2Promotes cell migration[202]
miR-196HAND1Promotes cell growth
Metastasis
[203]
miR-320VEGFR2
NRP-1
Induces cell growth and proliferation
Metastasis
[204]
miR-383IRF1Induces cell proliferation
Promotes cell migration
[190]
miR-424-5pARK5Promotes cell migration and invasion
Induces EMT
[205]
Table 4. Up- or down-regulated miRNAs employed in differential diagnosis in Cholangiocarcinoma.
Table 4. Up- or down-regulated miRNAs employed in differential diagnosis in Cholangiocarcinoma.
miRNAUp/Down-RegulatedRef.
miR-21Up-regulated[206]
miR-26Up-regulated[206]
miR-30aUp-regulated[198]
miR-106aDown-regulated[207]
miR-141Down-regulated[198]
miR-150Up-regulated[206]
miR-192Up-regulated[208]
miR-194Up-regulated[209]
miR-200cDown-regulated[198]
miR-425Down-regulated[198]

6.3. miRNAs as Biomarkers in Hepatocellular Carcinoma

Recently, numerous studies have analyzed the role of miRNAs in the development and progression of HCC [210]. miRNA profiles have proven that they are an important source for potential biomarkers as they made possible the distinction of liver cancer cells from hepatocytes [144] (Table 5). The WNT/β-catenin pathway is frequently up-regulated, and it is involved in the proliferation, migration, invasion, and survival of liver cancer cells. Moreover, it was discovered that several miRNAs implicated in this deregulation, including miR-21, miR-106b, miR-135, and miR-315, are overexpressed, and miR-122, miR-145, miR-214 are down-regulated [144]. Additionally, the down-regulation of miR-221/-222, miR-30, and miR-148a contribute to the E-cadherin loss, reducing cell–cell adhesion and promoting EMT [211]. Down-regulation of miR-23b and miR-34a plays a role in HCC progression by inducing cell proliferation, migration, invasion, and metastasis [212].
It has been demonstrated that down-regulation of different miRNAs, such as miR-199a-5p or miR-29c, promotes tumorigenesis and increases cell invasion in HCC by modulating MMP activities and regulating the cell cycle, respectively [213,214]. Instead, miR-9 over-expression promoted migration and invasion in HCC cells, likewise in other types of GI cancer such as colon cancer and GC [123,124,215,216]. Sun et al. studied the connection between the over-expression of miR-1246 and the putative target CADM1, involved in cell–cell interaction, and the capacity of the miRNA to induce metastasis in HCC [217]. Various miRNAs, in addition, enhance HCC progression mediating the EMT event, such as miR-330-3p and miR-192 [218].
About nineteen deregulated miRNAs are implicated in angiogenesis, invasion, and metastasis, such as miR-122, which enhance these processes by inhibiting the p53 signaling pathway and modulating disintegrin and metalloprotease 17 (ADAM17) [219]. miR-139 and miR-151 were also involved in the ADAM17 pathway facilitating angiogenesis, invasion, and metastasis [220].
Aberrant miRNA expression profiles between liver cancer and normal liver tissues have been identified. For instance, Nagy et al. identified several overexpressed miRNAs, such as miR-421, miR-183, miR-182, miR-96, and miR-301, in liver cancer patients. At the same time, they also identified a number of down-regulated miRNAs, including miR-195, miR-139, miR-326, and miR-145 [221].
Table 5. Aberrantly expressed miRNAs in Hepatocellular carcinoma, with their specific targets.
Table 5. Aberrantly expressed miRNAs in Hepatocellular carcinoma, with their specific targets.
miRNATargetEffectsRefs.
miR-9KLF17Promotes cell migration and invasion[123]
miR-21PTEN
PDCD4
Metastasis[222]
miR-23buPA
MET
Promotes cell proliferation
Induces cell invasion
[212,223]
miR-29cRPS15APromotes cell invasion
Regulates cell cycle
[214]
miR-30SNAILInduces EMT[211]
miR-34aHDAC1
D1 CYCLIN
CDK2/4
FOXMI
BCL-2
Induces cell proliferation
Promotes cell invasion and migration
Drug resistance
[212,224,225,226]
miR-96SOX6Induces cell proliferation
Promotes cell migration and invasion
[227]
miR-106bPTENInduces cell proliferation
Promotes cell migration and invasion
[228]
miR-122ADAM17
WNT1
TACE
LMNB2
Induces cell proliferation
Promotes cell invasion
Angiogenesis and metastasis
[219,229,230]
miR-124-3pCRKLPromotes cell migration and invasion
Metastasis
[231]
miR-130bNotch-Dll1Promotes cell migration and invasion[232]
miR-135APC
AXIN
Metastasis[222]
miR-139ADAM17
ROCK2
Induces cell proliferation
Metastasis
[229]
miR-144FOXK1Modulates glycolysis[233]
miR-145IRS1 IRS2 OCT4
β-Catenin IGF-IR
Induces cell proliferation
Promotes cell migration and invasion
[234]
miR-148ac-MET
HRIP
c-MYC WNT1
SNAIL1 DNMT1
Induces EMT
Metastasis
[211]
miR-151ADAM17
RHOGDIA
Promotes cell invasion
Angiogenesis and metastasis
[235]
miR-182FOXO3a
MTSSI
pRB
CEPBA
RASA1
c-MYC
Induces cell proliferation
Angiogenesis and metastasis
[236,237,238]
miR-183SOCS6Induces cell proliferation
Promotes cell invasion
[239]
miR-185AKT1Induces cell proliferation[240]
miR-195CDK6
CYCLIN D1
YAP
WNT3a
VEGF
Regulates cell cycle and apoptosis
Induces EMT
Angiogenesis and metastasis
[241,242]
miR-199DDR1
mTOR
c-Met
Promotes cell invasion
Regulates cell cycle
Drug resistance
[213,243]
miR-214HDGF
β-Catenin
Angiogenesis[244]
miR-221/-222PTEN
E-cadherin
Induces EMT[211]
miR-301GAXMetastasis[222]
miR-315APC
Axin
Metastasis[222]
miR-326LASP1
RAB21
Induces cell proliferation
Promotes cell invasion
[245]
miR-330-3pEREGRegulates EMT[218]
miR-409JAK2
STAT3
Inhibits apoptosis
Induces cell proliferation
Promotes cell viability
[246]
miR-421SOX9
PTEN
MMP-3
Induces cell proliferation and EMT
Promotes cell invasion
[247,248,249]
miR-520c-3pPTENPromotes cell migration and invasion[250]
miR-539MAP2K1Promotes cell migration and invasion
Induce cell proliferation
Inhibits apoptosis
[251]
miR-579-3pPIK3CATumor development[252]
miR-1246CADM1Metastasis[217]
miR-4521FAM129ARegulates cell growth and apoptosis[253]

6.4. miRNAs as Biomarkers in Colorectal Cancer

As mentioned above, CRC cells are characterized by a high capability of proliferation, invasion, and metastasis. Among the factors that are involved in CRC development, epigenetic modifications have been reported. CRC pathogenesis is characterized by a stage-specific miRNA expression. Table 6 lists miRNAs involved in the development and progression of CRC. Let-7, miR-21, miR-29a, and cluster miR-17-92 are relevant [254]. Let-7 is deregulated in several GI cancers and modulates Ras and Myc expression controlling tumor progression and metastasis in CRC [255]. The increased expression level of miR-21 is associated with invasion and liver and lymph node metastasis [256]. miR-29a is a metastasis-promoter due to its tumor suppressor target gene Kruppel-like factor 4 (KLF4) that up-regulates MMP-2 expression and, at the same time, down-regulates E-cadherin expression [257]. Cluster miR-17-92 is typically amplified and has several target genes involved in CRC progression and metastasis. Especially, miR-17 targets the MYC family, increasing tumor progression and invasion [258,259]. Sun et al. demonstrated the role of miR-103a-3p in the promotion of cell invasion and metastasis by regulating the glycolysis process mediated by the Hippo pathway [134]. MiR-152-3p also promotes cell adhesion and metastases. In particular, miR-152-3p negatively regulates the Aquaporin-11 (AQP11) protein, which is usually involved in the repression of cell growth and adhesion [135]. Moreover, Zhu et al. suggested that inhibition of miR-152-3p could stop the progression of CRC [135]. The aberrant expression of miR-23a-3p is also involved in CRC development. This miRNA, in fact, inhibits the expression of NDRG4 in cancer cells with the consequent increase in cell proliferation, migration, and invasion [129]. miRNAs are also implicated in drug resistance phenomena. It has been demonstrated that miR-93-5p is up-regulated in CRC cells that are resistant to chemotherapeutics and may regulate proteins involved in multidrug resistance and target cyclin-dependent kinase inhibitors [133]. Additionally, miR-101 was reported as a tumor suppressor that is down-regulated in CRC, and its direct target is Cyclooxygenase-2 (COX-2), whose high expression contributes to cell growth and invasion [153].
The over-expression of miR-125b promotes invasion and EMT, enhancing CXCR4 expression; in turn, the CXCL12/CXCR4 axis induces miR-125b expression. Moreover, miR-125b down-regulates the p53 pathway, promoting cell proliferation [260].
Recent studies have demonstrated that the tumor suppressor miR-137-3p is down-regulated in several cancers, such as non-small cell lung cancer (NSCLC), liver cancer, breast cancer, and CRC. miR-137-3p promotes migration, EMT, and invasion of CRC cells in a lysine-specific demethylase 1 (LDS-1)-dependent manner [260].
In addition, miR-106b up-regulation determines the enhancement of proliferation, invasion, and migration in CRC cells via PTEN modulation [144].
miR-145-5p targets cell cycle-associated protein-3 (CDCA3), determining cell growth and EMT suppression [261].
Finally, Volker et al. investigated miR-192, miR-17, and miR-200c down-regulation in CRC invasion and metastatic process. These three miRNAs negatively control target genes associated with ECM remodeling in fibroblasts. Notably, miR-192 targets integrins, such as ITGB1 and ITGAV, inhibiting cell adhesion and metastasis; miR-200c modulates the EMT process via E-cadherin repressors, ZEB1 and ZEB2 [259]. In fact, in this study, it was shown that the enhancement of miR-192, miR-17, and miR-200c suppresses the invasiveness and metastasis of CRC cells.
Table 6. Aberrantly expressed miRNAs in Colorectal cancer, with their specific targets.
Table 6. Aberrantly expressed miRNAs in Colorectal cancer, with their specific targets.
miRNATargetEffectsRefs.
let-7RAS
MYC
Induces cell progression
Metastasis
[255]
miR-9E-CadherinPromotes cell migration and invasion[215]
miR-17P130Induces cell progression[262]
miR-17-92 clusterC-MYC
E2F
Induces cell progression
Promotes cell invasion
[258,259]
miR-20a-5pSMAD4Promotes cell invasion and migration
Metastasis
[263]
miR-21TNF-α
PDCD4
RECK
PTEN
Promotes cell migration
Induces cell proliferation and EMT
Metastasis
[256,264]
miR-23a-3pNDRG4Induces cell proliferation
Promotes cell migration and invasion
[129]
miR-29aKLF4
MMP-2
Induces EMT
Metastasis
[257]
miR-31FIH1Promotes cell invasion and migration
Induces cell proliferation
[265]
miR-34TP53Induces cancer progression[266]
miR-93-5pCDK inhibitorDrug resistance[133]
miR-101COX-2
ZEB1
EZH2
Induces cell proliferation and EMT
Promotes cell invasion
[153]
miR-103a-3pHyppoPromotes cell invasion
Metastasis
[134]
miR-106bPTENInduces cell proliferation
Promotes cell migration and invasion
[144]
miR-125bBAK1
BMF
CXCR4
Induces cell proliferation and EMT
Promotes cell invasion
[260]
miR-126CXCR4Metastasis[267]
miR-135APCPromotes tumorigenesis[268]
miR-137-3pLDS-1Induces EMT
Promotes cell migration and invasion
[260]
miR-145-5pCDCA3Induces EMT
Promotes cell invasion
[261]
miR-148aMMP7Promotes cell invasion[269]
miR-152-3pAQP11Metastasis[135]
miR-192ITGB1
ITGAV
Promotes cell invasion
Metastasis
[259]
miR-200cSOX2
ZEB1
ZEB2
Promotes cell invasion and EMT[259,270]
miR-483EI24Induces cell proliferation
Promotes cell invasion
Metastasis
[271]

6.5. miRNAs as Biomarkers in Gastrointestinal Stromal Tumors

A growing number of research has shown that miRNAs play an important role not only in tumorigenesis but also in the risk stratification of GIST [272]. Table 7 briefly summarizes miRNAs of major significance in invasion, EMT, and metastasis in GIST.
miR-196a is one of the most relevant miRNAs involved in tumor progression. In fact, a positive correlation was found between the over-expression of miR-196a and tumor progression from metaplasia to adenocarcinoma [272]. In particular, miR-196a up-regulates annexin A1 (ANXA1), involved in the invasion process, increasing the risk of metastasis formation [273]. miR-186 inhibition is associated with the over-expression of genes implicated in GIST metastasis by modulating the Met/Akt signaling pathway [274]. Therefore, miR-196a over-expression and low expression of miR-186 are associated with poorer prognosis in GIST patients [47].
Yamamoto et al. demonstrated that the down-regulation of miR-133b determines the over-expression of Fascin-1, an actin-binding protein important in the regulation of cell adhesion and migration. In addition, they discovered the correlation between over-expression of Fascin-1 and worse prognosis in GIST patients. For this reason, Fascin-1 should be a useful biomarker to predict cancer aggressiveness [275].
It was found that the down-regulation of miR-137 can control EMT in GIST via TWIST1 inhibition [276]. Liu et al. demonstrated that miR-152 plays a tumor suppressor role by targeting genes associated with cell proliferation, migration, and invasion, and it results in down-regulated in GIST [277]. Recently, a study on miR-218 showed that its down-regulation inhibits cell proliferation, migration, and invasion through direct targeting of the KIT gene [278]. Many different tumors exhibit a down-regulation of let-7 family members, which is also implied in GIST migration, invasion, and metastasis [272].
Gyvyte et al. investigated two deregulated miRNAs in GIST: miR-375-3p and miR-200b-3p. Their down-regulation increases cell viability and migration through the modulation of different target gene expressions, such as KIT, EGFR, ETV1, and the JAK/STAT3 pathway [279].
Table 7. Aberrantly expressed miRNAs in Gastrointestinal Stromal cancer, with their specific targets.
Table 7. Aberrantly expressed miRNAs in Gastrointestinal Stromal cancer, with their specific targets.
miRNATargetEffectsRefs.
let-7-cHOXA1
MMP1
C/EBP-α
Induces cell proliferation
Promotes cell migration and invasion
[272]
miR-133bFSCN1Enhance cell proliferation
Promotes cell invasion
[276]
miR-137TWIST1Induces cell cycle arrest
Promotes cell migration and EMT
Apoptosis
[276]
miR-152CTSLInduces cell proliferation
Promotes cell migration and invasion
[277]
miR-186IGFBP3
AKT
HGFR
CXCR4
EFEMP1
Promotes cell migration and invasion
Metastasis
[274]
miR-196aANXA1Promotes cell invasion[272,273]
miR-218AKT
KIT
Induces cell proliferation
Promotes cell migration and invasion
[276,278]
miR-200b-3EGFR
ETV1
STAT1
Induces cell proliferation
Promotes cell migration and invasion
[279]
miR-375-3pKIT
PDGFRA JAK2
Induces cell proliferation
Promotes cell migration and invasion
[279]

7. Other Biomarkers in Gastrointestinal Cancers

In addition to the use of miRNAs as possible biomarkers, several molecules have emerged over the years as biomarkers helpful for the diagnosis, follow-up, and response to treatment in GI cancers. To date, the most widely used biomarker in the clinic for GI cancer is carcinoembryonic antigen (CEA), a protein found in CRC and CCA patients that plays an important role in cell adhesion and intracellular signaling [280]. It is used as a prognostic, diagnostic, and response-to-therapy biomarker and as a target for cancer immunotherapy (DOTAP) [281]. Indeed, high levels of CEA in the blood of patients with CRC are associated with disease progression, while a decrease is found in patients after surgery [282]. Moreover, tumor-associated antigens, such as CA19-9, a marker in the diagnosis and follow-up of some GI cancers such as pancreatic cancer, CRC, GC, and CCA [283,284,285], are extensively used. The first is found at high levels in the serum of CRC patients, while the second is considered the first tumor marker for GC [286,287,288]. Alpha-fetoprotein (AFP) is a glycoprotein capable of binding to different types of membrane receptors and intracytoplasmatic proteins, blocking or enhancing the responses of intracellular signaling pathways. It is an important biomarker used for early diagnosis and prognosis of HCC patients [289].

8. Nutraceuticals

Nowadays, conventional anticancer therapy implicates the use of radiotherapy and chemotherapy, but such treatments are still expensive and inefficient, especially due to drug resistance and severe adverse events. To date, there are many experimental studies showing the usefulness of nutraceuticals as a complementary approach to standard therapy, thanks to their low toxicity and multiple biological activities [290,291]. Nutraceuticals are also known as bioactive compounds isolated from plants or food and include dietary fibers, antioxidants, phytochemicals, polyunsaturated fatty acids, amino acids, prebiotics and probiotics, and other types of natural food [292].
Many of them have multiple therapeutic properties, such as antioxidant, anti-inflammatory and anticancer, and they seem to be very interesting in the management or treatment of malignancies, cardiovascular diseases, diabetes, obesity, osteoporosis, and the immune system, used alone or in combination with other drugs [293].
Much evidence has revealed that numerous amounts of bioactive substances or extracts from medical plants play an important role in the treatment of different types of GI tumors [290].
Cairicoside E (CE) is a natural herbal medical compound isolated from Ipomoea Cairica (Convolvulaceae). CE modulates the EMT through the down-regulation of aquaporine-5 (AQP5), determining an anti-metastatic effect [294].
Berberine (BBR) is an alkaloid used in Chinese plant-based medicines. It has a wide spectrum of therapeutic actions, such as inhibition of cell proliferation, migration, induction of cellular death, and the enhancement of chemosensitivity through the modulation of NF-κB, PI3K/Akt, and the MAPK signaling pathways. In addition, it is involved in immunity, inflammation [295,296], and, in some cases, in reversing gastric cancerogenesis [297].
Particularly, BBR up-regulates E-cadherin expression and down-regulates N-cadherin, fibronectin, and vimentin expressions to modulate the EMT. Furthermore, BBR suppresses migration and invasion of tumor cells via the IL-6/JAK2/STAT3 signaling pathway and inhibiting MMP-9 protein levels.
BBR up-regulates miR-203 expression, a tumor-suppressive miRNA that binds at the 3′-UTR of Bcl-w oncogene, also resulting in a decrease in chemoresistance [298].
Oleanolic acid (OA) is a pentacyclic triterpene isolated from several plants, including Olea europaea [299]. An antitumoral effect has been demonstrated through the over-expression of miR-98-5p involved in the regulation of Treg/Th17 balance in GC tissues [300].
Paeoniflorin is a bioactive substance of Radix Paeoniae Rubra and is potentially used as a novel therapeutic agent in GC TME. In fact, it improves the immune microenvironment through the up-regulation of miR-149 in gastric CAFs, inhibiting the secretion of IL-6 and leading to the inactivation of the IL-6/STAT3/MMP signaling axis [301].
Sulforaphane is an isothiocyanate derived from the Brassicaceae family, including cabbage and broccoli. It has several activities, including chemopreventive and chemotherapeutic, in different tumors such as lung, bladder, and CRC. As an anticancer drug, Sulforaphane affects all three stages of carcinogenesis. Many studies revealed that it is able to suppress progression and angiogenesis processes in CRC by the inhibition of HIF-1α and VEGF expression [302,303].
Resveratrol (RV) is a polyphenol found in red wine, grapes, peanuts, and other food products that has been reported to have antineoplastic activity on different malignancies [304]. It could present an antitumor potentiality in CCA. In fact, RV can inhibit the secretion of IL-6 from CAFs, preventing the induction of EMT and cancer cell migration. Furthermore, it strongly promotes E-cadherin expression while suppressing N-cadherin, thus, resulting in a reverse phenomenon of the EMT and a reduction in invasiveness and metastasis in cancer cells [305].
Curcumin (CUR) is a non-flavonoid polyphenol purified from the rhizome of the Curcuma longa. It is a pleiotropic molecule with anti-inflammatory, antioxidant, immunomodulatory, and antimicrobial properties [306]. Recent studies have shown that CUR has an antitumor effect on several malignancies, including GI cancers [307].
CUR is a multitarget drug capable of decreasing the expression of molecules involved in angiogenesis, such as VEGF, and tumor invasion, such as intracellular adhesion molecule-1 (ICAM-1), MMP-2, and MMP-9 in CCA [308].
Furthermore, CUR can suppress proliferation, migration, and invasion processes promoting apoptosis of cancer cells in HCC by targeting the circ_0078710/miR-378b/PRIM2 signaling pathway [309] and up-regulating the miR-200 family, involved in the EMT suppression [310].
CUR down-regulates miR-21 expression in CRC and GC. As mentioned above, miR-21 plays an important role in cancer cell migration and invasion through the activation of the PI3K/Akt/mTOR pathway and modulation of MMP-2, MMP-9, and MMP-14 [159,310].
Several plant- and food-derived compounds have a high potential to treat GI tumors in line with relatively low toxicity to normal cells. Indeed, they could be used in combination with conventional anticancer drugs in order to have a potential synergistic effect in cancer therapy [295,311].

9. Conclusions and Perspective

In summary, this review systematically describes aspects related to the mechanisms responsible for tumor migration and invasiveness in GI cancers. Metastasis is the leading cause of death from GI cancer, so it is of paramount importance to study the mechanisms that regulate tumor migration, invasion, and metastasis to propose novel therapeutic targets and biomarkers. The migration of tumor cells and their ability to form metastases are regulated by mechanisms driven by the TME. In GI tumors, the TME is composed of different cell types which cooperate to promote and trigger metastasis formation. Specifically, CAFs play a pivotal role in tumor progression by enhancing EMT and, thus, promoting migration and metastasis formation. These processes are further supported by the presence of an immunosuppressive infiltrate, mainly composed of Treg, MDSC, and TAM2 cells, and by the presence of chemokines and cytokines such as CXCL12, IL-6, and TGF- β.
Recent studies have also demonstrated the critical role of miRNAs in promoting tumor progression and metastasis formation in GI tumors. They may modulate signaling pathways involved in migration and tumor invasion, such as Wnt/β-catenin, Hedgehog, and Jak/Stat. Many miRNAs are found to be up- or down-regulated in tumor tissues compared to healthy tissues, and to date, they are being considered as potential predictors for prognosis, monitoring, and diagnosis of GI cancer patients. Of note, recent studies have shown that the use of nutraceuticals, bioactive compounds isolated from plants or foods, would be useful in GI cancers as a complementary approach to standard therapy. Indeed, they are able to target miRNAs and other molecules that regulate several mechanisms involved in tumor migration and invasiveness. This could pave the way for new targeted therapies for GI cancer treatment.
The high aggressiveness and late diagnosis in patients with GI tumors often result in a poor prognosis. Therefore, it is necessary to develop new tools to improve early diagnosis and prognosis for these patients. A growing number of studies are highlighting the important role of miRNAs as potential markers in many neoplasms. The use of miRNAs as biomarkers offers the great advantage of developing minimally invasive methods due to their stability, which renders them easy to detect in different body fluids, such as blood.
On the other hand, the main disadvantage of their use in large-scale therapeutic applications could be the difficulty of predicting the overall effect of the miRNAs of interest due to the possible high number of target genes. Especially in a therapeutic context, their application could represent a risk because of the occurrence of unknown and unpredictable side effects, possibly triggering physiological function disorders or the development of additional diseases. For this reason, Zhang et al. proposed the design of new delivery systems for miRNA that recognize specific features at the site of the malignant lesion [312].
Further investigations are, therefore, needed for the application of miRNAs as tumor biomarkers. In fact, they could offer important opportunities for the development of new diagnostic and prognostic strategies that would help to improve the clinical outcome of patients with GI tumors.

Author Contributions

V.L., D.N., V.A., M.B., C.F., R.C., Z.M.B., R.B. and L.M. contributed to the writing, reviewing, and editing of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Ministero della Salute, Piano Nazionale Complementare “D3 4 Health—Digital Driven Diagnostics, prognostics and therapeutics for sustainable Health care”. n. PNC0000001 (V.L. and L.M.); Progetto Ateneo Sapienza 2021 n. RM12117A7F986146 (L.M.); Ministero dell’Università e della Ricerca, PRIN 2020, BeiR20Prin, CUP: E85F22000060006 (R.B.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

V.A. is a recipient of the Sapienza Ph.D. program in Molecular Medicine. R.C. is a recipient of the Tor Vergata Ph.D. program in Tissue Engineering and Remodeling Biotechnologies for Body Functions.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ALCAMActivated leukocyte cell adhesion molecule
ANXA1Annexin A1
AQP5Aquaporine-5
AQP11Aquaporine-11
AFPAlpha-fetoprotein
bFGFBasic fibroblast growth factor
BBRBerberine
CECairicoside E
CAFsCancer-associated fibroblasts
CEACarcinoembryonic antigen
CTSLCathepsin L
C/EBP-αCCAAT Enhancer Binding Protein Alpha
CXCR4CXC chemokine receptor 4
CDCA3Cell cycle-associated protein-3
CCACholangiocarcinoma
JNKc-Jun amino-terminal kinases
CRCColorectal cancer
CURCurcumin
COX-2Cyclooxygenase-2
DCDendritic cells
ADAM17Disintegrin and metalloprotease 17
EFEMP1Epidermal growth factor-containing fibulin-like extracellular matrix protein 1
EGFREpidermal growth factor receptor
EMTEpithelial-mesenchyme transition
ETV1ETS transcription factor 1
ECMExtracellular matrix
ERK1/2Extracellular regulated kinase 1/2
FSCN1Fascin actin-bundling protein 1
FAPFibroblast activation protein
FFPEFormalin-fixed paraffin-embedded
GCGastric cancer
GIGastrointestinal
GISTGastrointestinal stromal tumor
HCCHepatocellular carcinoma
HGFRHepatocyte growth factor receptor
HGMB2High mobility group AT-hook 2
ICIsImmune checkpoint inhibitors
IDOIndoleamine-pyrrole2,3-dioxygenase
IGF-1Insulin-like growth factor-1
IGFBP3Insulin-like growth factor-binding protein 3
IRF1Interferon regulatory factor-1
ICCInterstitial cell of Cajal
ICAM-1Intracellular adhesion molecule-1
JAKJanus kinase
KLF4Kruppel-like factor 4
LDS-1Lysine-specific demethylase 1
MMPMatrix metalloproteases
MDX1MAX dimerisation protein 1
mRNAMessenger RNA
miRNAsMicroRNAs
MAPKsMitogen-activated protein kinase
MDSCsMyeloid-derived suppressor cells
15-PGDH/HPGDNAD(+)-linked 15-hydroxyprostaglandin dehydrogenase
NGSNext-Generation Sequencing
NSCLCNon-small cell lung cancer
NF-κBNuclear factor kappaB
OAOleanolic acid
PTENPhosphatase and tensin homolog
PI3KPhosphatidylinositol 3-kinase
PDGFPlatelet-derived growth factor
PDGFRAPlatelet-derived growth factor receptor A
PDCD4Programmed Cell Death 4
PGE2Prostaglandin E2
AKTProtein kinase B
qRT-PCRQuantitative reverse transcription-polymerase chain reaction
RDXRadixin
RKTReceptor tyrosine kinase
ROSReactive oxygen species
RVResveratrol
RECKReversion Inducing Cysteine Rich Protein with Kazal Motifs
STATSignal transducer and activator of transcription
TregsT regulatory cells
TGF-βTransforming growth factor-β
TMETumor microenvironment
TAMsTumor-associated macrophages
TANsTumor-associated neutrophils
TILsTumor-infiltrating lymphocytes
TWISTTwist family bHLH transcription factor 1
KITTyrosine kinase
UQCRC2Ubiquinol-cytochrome c reductase core protein 2
VEGFVascular endothelial growth factor
YAP1Yes-associated protein 1
α-SMAα-smooth muscle actin

References

  1. Losurdo, P.; de Manzini, N.; Palmisano, S.; Grassi, M.; Parisi, S.; Rizzolio, F.; Tierno, D.; Biasin, A.; Grassi, C.; Truong, N.H.; et al. Potential Application of Small Interfering RNA in Gastro-Intestinal Tumors. Pharmaceuticals 2022, 15, 1295. [Google Scholar] [CrossRef] [PubMed]
  2. Arnold, M.; Abnet, C.C.; Neale, R.E.; Vignat, J.; Giovannucci, E.L.; McGlynn, K.A.; Bray, F. Global Burden of 5 Major Types of Gastrointestinal Cancer. Gastroenterology 2020, 159, 335–349.e15. [Google Scholar] [CrossRef] [PubMed]
  3. Wang, D.-K.; Zuo, Q.; He, Q.-Y.; Li, B. Targeted Immunotherapies in Gastrointestinal Cancer: From Molecular Mechanisms to Implications. Front. Immunol. 2021, 12, 705999. [Google Scholar] [CrossRef] [PubMed]
  4. Petricevic, B.; Kabiljo, J.; Zirnbauer, R.; Walczak, H.; Laengle, J.; Bergmann, M. Neoadjuvant immunotherapy in gastrointestinal cancers–The new standard of care? Semin. Cancer Biol. 2022, 86 Pt 2, 834–850. [Google Scholar] [CrossRef] [PubMed]
  5. Tan, Z.; Sun, W.; Li, Y.; Jiao, X.; Zhu, M.; Zhang, J.; Qing, C.; Jia, Y. Current Progress of EMT: A New Direction of Targeted Therapy for Colorectal Cancer with Invasion and Metastasis. Biomolecules 2022, 12, 1723. [Google Scholar] [CrossRef]
  6. Li, D.; Zhang, X.; Jiang, L. Molecular mechanism and potential therapeutic targets of liver metastasis from gastric cancer. Front. Oncol. 2022, 12, 1000807. [Google Scholar] [CrossRef]
  7. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  8. Joshi, S.S.; Badgwell, B.D. Current treatment and recent progress in gastric cancer. CA Cancer J. Clin. 2021, 71, 264–279. [Google Scholar] [CrossRef]
  9. The Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature 2014, 513, 202–209. [Google Scholar] [CrossRef] [Green Version]
  10. Laurén, P. The Two Histological Main Types of Gastric Carcinoma: Diffuse and so-called Intestinal-Type Carcinoma. An Attempt at a Histo-Clinical Classification. Acta Pathol. Microbiol. Scand. 1965, 64, 31–49. [Google Scholar] [CrossRef]
  11. Yoon, H.; Kim, N. Diagnosis and Management of high risk Group for Gastric Cancer. Gut Liver 2015, 9, 5–17. [Google Scholar] [CrossRef] [Green Version]
  12. Malfertheiner, P.; Camargo, M.C.; El-Omar, E.; Liou, J.-M.; Peek, R.; Schulz, C.; Smith, S.I.; Suerbaum, S. Helicobacter pylori infection. Nat. Rev. Dis. Prim. 2023, 9, 19. [Google Scholar] [CrossRef]
  13. Sato, Y.; Okamoto, K.; Kida, Y.; Mitsui, Y.; Kawano, Y.; Sogabe, M.; Miyamoto, H.; Takayama, T. Overview of Chemotherapy for Gastric Cancer. J. Clin. Med. 2023, 12, 1336. [Google Scholar] [CrossRef]
  14. Shen, J.; Wang, Z. Recent advances in the progress of immune checkpoint inhibitors in the treatment of advanced gastric cancer: A review. Front. Oncol. 2022, 12, 934249. [Google Scholar] [CrossRef]
  15. Li, Y.; Feng, A.; Zheng, S.; Chen, C.; Lyu, J. Recent Estimates and Predictions of 5-Year Survival in Patients with Gastric Cancer: A Model-Based Period Analysis. Cancer Control 2022, 29, 10732748221099227. [Google Scholar] [CrossRef]
  16. Machlowska, J.; Baj, J.; Sitarz, M.; Maciejewski, R.; Sitarz, R. Gastric Cancer: Epidemiology, Risk Factors, Classification, Genomic Characteristics and Treatment Strategies. Int. J. Mol. Sci. 2020, 21, 4012. [Google Scholar] [CrossRef]
  17. Oya, Y.; Hayakawa, Y.; Koike, K. Tumor microenvironment in gastric cancers. Cancer Sci. 2020, 111, 2696–2707. [Google Scholar] [CrossRef]
  18. Silvestri, M.; Vu, T.N.; Nichetti, F.; Niger, M.; Di Cosimo, S.; De Braud, F.; Pruneri, G.; Pawitan, Y.; Calza, S.; Cappelletti, V. Comprehensive transcriptomic analysis to identify biological and clinical differences in cholangiocarcinoma. Cancer Med. 2023, 12, 10156–10168. [Google Scholar] [CrossRef]
  19. Banales, J.M.; Marin, J.J.G.; Lamarca, A.; Rodrigues, P.M.; Khan, S.A.; Roberts, L.R.; Cardinale, V.; Carpino, G.; Andersen, J.B.; Braconi, C.; et al. Cholangiocarcinoma 2020: The next horizon in mechanisms and management. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 557–588. [Google Scholar] [CrossRef]
  20. Testa, U.; Pelosi, E.; Castelli, G. Cholangiocarcinoma: Molecular Abnormalities and Cells of Origin. Technol. Cancer Res. Treat. 2023, 22, 15330338221128689. [Google Scholar] [CrossRef]
  21. Moazzami, B.; Majidzadeh-A, K.; Dooghaie-Moghadam, A.; Eslami, P.; Razavi-Khorasani, N.; Iravani, S.; Khoshdel, A.; Shahi, F.; Dashti, H.; Mehrvar, A.; et al. Cholangiocarcinoma: State of the Art. J. Gastrointest. Cancer 2020, 51, 774–781. [Google Scholar] [CrossRef] [PubMed]
  22. Fabris, L.; Perugorria, M.J.; Mertens, J.; Björkström, N.K.; Cramer, T.; Lleo, A.; Solinas, A.; Sänger, H.; Lukacs-Kornek, V.; Moncsek, A.; et al. The tumour microenvironment and immune milieu of cholangiocarcinoma. Liver Int. 2019, 39 (Suppl. S1), 63–78. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Høgdall, D.; Lewinska, M.; Andersen, J.B. Desmoplastic Tumor Microenvironment and Immunotherapy in Cholangiocarcinoma. Trends Cancer 2018, 4, 239–255. [Google Scholar] [CrossRef] [PubMed]
  24. Carloni, R.; Rizzo, A.; Ricci, A.D.; Di Federico, A.; De Luca, R.; Guven, D.C.; Yalcin, S.; Brandi, G. Targeting tumor microenvironment for cholangiocarcinoma: Opportunities for precision medicine. Transl. Oncol. 2022, 25, 101514. [Google Scholar] [CrossRef]
  25. Chidambaranathan-Reghupaty, S.; Fisher, P.B.; Sarkar, D. Hepatocellular carcinoma (HCC): Epidemiology, etiology and molecular classification. Adv. Cancer Res. 2021, 149, 1–61. [Google Scholar] [CrossRef]
  26. Vogel, A.; Meyer, T.; Sapisochin, G.; Salem, R.; Saborowski, A. Hepatocellular carcinoma. Lancet 2022, 400, 1345–1362. [Google Scholar] [CrossRef]
  27. Ailia, M.J.; Heo, J.; Yoo, S.Y. Navigating through the PD-1/PDL-1 Landscape: A Systematic Review and Meta-Analysis of Clinical Outcomes in Hepatocellular Carcinoma and Their Influence on Immunotherapy and Tumor Microenvironment. Int. J. Mol. Sci. 2023, 24, 6495. [Google Scholar] [CrossRef]
  28. Rizvi, S.; Khan, S.A.; Hallemeier, C.L.; Kelley, R.K.; Gores, G.J. Cholangiocarcinoma—Evolving concepts and therapeutic strategies. Nat. Rev. Clin. Oncol. 2018, 15, 95–111. [Google Scholar] [CrossRef] [Green Version]
  29. Liu, P.; Kong, L.; Liu, Y.; Li, G.; Xie, J.; Lu, X. A key driver to promote HCC: Cellular crosstalk in tumor microenvironment. Front. Oncol. 2023, 13, 1135122. [Google Scholar] [CrossRef]
  30. Tu, S.; Huang, W.; Huang, C.; Luo, Z.; Yan, X. Contextual Regulation of TGF-β Signaling in Liver Cancer. Cells 2019, 8, 1235. [Google Scholar] [CrossRef] [Green Version]
  31. Sawicki, T.; Ruszkowska, M.; Danielewicz, A.; Niedźwiedzka, E.; Arłukowicz, T.; Przybyłowicz, K.E. A Review of Colorectal Cancer in Terms of Epidemiology, Risk Factors, Development, Symptoms and Diagnosis. Cancers 2021, 13, 2025. [Google Scholar] [CrossRef]
  32. Citarella, A.; Catanzaro, G.; Besharat, Z.M.; Trocchianesi, S.; Barbagallo, F.; Gosti, G.; Leonetti, M.; Di Fiore, A.; Coppola, L.; Autilio, T.M.; et al. Hedgehog-GLI and Notch Pathways Sustain Chemoresistance and Invasiveness in Colorectal Cancer and Their Inhibition Restores Chemotherapy Efficacy. Cancers 2023, 15, 1471. [Google Scholar] [CrossRef]
  33. Testa, U.; Pelosi, E.; Castelli, G. Colorectal cancer: Genetic abnormalities, tumor progression, tumor heterogeneity, clonal evolution and tumor-initiating cells. Med. Sci. 2018, 6, 31. [Google Scholar] [CrossRef] [Green Version]
  34. Taieb, J.; Svrcek, M.; Cohen, R.; Basile, D.; Tougeron, D.; Phelip, J.-M. Deficient mismatch repair/microsatellite unstable colorectal cancer: Diagnosis, prognosis and treatment. Eur. J. Cancer 2022, 175, 136–157. [Google Scholar] [CrossRef]
  35. Zhu, J.; Lian, J.; Xu, B.; Pang, X.; Ji, S.; Zhao, Y.; Lu, H. Neoadjuvant immunotherapy for colorectal cancer: Right regimens, right patients, right directions? Front. Immunol. 2023, 14, 1120684. [Google Scholar] [CrossRef]
  36. Krasteva, N.; Georgieva, M. Promising Therapeutic Strategies for Colorectal Cancer Treatment Based on Nanomaterials. Pharmaceutics 2022, 14, 1213. [Google Scholar] [CrossRef]
  37. Deshmukh, R.; Prajapati, M.; Harwansh, R.K. A review on emerging targeted therapies for the management of metastatic colorectal cancers. Med. Oncol. 2023, 40, 159. [Google Scholar] [CrossRef]
  38. Ben Hamouda, S.; Essafi-Benkhadir, K. Interplay between Signaling Pathways and Tumor Microenvironment Components: A Paradoxical Role in Colorectal Cancer. Int. J. Mol. Sci. 2023, 24, 5600. [Google Scholar] [CrossRef]
  39. Parab, T.M.; DeRogatis, M.J.; Boaz, A.M.; Grasso, S.A.; Issack, P.S.; Duarte, D.A.; Urayeneza, O.; Vahdat, S.; Qiao, J.-H.; Hinika, G.S. Gastrointestinal stromal tumors: A comprehensive review. J. Gastrointest. Oncol. 2019, 10, 144–154. [Google Scholar] [CrossRef]
  40. Von Mehren, M.; Joensuu, H. Gastrointestinal Stromal Tumors. J. Clin. Oncol. 2018, 36, 136–143. [Google Scholar] [CrossRef]
  41. Søreide, K.; Sandvik, O.M.; Søreide, J.A.; Giljaca, V.; Jureckova, A.; Bulusu, V.R. Global epidemiology of gastrointestinal stromal tumours (GIST): A systematic review of population-based cohort studies. Cancer Epidemiol. 2016, 40, 39–46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Dudzisz-Śledź, M.; Klimczak, A.; Bylina, E.; Rutkowski, P. Treatment of Gastrointestinal Stromal Tumors (GISTs): A Focus on Younger Patients. Cancers 2022, 14, 2831. [Google Scholar] [CrossRef] [PubMed]
  43. Min, K.W.; Leabu, M. Interstitial Cells of Cajal (ICC) and Gastrointestinal Stromal Tumor (GIST): Facts, speculations, and myths. J. Cell. Mol. Med. 2006, 10, 995–1013. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Radu, P.; Zurzu, M.; Paic, V.; Bratucu, M.; Garofil, D.; Tigora, A.; Georgescu, V.; Prunoiu, V.; Popa, F.; Surlin, V.; et al. Interstitial Cells of Cajal—Origin, Distribution and Relationship with Gastrointestinal Tumors. Medicina 2022, 59, 63. [Google Scholar] [CrossRef]
  45. Sun, Y.; Yue, L.; Xu, P.; Hu, W. An overview of agents and treatments for PDGFRA-mutated gastrointestinal stromal tumors. Front. Oncol. 2022, 12, 927587. [Google Scholar] [CrossRef]
  46. Serrano, C.; George, S. Gastrointestinal Stromal Tumor: Challenges and Opportunities for a New Decade. Clin. Cancer Res. 2020, 26, 5078–5085. [Google Scholar] [CrossRef]
  47. Fudalej, M.M.; Badowska-Kozakiewicz, A.M. Improved understanding of gastrointestinal stromal tumors biology as a step for developing new diagnostic and therapeutic schemes. Oncol. Lett. 2021, 21, 417. [Google Scholar] [CrossRef]
  48. Wang, M.; Zhao, J.; Zhang, L.; Wei, F.; Lian, Y.; Wu, Y.; Gong, Z.; Zhang, S.; Zhou, J.; Cao, K.; et al. Role of tumor microenvironment in tumorigenesis. J. Cancer 2017, 8, 761–773. [Google Scholar] [CrossRef] [Green Version]
  49. Neophytou, C.M.; Panagi, M.; Stylianopoulos, T.; Papageorgis, P. The Role of Tumor Microenvironment in Cancer Metastasis: Molecular Mechanisms and Therapeutic Opportunities. Cancers 2021, 13, 2053. [Google Scholar] [CrossRef]
  50. Belli, C.; Trapani, D.; Viale, G.; D’Amico, P.; Duso, B.A.; Della Vigna, P.; Orsi, F.; Curigliano, G. Targeting the microenvironment in solid tumors. Cancer Treat. Rev. 2018, 65, 22–32. [Google Scholar] [CrossRef]
  51. Baghban, R.; Roshangar, L.; Jahanban-Esfahlan, R.; Seidi, K.; Ebrahimi-Kalan, A.; Jaymand, M.; Kolahian, S.; Javaheri, T.; Zare, P. Tumor microenvironment complexity and therapeutic implications at a glance. Cell Commun. Signal. 2020, 18, 59. [Google Scholar] [CrossRef] [Green Version]
  52. Khalaf, K.; Hana, D.; Chou, J.T.-T.; Singh, C.; Mackiewicz, A.; Kaczmarek, M. Aspects of the Tumor Microenvironment Involved in Immune Resistance and Drug Resistance. Front. Immunol. 2021, 12, 656364. [Google Scholar] [CrossRef]
  53. Melissari, M.-T.; Chalkidi, N.; Sarris, M.E.; Koliaraki, V. Fibroblast Reprogramming in Gastrointestinal Cancer. Front. Cell Dev. Biol. 2020, 8, 630. [Google Scholar] [CrossRef]
  54. Wang, Y.; Gao, P.; Hao, Z.; Chen, L.; Li, X.; Jiao, Y.; Liu, J.; Li, J.; Zhang, Y.; Peng, X.; et al. The effect of neoadjuvant chemotherapy on the tumor immune microenvironment in gastrointestinal tumors. Front. Oncol. 2022, 12, 1054598. [Google Scholar] [CrossRef]
  55. Ham, I.-H.; Lee, D.; Hur, H. Cancer-Associated Fibroblast-Induced Resistance to Chemotherapy and Radiotherapy in Gastrointestinal Cancers. Cancers 2021, 13, 1172. [Google Scholar] [CrossRef]
  56. Yang, P.; Zhang, D.; Wang, T.; Ji, J.; Jin, C.; Peng, C.; Tan, Y.; Zhou, J.; Wang, L.; Feng, Y.; et al. CAF-derived exosomal WEE2-AS1 facilitates colorectal cancer progression via promoting degradation of MOB1A to inhibit the Hippo pathway. Cell Death Dis. 2022, 13, 796. [Google Scholar] [CrossRef]
  57. Kalluri, R. The biology and function of fibroblasts in cancer. Nat. Rev. Cancer 2016, 16, 582–598. [Google Scholar] [CrossRef]
  58. Zonouzi, S.K.; Pezeshki, P.S.; Razi, S.; Rezaei, N. Cancer-associated fibroblasts in colorectal cancer. Clin. Transl. Oncol. 2022, 24, 757–769. [Google Scholar] [CrossRef]
  59. Zulaziz, N.; Chai, S.J.; Lim, K.P. The origins, roles and therapies of cancer associated fibroblast in liver cancer. Front. Oncol. 2023, 13, 1151373. [Google Scholar] [CrossRef]
  60. Joshi, R.S.; Kanugula, S.S.; Sudhir, S.; Pereira, M.P.; Jain, S.; Aghi, M.K. The Role of Cancer-Associated Fibroblasts in Tumor Progression. Cancers 2021, 13, 1399. [Google Scholar] [CrossRef]
  61. Sun, H.; Wang, X.; Wang, X.; Xu, M.; Sheng, W. The role of cancer-associated fibroblasts in tumorigenesis of gastric cancer. Cell Death Dis. 2022, 13, 874. [Google Scholar] [CrossRef] [PubMed]
  62. Wu, X.; Tao, P.; Zhou, Q.; Li, J.; Yu, Z.; Wang, X.; Li, J.; Li, C.; Yan, M.; Zhu, Z.; et al. IL-6 secreted by cancer-associated fibroblasts promotes epithelial-mesenchymal transition and metastasis of gastric cancer via JAK2/STAT3 signaling pathway. Oncotarget 2017, 8, 20741–20750. [Google Scholar] [CrossRef] [Green Version]
  63. Montori, M.; Scorzoni, C.; Argenziano, M.E.; Balducci, D.; De Blasio, F.; Martini, F.; Buono, T.; Benedetti, A.; Marzioni, M.; Maroni, L. Cancer-Associated Fibroblasts in Cholangiocarcinoma: Current Knowledge and Possible Implications for Therapy. J. Clin. Med. 2022, 11, 6498. [Google Scholar] [CrossRef] [PubMed]
  64. Ishimoto, T.; Miyake, K.; Nandi, T.; Yashiro, M.; Onishi, N.; Huang, K.K.; Lin, S.J.; Kalpana, R.; Tay, S.T.; Suzuki, Y.; et al. Activation of Transforming Growth Factor Beta 1 Signaling in Gastric Cancer-associated Fibroblasts Increases Their Motility, via Expression of Rhomboid 5 Homolog 2, and Ability to Induce Invasiveness of Gastric Cancer Cells. Gastroenterology 2017, 153, 191–204.e16. [Google Scholar] [CrossRef] [PubMed]
  65. Jiang, H.; Ge, H.; Shi, Y.; Yuan, F.; Yue, H. CAFs secrete CXCL12 to accelerate the progression and cisplatin resistance of colorectal cancer through promoting M2 polarization of macrophages. Med. Oncol. 2023, 40, 90. [Google Scholar] [CrossRef] [PubMed]
  66. Mao, X.; Xu, J.; Wang, W.; Liang, C.; Hua, J.; Liu, J.; Zhang, B.; Meng, Q.; Yu, X.; Shi, S. Crosstalk between cancer-associated fibroblasts and immune cells in the tumor microenvironment: New findings and future perspectives. Mol. Cancer 2021, 20, 131. [Google Scholar] [CrossRef]
  67. Zhang, A.; Qian, Y.; Ye, Z.; Chen, H.; Xie, H.; Zhou, L.; Shen, Y.; Zheng, S. Cancer-associated fibroblasts promote M2 polarization of macrophages in pancreatic ductal adenocarcinoma. Cancer Med. 2017, 6, 463–470. [Google Scholar] [CrossRef]
  68. Kinoshita, T.; Ishii, G.; Hiraoka, N.; Hirayama, S.; Yamauchi, C.; Aokage, K.; Hishida, T.; Yoshida, J.; Nagai, K.; Ochiai, A. Forkhead box P3 regulatory T cells coexisting with cancer associated fibroblasts are correlated with a poor outcome in lung adenocarcinoma. Cancer Sci. 2013, 104, 409–415. [Google Scholar] [CrossRef]
  69. Chun, E.; Lavoie, S.; Michaud, M.; Gallini, C.A.; Kim, J.; Soucy, G.; Odze, R.; Glickman, J.N.; Garrett, W.S. CCL2 Promotes Colorectal Carcinogenesis by Enhancing Polymorphonuclear Myeloid-Derived Suppressor Cell Population and Function. Cell Rep. 2015, 12, 244–257. [Google Scholar] [CrossRef] [Green Version]
  70. Linares, J.; Sallent-Aragay, A.; Badia-Ramentol, J.; Recort-Bascuas, A.; Méndez, A.; Manero-Rupérez, N.; Re, D.L.; Rivas, E.I.; Guiu, M.; Zwick, M.; et al. Long-term platinum-based drug accumulation in cancer-associated fibroblasts promotes colorectal cancer progression and resistance to therapy. Nat. Commun. 2023, 14, 746. [Google Scholar] [CrossRef]
  71. Lotti, F.; Jarrar, A.M.; Pai, R.K.; Hitomi, M.; Lathia, J.; Mace, A.; Gantt, G.A., Jr.; Sukhdeo, K.; DeVecchio, J.; Vasanji, A.; et al. Chemotherapy activates cancer-associated fibroblasts to maintain colorectal cancer-initiating cells by IL-17A. J. Exp. Med. 2013, 210, 2851–2872. [Google Scholar] [CrossRef] [Green Version]
  72. Caligiuri, A.; Pastore, M.; Lori, G.; Raggi, C.; Di Maira, G.; Marra, F.; Gentilini, A. Role of Chemokines in the Biology of Cholangiocarcinoma. Cancers 2020, 12, 2215. [Google Scholar] [CrossRef]
  73. Zhong, B.; Cheng, B.; Huang, X.; Xiao, Q.; Niu, Z.; Chen, Y.-F.; Yu, Q.; Wang, W.; Wu, X.-J. Colorectal cancer-associated fibroblasts promote metastasis by up-regulating LRG1 through stromal IL-6/STAT3 signaling. Cell Death Dis. 2021, 13, 16. [Google Scholar] [CrossRef]
  74. Xu, X.; Yang, C.; Chen, J.; Liu, J.; Li, P.; Shi, Y.; Yu, P. Interleukin-23 promotes the migration and invasion of gastric cancer cells by inducing epithelial-to-mesenchymal transition via the STAT3 pathway. Biochem. Biophys. Res. Commun. 2018, 499, 273–278. [Google Scholar] [CrossRef]
  75. Bandapalli, O.R.; Ehrmann, F.; Ehemann, V.; Gaida, M.; Macher-Goeppinger, S.; Wente, M.; Schirmacher, P.; Brand, K. Down-regulation of CXCL1 inhibits tumor growth in colorectal liver metastasis. Cytokine 2012, 57, 46–53. [Google Scholar] [CrossRef]
  76. Zhuo, C.; Ruan, Q.; Zhao, X.; Shen, Y.; Lin, R. CXCL1 promotes colon cancer progression through activation of NF-κB/P300 signaling pathway. Biol. Direct 2022, 17, 34. [Google Scholar] [CrossRef]
  77. Mao, Z.; Zhang, J.; Shi, Y.; Li, W.; Shi, H.; Ji, R.; Mao, F.; Qian, H.; Xu, W.; Zhang, X. CXCL5 promotes gastric cancer metastasis by inducing epithelial-mesenchymal transition and activating neutrophils. Oncogenesis 2020, 9, 63. [Google Scholar] [CrossRef]
  78. Zhao, J.; Ou, B.; Han, D.; Wang, P.; Zong, Y.; Zhu, C.; Di Liu, D.; Zheng, M.; Sun, J.; Feng, H.; et al. Tumor-derived CXCL5 promotes human colorectal cancer metastasis through activation of the ERK/Elk-1/Snail and AKT/GSK3β/β-catenin pathways. Mol. Cancer 2017, 16, 70. [Google Scholar] [CrossRef] [Green Version]
  79. Daniel, S.K.; Seo, Y.D.; Pillarisetty, V.G. The CXCL12-CXCR4/CXCR7 axis as a mechanism of immune resistance in gastrointestinal malignancies. Semin. Cancer Biol. 2020, 65, 176–188. [Google Scholar] [CrossRef]
  80. Wang, D.; Wang, X.; Song, Y.; Si, M.; Sun, Y.; Liu, X.; Cui, S.; Qu, X.; Yu, X. Exosomal miR-146a-5p and miR-155-5p promote CXCL12/CXCR7-induced metastasis of colorectal cancer by crosstalk with cancer-associated fibroblasts. Cell Death Dis. 2022, 13, 380. [Google Scholar] [CrossRef]
  81. Goïta, A.A.; Guenot, D. Colorectal Cancer: The Contribution of CXCL12 and Its Receptors CXCR4 and CXCR7. Cancers 2022, 14, 1810. [Google Scholar] [CrossRef] [PubMed]
  82. Xiong, X.; Liao, X.; Qiu, S.; Xu, H.; Zhang, S.; Wang, S.; Ai, J.; Yang, L. CXCL8 in Tumor Biology and Its Implications for Clinical Translation. Front. Mol. Biosci. 2022, 9, 723846. [Google Scholar] [CrossRef] [PubMed]
  83. Ali, S.; Rehman, M.U.; Yatoo, A.M.; Arafah, A.; Khan, A.; Rashid, S.; Majid, S.; Ali, A.; Ali, N. TGF-β signaling pathway: Therapeutic targeting and potential for anti-cancer immunity. Eur. J. Pharmacol. 2023, 947, 175678. [Google Scholar] [CrossRef]
  84. Cao, P.; Sun, Z.; Zhang, F.; Zhang, J.; Zheng, X.; Yu, B.; Zhao, Y.; Wang, W. TGF-β Enhances Immunosuppression of Myeloid-Derived Suppressor Cells to Induce Transplant Immune Tolerance Through Affecting Arg-1 Expression. Front. Immunol. 2022, 13, 919674. [Google Scholar] [CrossRef] [PubMed]
  85. Sabbadini, F.; Bertolini, M.; De Matteis, S.; Mangiameli, D.; Contarelli, S.; Pietrobono, S.; Melisi, D. The Multifaceted Role of TGF-β in Gastrointestinal Tumors. Cancers 2021, 13, 3960. [Google Scholar] [CrossRef] [PubMed]
  86. Lucarini, V.; Melaiu, O.; D’amico, S.; Pastorino, F.; Tempora, P.; Scarsella, M.; Pezzullo, M.; De Ninno, A.; D’oria, V.; Cilli, M.; et al. Combined mitoxantrone and anti-TGFβ treatment with PD-1 blockade enhances antitumor immunity by remodelling the tumor immune landscape in neuroblastoma. J. Exp. Clin. Cancer Res. 2022, 41, 326. [Google Scholar] [CrossRef]
  87. Qin, G.; Luo, M.; Chen, J.; Dang, Y.; Chen, G.; Li, L.; Zeng, J.; Lu, Y.; Yang, J. Reciprocal activation between MMP-8 and TGF-β1 stimulates EMT and malignant progression of hepatocellular carcinoma. Cancer Lett. 2016, 374, 85–95. [Google Scholar] [CrossRef]
  88. Cevenini, A.; Orrù, S.; Mancini, A.; Alfieri, A.; Buono, P.; Imperlini, E. Molecular Signatures of the Insulin-like Growth Factor 1-mediated Epithelial-Mesenchymal Transition in Breast, Lung and Gastric Cancers. Int. J. Mol. Sci. 2018, 19, 2411. [Google Scholar] [CrossRef] [Green Version]
  89. Jin, M.-Z.; Jin, W.-L. The updated landscape of tumor microenvironment and drug repurposing. Signal Transduct. Target. Ther. 2020, 5, 166. [Google Scholar] [CrossRef]
  90. Galon, J.; Costes, A.; Sanchez-Cabo, F.; Kirilovsky, A.; Mlecnik, B.; Lagorce-Pagès, C.; Tosolini, M.; Camus, M.; Berger, A.; Wind, P.; et al. Type, Density, and Location of Immune Cells Within Human Colorectal Tumors Predict Clinical Outcome. Science 2006, 313, 1960–1964. [Google Scholar] [CrossRef] [Green Version]
  91. Chen, L.; Huang, H.; Huang, Z.; Chen, J.; Liu, Y.; Wu, Y.; Li, A.; Ge, J.; Fang, Z.; Xu, B.; et al. Prognostic values of tissue-resident CD8+T cells in human hepatocellular carcinoma and intrahepatic cholangiocarcinoma. World J. Surg. Oncol. 2023, 21, 124. [Google Scholar] [CrossRef] [PubMed]
  92. Geng, Y.; Feng, J.; Huang, H.; Wang, Y.; Yi, X.; Wei, S.; Zhang, M.; Li, Z.; Wang, W.; Hu, W. Single-cell transcriptome analysis of tumor immune microenvironment characteristics in colorectal cancer liver metastasis. Ann. Transl. Med. 2022, 10, 1170. [Google Scholar] [CrossRef] [PubMed]
  93. Vita, F.; Olaizola, I.; Amato, F.; Rae, C.; Marco, S.; Banales, J.M.; Braconi, C. Heterogeneity of Cholangiocarcinoma Immune Biology. Cells 2023, 12, 846. [Google Scholar] [CrossRef] [PubMed]
  94. Väyrynen, J.P.; Haruki, K.; Väyrynen, S.A.; Lau, M.C.; Costa, A.D.; Borowsky, J.; Zhao, M.; Ugai, T.; Kishikawa, J.; Akimoto, N.; et al. Prognostic significance of myeloid immune cells and their spatial distribution in the colorectal cancer microenvironment. J. Immunother. Cancer 2021, 9, e002297. [Google Scholar] [CrossRef] [PubMed]
  95. Han, B.; Fang, T.; Zhang, Y.; Zhang, Y.; Gao, J.; Xue, Y. Association of the TGFβ gene family with microenvironmental features of gastric cancer and prediction of response to immunotherapy. Front. Oncol. 2022, 12, 920599. [Google Scholar] [CrossRef] [PubMed]
  96. Farshidpour, M.; Ahmed, M.; Junna, S.; Merchant, J.L. Myeloid-derived suppressor cells in gastrointestinal cancers: A systemic review. World J. Gastrointest. Oncol. 2021, 13, 1–11. [Google Scholar] [CrossRef]
  97. Qian, B.-Z.; Pollard, J.W. Macrophage diversity enhances tumor progression and metastasis. Cell 2010, 141, 39–51. [Google Scholar] [CrossRef] [Green Version]
  98. Mantovani, A.; Sica, A.; Sozzani, S.; Allavena, P.; Vecchi, A.; Locati, M. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 2004, 25, 677–686. [Google Scholar] [CrossRef]
  99. Wang, H.; Tian, T.; Zhang, J. Tumor-Associated Macrophages (TAMs) in Colorectal Cancer (CRC): From Mechanism to Therapy and Prognosis. Int. J. Mol. Sci. 2021, 22, 8470. [Google Scholar] [CrossRef]
  100. Gambardella, V.; Castillo, J.; Tarazona, N.; Gimeno-Valiente, F.; Martínez-Ciarpaglini, C.; Cabeza-Segura, M.; Roselló, S.; Roda, D.; Huerta, M.; Cervantes, A.; et al. The role of tumor-associated macrophages in gastric cancer development and their potential as a therapeutic target. Cancer Treat. Rev. 2020, 86, 102015. [Google Scholar] [CrossRef] [Green Version]
  101. Dimino, A.; Brando, C.; Algeri, L.; Gristina, V.; Pedone, E.; Peri, M.; Perez, A.; De Luca, I.; Sciacchitano, R.; Magrin, L.; et al. Exploring the Dynamic Crosstalk between the Immune System and Genetics in Gastrointestinal Stromal Tumors. Cancers 2022, 15, 216. [Google Scholar] [CrossRef]
  102. Cui, C.; Lan, P.; Fu, L. The role of myeloid-derived suppressor cells in gastrointestinal cancer. Cancer Commun. 2021, 41, 442–471. [Google Scholar] [CrossRef]
  103. Tang, Y.; Zhou, C.; Li, Q.; Cheng, X.; Huang, T.; Li, F.; He, L.; Zhang, B.; Tu, S. Targeting depletion of myeloid-derived suppressor cells potentiates PD-L1 blockade efficacy in gastric and colon cancers. Oncoimmunology 2022, 11, 2131084. [Google Scholar] [CrossRef]
  104. Yaseen, M.M.; Abuharfeil, N.M.; Darmani, H.; Daoud, A. Mechanisms of immune suppression by myeloid-derived suppressor cells: The role of interleukin-10 as a key immunoregulatory cytokine. Open Biol. 2020, 10, 200111. [Google Scholar] [CrossRef]
  105. Yang, Y.; Li, C.; Liu, T.; Dai, X.; Bazhin, A.V. Myeloid-Derived Suppressor Cells in Tumors: From Mechanisms to Antigen Specificity and Microenvironmental Regulation. Front. Immunol. 2020, 11, 1371. [Google Scholar] [CrossRef]
  106. Umansky, V.; Blattner, C.; Gebhardt, C.; Utikal, J. The Role of Myeloid-Derived Suppressor Cells (MDSC) in Cancer Progression. Vaccines 2016, 4, 36. [Google Scholar] [CrossRef] [Green Version]
  107. Zheng, W.; Wu, J.; Peng, Y.; Sun, J.; Cheng, P.; Huang, Q. Tumor-Associated Neutrophils in Colorectal Cancer Development, Progression and Immunotherapy. Cancers 2022, 14, 4755. [Google Scholar] [CrossRef]
  108. Zhang, Y.; Song, J.; Zhang, Y.; Li, T.; Peng, J.; Zhou, H.; Zong, Z. Emerging Role of Neutrophil Extracellular Traps in Gastrointestinal Tumors: A Narrative Review. Int. J. Mol. Sci. 2022, 24, 334. [Google Scholar] [CrossRef]
  109. Zhou, S.-L.; Zhou, Z.-J.; Hu, Z.-Q.; Huang, X.-W.; Wang, Z.; Chen, E.-B.; Fan, J.; Cao, Y.; Dai, Z.; Zhou, J. Tumor-Associated Neutrophils Recruit Macrophages and T-Regulatory Cells to Promote Progression of Hepatocellular Carcinoma and Resistance to Sorafenib. Gastroenterology 2016, 150, 1646–1658.e17. [Google Scholar] [CrossRef] [Green Version]
  110. Olguín, J.E.; Medina-Andrade, I.; Rodríguez, T.; Rodríguez-Sosa, M.; Terrazas, L.I. Relevance of Regulatory T Cells during Colorectal Cancer Development. Cancers 2020, 12, 1888. [Google Scholar] [CrossRef]
  111. Wang, B.; Zhang, Z.; Liu, W.; Tan, B. Targeting regulatory T cells in gastric cancer: Pathogenesis, immunotherapy, and prognosis. Biomed. Pharmacother. 2023, 158, 114180. [Google Scholar] [CrossRef] [PubMed]
  112. Zhang, L.; Yu, X.; Zheng, L.; Zhang, Y.; Li, Y.; Fang, Q.; Gao, R.; Kang, B.; Zhang, Q.; Huang, J.Y.; et al. Lineage tracking reveals dynamic relationships of T cells in colorectal cancer. Nature 2018, 564, 268–272. [Google Scholar] [CrossRef] [PubMed]
  113. Ahrends, T.; Borst, J. The opposing roles of CD4+T cells in anti-tumour immunity. Immunology 2018, 154, 582–592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Revilla, S.A.; Kranenburg, O.; Coffer, P.J. Colorectal Cancer-Infiltrating Regulatory T Cells: Functional Heterogeneity, Metabolic Adaptation, and Therapeutic Targeting. Front. Immunol. 2022, 13, 903564. [Google Scholar] [CrossRef]
  115. Ali Syeda, Z.; Langden, S.S.S.; Munkhzul, C.; Lee, M.; Song, S.J. Regulatory Mechanism of MicroRNA Expression in Cancer. Int. J. Mol. Sci. 2020, 21, 1723. [Google Scholar] [CrossRef] [Green Version]
  116. Zaki, M.B.; Abulsoud, A.I.; Elshaer, S.S.; Fathi, D.; Abdelmaksoud, N.M.; El-Mahdy, H.A.; Ismail, A.; Elsakka, E.G.; Sallam, A.-A.M.; Doghish, A.S. The interplay of signaling pathways with miRNAs in cholangiocarcinoma pathogenicity and targeted therapy. Pathol. Res. Pract. 2023, 245, 154437. [Google Scholar] [CrossRef]
  117. Ventura, A.; Jacks, T. MicroRNAs and cancer: Short RNAs go a long way. Cell 2009, 136, 586–591. [Google Scholar] [CrossRef] [Green Version]
  118. 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]
  119. Gramantieri, L.; Ferracin, M.; Fornari, F.; Veronese, A.; Sabbioni, S.; Liu, C.-G.; Calin, G.A.; Giovannini, C.; Ferrazzi, E.; Grazi, G.L.; et al. Cyclin G1 Is a Target of miR-122a, a MicroRNA frequently down-regulated in human hepatocellular carcinoma. Cancer Res. 2007, 67, 6092–6099. [Google Scholar] [CrossRef] [Green Version]
  120. Zhang, H.-H.; Wang, X.-J.; Li, G.-X.; Yang, E.; Yang, N.-M. Detection of let-7a microRNA by real-time PCR in gastric carcinoma. World J. Gastroenterol. 2007, 13, 2883–2888. [Google Scholar] [CrossRef]
  121. Kutay, H.; Bai, S.; Datta, J.; Motiwala, T.; Pogribny, I.; Frankel, W.; Jacob, S.T.; Ghoshal, K. Downregulation of miR-122 in the rodent and human hepatocellular carcinomas. J. Cell. Biochem. 2006, 99, 671–678. [Google Scholar] [CrossRef] [Green Version]
  122. Xie, Y.; Zhang, H.; Guo, X.-J.; Feng, Y.-C.; He, R.-Z.; Li, X.; Yu, S.; Zhao, Y.; Shen, M.; Zhu, F.; et al. Let-7c inhibits cholangiocarcinoma growth but promotes tumor cell invasion and growth at extrahepatic sites. Cell Death Dis. 2018, 9, 249. [Google Scholar] [CrossRef] [Green Version]
  123. Sun, Z.; Han, Q.; Zhou, N.; Wang, S.; Lu, S.; Bai, C.; Zhao, R.C. MicroRNA-9 enhances migration and invasion through KLF17 in hepatocellular carcinoma. Mol. Oncol. 2013, 7, 884–894. [Google Scholar] [CrossRef]
  124. Shirmohammadi, K.; Sohrabi, S.; Samani, Z.J.; Effatpanah, H.; Yadegarazari, R.; Saidijam, M. Evaluation of altered expression of miR-9 and miR-106a as an early diagnostic approach in gastric cancer. J. Gastrointest. Oncol. 2018, 9, 46–51. [Google Scholar] [CrossRef] [Green Version]
  125. Gao, L.; Yang, X.; Zhang, H.; Yu, M.; Long, J.; Yang, T. Inhibition of miR-10a-5p suppresses cholangiocarcinoma cell growth through downregulation of Akt pathway. OncoTargets Ther. 2018, 11, 6981–6994. [Google Scholar] [CrossRef] [Green Version]
  126. Asangani, I.A.; Rasheed, S.A.K.; Nikolova, D.A.; Leupold, J.H.; Colburn, N.H.; Post, S.; Allgayer, H. MicroRNA-21 (miR-21) post-transcriptionally downregulates tumor suppressor Pdcd4 and stimulates invasion, intravasation and metastasis in colorectal cancer. Oncogene 2008, 27, 2128–2136. [Google Scholar] [CrossRef] [Green Version]
  127. Li, Q.; Zhang, D.; Wang, Y.; Sun, P.; Hou, X.; Larner, J.; Xiong, W.; Mi, J. MiR-21/Smad 7 signaling determines TGF-β1-induced CAF formation. Sci. Rep. 2013, 3, 2038. [Google Scholar] [CrossRef] [Green Version]
  128. Meng, F.; Henson, R.; Lang, M.; Wehbe, H.; Maheshwari, S.; Mendell, J.T.; Jiang, J.; Schmittgen, T.D.; Patel, T. Involvement of Human Micro-RNA in growth and response to chemotherapy in human cholangiocarcinoma cell lines. Gastroenterology 2006, 130, 2113–2129. [Google Scholar] [CrossRef] [Green Version]
  129. Zuo, H.; Liu, S.; Li, X.; Hou, G. miR-23a-3p promotes the development of colon cancer by inhibiting the expression of NDRG4. Clin. Transl. Oncol. 2023, 25, 933–940. [Google Scholar] [CrossRef]
  130. Wang, J.; Guan, X.; Zhang, Y.; Ge, S.; Zhang, L.; Li, H.; Wang, X.; Liu, R.; Ning, T.; Deng, T.; et al. Exosomal miR-27a Derived from Gastric Cancer Cells Regulates the Transformation of Fibroblasts into Cancer-Associated Fibroblasts. Cell. Physiol. Biochem. 2018, 49, 869–883. [Google Scholar] [CrossRef]
  131. Mott, J.L.; Kobayashi, S.; Bronk, S.F.; Gores, G.J. mir-29 regulates Mcl-1 protein expression and apoptosis. Oncogene 2007, 26, 6133–6140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Bandrés, E.; Cubedo, E.; Agirre, X.; Malumbres, R.; Zárate, R.; Ramirez, N.; Abajo, A.; Navarro, A.; Moreno, I.; Monzó, M.; et al. Identification by Real-time PCR of 13 mature microRNAs differentially expressed in colorectal cancer and non-tumoral tissues. Mol. Cancer 2006, 5, 29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Wang, S.-J.; Cao, Y.-F.; Yang, Z.-Q.; Jiang, Z.-Y.; Cai, B.; Guo, J.; Zhang, S.; Zhang, X.-L.; Gao, F. MicroRNA-93-5p increases multidrug resistance in human colorectal carcinoma cells by downregulating cyclin dependent kinase inhibitor 1A gene expression. Oncol. Lett. 2016, 13, 722–730. [Google Scholar] [CrossRef] [Green Version]
  134. Sun, Z.; Zhang, Q.; Yuan, W.; Li, X.; Chen, C.; Guo, Y.; Shao, B.; Dang, Q.; Zhou, Q.; Wang, Q.; et al. MiR-103a-3p promotes tumour glycolysis in colorectal cancer via hippo/YAP1/HIF1A axis. J. Exp. Clin. Cancer Res. 2020, 39, 250. [Google Scholar] [CrossRef] [PubMed]
  135. Zhu, X.; Jin, X.; Li, Z.; Chen, X.; Zhao, J. miR-152–3p facilitates cell adhesion and hepatic metastases in colorectal cancer via targeting AQP11. Pathol. Res. Pract. 2023, 244, 154389. [Google Scholar] [CrossRef]
  136. Murakami, Y.; Yasuda, T.; Saigo, K.; Urashima, T.; Toyoda, H.; Okanoue, T.; Shimotohno, K. Comprehensive analysis of microRNA expression patterns in hepatocellular carcinoma and non-tumorous tissues. Oncogene 2006, 25, 2537–2545. [Google Scholar] [CrossRef]
  137. Li, Z.-Y.; Xie, Y.; Deng, M.; Zhu, L.; Wu, X.; Li, G.; Shi, N.-X.; Wen, C.; Huang, W.; Duan, Y.; et al. c-Myc-activated intronic miR-210 and lncRNA MIR210HG synergistically promote the metastasis of gastric cancer. Cancer Lett. 2022, 526, 322–334. [Google Scholar] [CrossRef]
  138. Shayimu, P.; Wang, J.B.; Liu, L.; Tuerdi, R.; Yu, C.G.; Yusufu, A. miR-922 regulates apoptosis, migration, and invasion by targeting SOCS1 in gastric cancer. Kaohsiung J. Med. Sci. 2020, 36, 178–185. [Google Scholar] [CrossRef]
  139. Santos, P.; Almeida, F. Role of Exosomal miRNAs and the Tumor Microenvironment in Drug Resistance. Cells 2020, 9, 1450. [Google Scholar] [CrossRef]
  140. Wei, C.; Yang, C.; Wang, S.; Shi, D.; Zhang, C.; Lin, X.; Liu, Q.; Dou, R.; Xiong, B. Crosstalk between cancer cells and tumor associated macrophages is required for mesenchymal circulating tumor cell-mediated colorectal cancer metastasis. Mol. Cancer 2019, 18, 64. [Google Scholar] [CrossRef]
  141. Toshida, K.; Itoh, S.; Harada, N.; Morinaga, A.; Yugawa, K.; Tomiyama, T.; Kosai-Fujimoto, Y.; Tomino, T.; Kurihara, T.; Nagao, Y.; et al. Cancer-associated fibroblasts promote tumor cell growth via miR-493-5p in intrahepatic cholangiocarcinoma. Cancer Sci. 2022, 114, 937–947. [Google Scholar] [CrossRef] [PubMed]
  142. Zhou, Y.; Ren, H.; Dai, B.; Li, J.; Shang, L.; Huang, J.; Shi, X. Hepatocellular carcinoma-derived exosomal miRNA-21 contributes to tumor progression by converting hepatocyte stellate cells to cancer-associated fibroblasts. J. Exp. Clin. Cancer Res. 2018, 37, 324. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Fang, T.; Lv, H.; Lv, G.; Li, T.; Wang, C.; Han, Q.; Yu, L.; Su, B.; Guo, L.; Huang, S.; et al. Tumor-derived exosomal miR-1247-3p induces cancer-associated fibroblast activation to foster lung metastasis of liver cancer. Nat. Commun. 2018, 9, 191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Yang, T.-S.; Yang, X.-H.; Chen, X.; Wang, X.-D.; Hua, J.; Zhou, D.-L.; Zhou, B.; Song, Z.-S. MicroRNA-106b in cancer-associated fibroblasts from gastric cancer promotes cell migration and invasion by targeting PTEN. FEBS Lett. 2014, 588, 2162–2169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Zhang, H.; Deng, T.; Liu, R.; Ning, T.; Yang, H.; Liu, D.; Zhang, Q.; Lin, D.; Ge, S.; Bai, M.; et al. CAF secreted miR-522 suppresses ferroptosis and promotes acquired chemo-resistance in gastric cancer. Mol. Cancer 2020, 19, 43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Jiang, Y.; Qiu, Q.; Jing, X.; Song, Z.; Zhang, Y.; Wang, C.; Liu, K.; Ye, F.; Ji, X.; Luo, F.; et al. Cancer-associated fibroblast-derived exosome miR-181b-3p promotes the occurrence and development of colorectal cancer by regulating SNX2 expression. Biochem. Biophys. Res. Commun. 2023, 641, 177–185. [Google Scholar] [CrossRef]
  147. Liu, K.; Dou, R.; Yang, C.; Di, Z.; Shi, D.; Zhang, C.; Song, J.; Fang, Y.; Huang, S.; Xiang, Z.; et al. Exosome-transmitted miR-29a induces colorectal cancer metastasis by destroying the vascular endothelial barrier. Carcinogenesis, 2023; online ahead of print. [Google Scholar] [CrossRef]
  148. Zheng, P.; Chen, L.; Yuan, X.; Luo, Q.; Liu, Y.; Xie, G.; Ma, Y.; Shen, L. Exosomal transfer of tumor-associated macrophage-derived miR-21 confers cisplatin resistance in gastric cancer cells. J. Exp. Clin. Cancer Res. 2017, 36, 53. [Google Scholar] [CrossRef] [Green Version]
  149. Song, J.; Lin, Z.; Liu, Q.; Huang, S.; Han, L.; Fang, Y.; Zhong, P.; Dou, R.; Xiang, Z.; Zheng, J.; et al. MiR-192-5p/RB1/NF-κBp65 signaling axis promotes IL-10 secretion during gastric cancer EMT to induce Treg cell differentiation in the tumour microenvironment. Clin. Transl. Med. 2022, 12, e992. [Google Scholar] [CrossRef]
  150. Bandres, E.; Bitarte, N.; Arias, F.; Agorreta, J.; Fortes, P.; Agirre, X.; Zarate, R.; Diaz-Gonzalez, J.A.; Ramirez, N.; Sola, J.J.; et al. microRNA-451 regulates macrophage migration inhibitory factor production and proliferation of gastrointestinal cancer cells. Clin. Cancer Res. 2009, 15, 2281–2290. [Google Scholar] [CrossRef] [Green Version]
  151. Tsai, M.-M.; Wang, C.-S.; Tsai, C.-Y.; Huang, H.-W.; Chi, H.-C.; Lin, Y.-H.; Lu, P.-H.; Lin, K.-H. Potential Diagnostic, Prognostic and Therapeutic Targets of MicroRNAs in Human Gastric Cancer. Int. J. Mol. Sci. 2016, 17, 945. [Google Scholar] [CrossRef] [Green Version]
  152. Tazawa, H.; Kagawa, S.; Fujiwara, T. MicroRNAs as potential target gene in cancer gene therapy of gastrointestinal tumors. Expert Opin. Biol. Ther. 2011, 11, 145–155. [Google Scholar] [CrossRef]
  153. Song, B.; Ju, J. Impact of miRNAs in gastrointestinal cancer diagnosis and prognosis. Expert Rev. Mol. Med. 2010, 12, e33. [Google Scholar] [CrossRef]
  154. Hu, S.; Zheng, Q.; Wu, H.; Wang, C.; Liu, T.; Zhou, W. miR-532 promoted gastric cancer migration and invasion by targeting NKD1. Life Sci. 2017, 177, 15–19. [Google Scholar] [CrossRef]
  155. Zang, Y.; Wang, T.; Pan, J.; Gao, F. miR-215 promotes cell migration and invasion of gastric cancer cell lines by targeting FOXO1. Neoplasma 2017, 64, 579–587. [Google Scholar] [CrossRef]
  156. Cao, C.; Sun, D.; Zhang, L.; Song, L. miR-186 affects the proliferation, invasion and migration of human gastric cancer by inhibition of Twist1. Oncotarget 2016, 7, 79956–79963. [Google Scholar] [CrossRef] [Green Version]
  157. De Craene, B.; Berx, G. Regulatory networks defining EMT during cancer initiation and progression. Nat. Rev. Cancer 2013, 13, 97–110. [Google Scholar] [CrossRef]
  158. Zhang, X.; Peng, Y.; Huang, Y.; Yang, M.; Yan, R.; Zhao, Y.; Cheng, Y.; Liu, X.; Deng, S.; Feng, X.; et al. SMG-1 inhibition by miR-192/-215 causes epithelial-mesenchymal transition in gastric carcinogenesis via activation of Wnt signaling. Cancer Med. 2018, 7, 146–156. [Google Scholar] [CrossRef]
  159. Zhang, Z.; Li, Z.; Gao, C.; Chen, P.; Chen, J.; Liu, W.; Xiao, S.; Lu, H. miR-21 plays a pivotal role in gastric cancer pathogenesis and progression. Lab. Investig. 2008, 88, 1358–1366. [Google Scholar] [CrossRef] [Green Version]
  160. Tse, J.; Pierce, T.; Carli, A.L.E.; Alorro, M.G.; Thiem, S.; Marcusson, E.G.; Ernst, M.; Buchert, M. Onco-miR-21 Promotes Stat3-Dependent Gastric Cancer Progression. Cancers 2022, 14, 264. [Google Scholar] [CrossRef]
  161. Tsai, M.-M.; Wang, C.-S.; Tsai, C.-Y.; Chen, C.-Y.; Chi, H.-C.; Tseng, Y.-H.; Chung, P.-J.; Lin, Y.-H.; Chung, I.-H.; Chen, C.-Y.; et al. MicroRNA-196a/-196b promote cell metastasis via negative regulation of radixin in human gastric cancer. Cancer Lett. 2014, 351, 222–231. [Google Scholar] [CrossRef] [PubMed]
  162. Wu, Q.; Yang, Z.; An, Y.; Hu, H.; Yin, J.; Zhang, P.; Nie, Y.; Wu, K.; Shi, Y.; Fan, D. MiR-19a/b modulate the metastasis of gastric cancer cells by targeting the tumour suppressor MXD1. Cell Death Dis. 2014, 5, e1144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Wang, D.-W.; Su, F.; Zhang, T.; Yang, T.-C.; Wang, H.-Q.; Yang, L.-J.; Zhou, F.-F.; Feng, M.-H. The miR-370/UQCRC2 axis facilitates tumorigenesis by regulating epithelial-mesenchymal transition in Gastric Cancer. J. Cancer 2020, 11, 5042–5055. [Google Scholar] [CrossRef] [PubMed]
  164. Lo, S.-S.; Hung, P.-S.; Chen, J.-H.; Tu, H.-F.; Fang, W.-L.; Chen, C.-Y.; Chen, W.-T.; Gong, N.-R.; Wu, C.-W. Overexpression of miR-370 and downregulation of its novel target TGFβ-RII contribute to the progression of gastric carcinoma. Oncogene 2012, 31, 226–237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Tie, J.; Pan, Y.; Zhao, L.; Wu, K.; Liu, J.; Sun, S.; Guo, X.; Wang, B.; Gang, Y.; Zhang, Y.; et al. MiR-218 inhibits invasion and metastasis of gastric cancer by targeting the Robo1 receptor. PLoS Genet. 2010, 6, e1000879. [Google Scholar] [CrossRef]
  166. Yang, Q.; Jie, Z.; Cao, H.; Greenlee, A.R.; Yang, C.; Zou, F.; Jiang, Y. Low-level expression of let-7a in gastric cancer and its involvement in tumorigenesis by targeting RAB40C. Carcinogenesis 2011, 32, 713–722. [Google Scholar] [CrossRef] [Green Version]
  167. Korpal, M.; Lee, E.S.; Hu, G.; Kang, Y. The miR-200 family inhibits epithelial-mesenchymal transition and cancer cell migration by direct targeting of E-cadherin transcriptional repressors ZEB1 and ZEB2. J. Biol. Chem. 2008, 283, 14910–14914. [Google Scholar] [CrossRef] [Green Version]
  168. Xu, Y.; Zhao, F.; Wang, Z.; Song, Y.; Luo, Y.; Zhang, X.; Jiang, L.; Sun, Z.; Miao, Z.; Xu, H. MicroRNA-335 acts as a metastasis suppressor in gastric cancer by targeting Bcl-w and specificity protein 1. Oncogene 2011, 31, 1398–1407. [Google Scholar] [CrossRef] [Green Version]
  169. Wang, Z.; Liu, C. MiR-153 regulates metastases of gastric cancer through Snail. Tumor Biol. 2015, 37, 15509–15515. [Google Scholar] [CrossRef]
  170. Luo, H.; Zhang, H.; Zhang, Z.; Zhang, X.; Ning, B.; Guo, J.; Nie, N.; Liu, B.; Wu, X. Down-regulated miR-9 and miR-433 in human gastric carcinoma. J. Exp. Clin. Cancer Res. 2009, 28, 82–89. [Google Scholar] [CrossRef] [Green Version]
  171. Wan, H.-Y.; Guo, L.-M.; Liu, T.; Liu, M.; Li, X.; Tang, H. Regulation of the transcription factor NF-κB1 by microRNA-9 in human gastric adenocarcinoma. Mol. Cancer 2010, 9, 16. [Google Scholar] [CrossRef] [Green Version]
  172. Wang, Y.-Y.; Li, L.; Ye, Z.-Y.; Zhao, Z.-S.; Yan, Z.-L. MicroRNA-10b promotes migration and invasion through Hoxd10 in human gastric cancer. World J. Surg. Oncol. 2015, 13, 259. [Google Scholar] [CrossRef] [Green Version]
  173. Chen, J.; Zhou, C.; Li, J.; Xiang, X.; Zhang, L.; Deng, J.; Xiong, J. miR-21-5p confers doxorubicin resistance in gastric cancer cells by targeting PTEN and TIMP3. Int. J. Mol. Med. 2018, 41, 1855–1866. [Google Scholar] [CrossRef] [Green Version]
  174. Xiao, B.; Guo, J.; Miao, Y.; Jiang, Z.; Huan, R.; Zhang, Y.; Li, D.; Zhong, J. Detection of miR-106a in gastric carcinoma and its clinical significance. Clin. Chim. Acta 2009, 400, 97–102. [Google Scholar] [CrossRef]
  175. Li, X.; Zhang, Y.; Shi, Y.; Dong, G.; Liang, J.; Han, Y.; Wang, X.; Zhao, Q.; Ding, J.; Wu, K.; et al. MicroRNA-107, an oncogene microRNA that regulates tumour invasion and metastasis by targeting DICER1 in gastric cancer. J. Cell. Mol. Med. 2011, 15, 1887–1895. [Google Scholar] [CrossRef] [Green Version]
  176. Hu, C.-B.; Li, Q.-L.; Hu, J.-F.; Zhang, Q.; Xie, J.-P.; Deng, L. miR-124 inhibits growth and invasion of gastric cancer by targeting ROCK1. Asian Pac. J. Cancer Prev. 2014, 15, 6543–6546. [Google Scholar] [CrossRef] [Green Version]
  177. Feng, R.; Chen, X.; Yu, Y.; Su, L.; Yu, B.; Li, J.; Cai, Q.; Yan, M.; Liu, B.; Zhu, Z. miR-126 functions as a tumour suppressor in human gastric cancer. Cancer Lett. 2010, 298, 50–63. [Google Scholar] [CrossRef]
  178. Li, X.; Wang, F.; Qi, Y. MiR-126 inhibits the invasion of gastric cancer cell in part by targeting Crk. Eur. Rev. Med. Pharmacol. Sci. 2014, 18, 2031–2037. [Google Scholar]
  179. Jiang, H.; Yu, W.-W.; Wang, L.-L.; Peng, Y. miR-130a acts as a potential diagnostic biomarker and promotes gastric cancer migration, invasion and proliferation by targeting RUNX3. Oncol. Rep. 2015, 34, 1153–1161. [Google Scholar] [CrossRef] [Green Version]
  180. Wu, Y.; Hu, G.; Wu, R.; Gong, N. High expression of miR-135b predicts malignant transformation and poor prognosis of gastric cancer. Life Sci. 2020, 257, 118133. [Google Scholar] [CrossRef]
  181. Tseng, C.-W.; Lin, C.-C.; Chen, C.-N.; Huang, H.-C.; Juan, H.-F. Integrative network analysis reveals active microRNAs and their functions in gastric cancer. BMC Syst. Biol. 2011, 5, 99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Du, Y.; Wang, D.; Luo, L.; Guo, J. miR-129-1-3p promote BGC-823 cell proliferation by targeting PDCD2. Anat. Rec. 2014, 297, 2273–2279. [Google Scholar] [CrossRef] [PubMed]
  183. Li, Z.; Wang, Y.; Liu, S.; Li, W.; Wang, Z.; Jia, Z.; Zhu, Z.; Bao, Y. MiR-200a-3p promotes gastric cancer progression by targeting DLC-1. Histochem. J. 2021, 53, 39–49. [Google Scholar] [CrossRef] [PubMed]
  184. Li, X.; Zhang, Y.; Zhang, H.; Liu, X.; Gong, T.; Li, M.; Sun, L.; Ji, G.; Shi, Y.; Han, Z.; et al. miRNA-223 promotes gastric cancer invasion and metastasis by targeting tumor suppressor EPB41L3. Mol. Cancer Res. 2011, 9, 824–833. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Kang, W.; Tong, J.H.M.; Chan, A.W.H.; Lung, R.W.M.; Chau, S.L.; Wong, Q.W.L.; Wong, N.; Yu, J.; Cheng, A.S.L.; To, K.F. Stathmin1 plays oncogenic role and is a target of microRNA-223 in gastric cancer. PLoS ONE 2012, 7, e33919. [Google Scholar] [CrossRef]
  186. Zheng, Z.; Li, J.; An, J.; Feng, Y.; Wang, L. High miR-324-5p expression predicts unfavorable prognosis of gastric cancer and facilitates tumor progression in tumor cells. Diagn. Pathol. 2021, 16, 5. [Google Scholar] [CrossRef]
  187. He, X.; Shu, Y. miR-452 promotes the development of gastric cancer via targeting EPB41L3. Pathol. Res. Pract. 2020, 216, 152725. [Google Scholar] [CrossRef]
  188. Wang, Z.-X.; Zhang, G.-J.; Yang, X.-F.; Feng, S.-J.; Ji, S.-S.; Qi, Y.-B. miRNA-633 and KAI1 as potential biomarkers of malignant melanoma with gastric cancer. Comb. Chem. High Throughput Screen 2022, 26, 1001–1014. [Google Scholar] [CrossRef]
  189. Zhang, J.W.; Wang, X.; Li, G.C.; Wang, D.; Han, S.; Zhang, Y.D.; Luo, C.H.; Wang, H.W.; Jiang, W.J.; Li, C.X.; et al. MiR-30a-5p promotes cholangiocarcinoma cell proliferation through targeting SOCS3. J. Cancer 2020, 11, 3604–3614. [Google Scholar] [CrossRef] [Green Version]
  190. Wan, P.; Chi, X.; Du, Q.; Luo, J.; Cui, X.; Dong, K.; Bing, Y.; Heres, C.; Geller, D.A. miR-383 promotes cholangiocarcinoma cell proliferation, migration, and invasion through targeting IRF1. J. Cell. Biochem. 2018, 119, 9720–9729. [Google Scholar] [CrossRef]
  191. Liu, C.-H.; Huang, Q.; Jin, Z.-Y.; Zhu, C.-L.; Liu, Z.; Wang, C. miR-21 and KLF4 jointly augment epithelial-mesenchymal transition via the Akt/ERK1/2 pathway. Int. J. Oncol. 2017, 50, 1109–1115. [Google Scholar] [CrossRef] [Green Version]
  192. Tomimaru, Y.; Eguchi, H.; Nagano, H.; Wada, H.; Kobayashi, S.; Marubashi, S.; Tanemura, M.; Tomokuni, A.; Takemasa, I.; Umeshita, K.; et al. Circulating microRNA-21 as a novel biomarker for hepatocellular carcinoma. J. Hepatol. 2012, 56, 167–175. [Google Scholar] [CrossRef]
  193. Collins, A.L.; Wojcik, S.; Liu, J.; Frankel, W.L.; Alder, H.; Yu, L.; Schmittgen, T.D.; Croce, C.M.; Bloomston, M. A differential microRNA profile distinguishes cholangiocarcinoma from pancreatic adenocarcinoma. Ann. Surg. Oncol. 2014, 21, 133–138. [Google Scholar] [CrossRef] [Green Version]
  194. Liu, Y.; Liu, X.; Zhou, Y.; Liu, T.; Li, J. Overexpression of miR-27a predicts poor prognosis and promotes the progression in cholangiocarcinoma. Clin. Exp. Med. 2021, 21, 121–128. [Google Scholar] [CrossRef]
  195. Cao, K.; Li, B.; Zhang, Y.-W.; Song, H.; Chen, Y.-G.; Gong, Y.-J.; Li, H.-Y.; Zuo, S. miR-29b restrains cholangiocarcinoma progression by relieving DNMT3B-mediated repression of CDKN2B expression. Aging 2021, 13, 6055–6065. [Google Scholar] [CrossRef]
  196. Han, S.; Wang, D.; Tang, G.; Yang, X.; Jiao, C.; Yang, R.; Zhang, Y.; Huo, L.; Shao, Z.; Lu, Z.; et al. Suppression of miR-16 promotes tumor growth and metastasis through reversely regulating YAP1 in human cholangiocarcinoma. Oncotarget 2017, 8, 56635–56650. [Google Scholar] [CrossRef] [Green Version]
  197. Sun, C.; Zhu, J.; Wu, B.; Chen, J.; Zhu, Z.; Cai, P.; Guo, W.; Gu, Z.; Wang, J.; Huang, S. Diagnostic and prognostic value of microRNAs in cholangiocarcinoma: A systematic review and meta-analysis. Cancer Manag. Res. 2018, 10, 2125–2139. [Google Scholar] [CrossRef] [Green Version]
  198. Park, J.W.; Jeong, J.M.; Cho, K.S.; Cho, S.Y.; Cheon, J.H.; Choi, D.H.; Park, S.J.; Kim, H.K. MiR-30a and miR-200c differentiate cholangiocarcinomas from gastrointestinal cancer liver metastases. PLoS ONE 2021, 16, e0250083. [Google Scholar] [CrossRef]
  199. Ni, Q.; Zhang, H.; Shi, X.; Li, X. Exosomal microRNA-23a-3p contributes to the progression of cholangiocarcinoma by interaction with Dynamin3. Bioengineered 2022, 13, 6208–6221. [Google Scholar] [CrossRef]
  200. Yin, X.; Chai, Z.; Sun, X.; Chen, J.; Wu, X.; Yang, L.; Zhou, X.; Liu, F. Overexpression of microRNA-96 is associated with poor prognosis and promotes proliferation, migration and invasion in cholangiocarcinoma cells via MTSS1. Exp. Ther. Med. 2020, 19, 2757–2765. [Google Scholar] [CrossRef] [Green Version]
  201. Chen, T.; Lei, S.; Zeng, Z.; Pan, S.; Zhang, J.; Xue, Y.; Sun, Y.; Lan, J.; Xu, S.; Mao, D.; et al. MicroRNA-137 suppresses the proliferation, migration and invasion of cholangiocarcinoma cells by targeting WNT2B. Int. J. Mol. Med. 2020, 45, 886–896. [Google Scholar] [CrossRef] [PubMed]
  202. Jiang, Z.-L.; Zhang, F.-X.; Zhan, H.-L.; Yang, H.-J.; Zhang, S.-Y.; Liu, Z.-H.; Jiang, Y.; Lv, L.-Z.; Ke, R.-S. miR-181b-5p Promotes the Progression of Cholangiocarcinoma by Targeting PARK2 via PTEN/PI3K/AKT Signaling Pathway. Biochem. Genet. 2022, 60, 223–240. [Google Scholar] [CrossRef] [PubMed]
  203. Liu, C.; Li, Y.; Zhang, L.; Zhang, P.; Yu, N.; Liu, X.; Lu, H.; Du, H.; Hou, S. MiRNA-196-5p Promotes Proliferation and Migration in Cholangiocarcinoma via HAND1/Wnt/β-Catenin Signaling Pathway. J. Oncol. 2022, 2022, 4599676. [Google Scholar] [CrossRef] [PubMed]
  204. Zhu, H.; Jiang, X.; Zhou, X.; Dong, X.; Xie, K.; Yang, C.; Jiang, H.; Sun, X.; Lu, J. Neuropilin-1 regulated by miR-320 contributes to the growth and metastasis of cholangiocarcinoma cells. Liver Int. 2018, 38, 125–135. [Google Scholar] [CrossRef]
  205. Wu, J.; Yang, B.; Zhang, Y.; Feng, X.; He, B.; Xie, H.; Zhou, L.; Wu, J.; Zheng, S. miR-424-5p represses the metastasis and invasion of intrahepatic cholangiocarcinoma by targeting ARK5. Int. J. Biol. Sci. 2019, 15, 1591–1599. [Google Scholar] [CrossRef] [Green Version]
  206. Puik, J.R.; Meijer, L.L.; Le Large, T.Y.; Prado, M.M.; Frampton, A.E.; Kazemier, G.; Giovannetti, E. miRNA profiling for diagnosis, prognosis and stratification of cancer treatment in cholangiocarcinoma. Pharmacogenomics 2017, 18, 1343–1358. [Google Scholar] [CrossRef]
  207. Cheng, Q.; Feng, F.; Zhu, L.; Zheng, Y.; Luo, X.; Liu, C.; Yi, B.; Jiang, X. Circulating miR-106a is a Novel Prognostic and Lymph Node Metastasis Indicator for Cholangiocarcinoma. Sci. Rep. 2015, 5, 16103. [Google Scholar] [CrossRef] [Green Version]
  208. Loosen, S.H.; Lurje, G.; Wiltberger, G.; Vucur, M.; Koch, A.; Kather, J.N.; Paffenholz, P.; Tacke, F.; Ulmer, F.T.; Trautwein, C.; et al. Serum levels of miR-29, miR-122, miR-155 and miR-192 are elevated in patients with cholangiocarcinoma. PLoS ONE 2019, 14, e0210944. [Google Scholar] [CrossRef] [Green Version]
  209. Gao, J.; Dai, C.; Yu, X.; Yin, X.; Zhou, F. Upregulated microRNA-194 impairs stemness of cholangiocarcinoma cells through the Rho pathway via inhibition of ECT2. J. Cell. Biochem. 2020, 121, 4239–4250. [Google Scholar] [CrossRef]
  210. Giordano, S.; Columbano, A. MicroRNAs: New tools for diagnosis, prognosis, and therapy in hepatocellular carcinoma? Hepatology 2012, 57, 840–847. [Google Scholar] [CrossRef] [Green Version]
  211. Vilchez, V.; Turcios, L.; Marti, F.; Gedaly, R. Targeting Wnt/β-catenin pathway in hepatocellular carcinoma treatment. World J. Gastroenterol. 2016, 22, 823–832. [Google Scholar] [CrossRef]
  212. Lai, M.; Du, G.; Shi, R.; Yao, J.; Yang, G.; Wei, Y.; Zhang, D.; Xu, Z.; Zhang, R.; Li, Y.; et al. miR-34a inhibits migration and invasion by regulating the SIRT1/p53 pathway in human SW480 cells. Mol. Med. Rep. 2015, 11, 3301–3307. [Google Scholar] [CrossRef] [Green Version]
  213. Shen, Q.; Cicinnati, V.R.; Zhang, X.; Iacob, S.; Weber, F.; Sotiropoulos, G.C.; Radtke, A.; Lu, M.; Paul, A.; Gerken, G.; et al. Role of microRNA-199a-5p and discoidin domain receptor 1 in human hepatocellular carcinoma invasion. Mol. Cancer 2010, 9, 227. [Google Scholar] [CrossRef] [Green Version]
  214. Zhang, N.-S.; Dai, G.-L.; Liu, S.-J. MicroRNA-29 family functions as a tumor suppressor by targeting RPS15A and regulating cell cycle in hepatocellular carcinoma. Int. J. Clin. Exp. Pathol. 2017, 10, 8031–8042. [Google Scholar]
  215. Tavakolian, S.; Goudarzi, H.; Faghihloo, E. Evaluating the expression level of miR-9-5p and miR-192-5p in gastrointestinal cancer: Introducing novel screening biomarkers for patients. BMC Res. Notes 2020, 13, 226. [Google Scholar] [CrossRef] [Green Version]
  216. Lima, J.F.; Carvalho, J.; Pinto-Ribeiro, I.; Almeida, C.; Wengel, J.; Cerqueira, L.; Figueiredo, C.; Oliveira, C.; Azevedo, N.F. Targeting miR-9 in gastric cancer cells using locked nucleic acid oligonucleotides. BMC Mol. Biol. 2018, 19, 6. [Google Scholar] [CrossRef] [Green Version]
  217. Sun, Z.; Meng, C.; Wang, S.; Zhou, N.; Guan, M.; Bai, C.; Lu, S.; Han, Q.; Zhao, R.C. MicroRNA-1246 enhances migration and invasion through CADM1 in hepatocellular carcinoma. BMC Cancer 2014, 14, 616. [Google Scholar] [CrossRef] [Green Version]
  218. Yang, S.; Zhang, Y.; Guo, C.; Liu, R.; Elkharti, M.; Ge, Z.; Liu, Q.; Liu, S.; Sun, M.Z. The homeostatic malfunction of a novel feedback pathway formed by lncRNA021545, miR-330-3p and epiregulin contributes in hepatocarcinoma progression via mediating epithelial-mesenchymal transition. Am. J. Cancer Res. 2022, 12, 2492–2525. [Google Scholar]
  219. Fornari, F.; Gramantieri, L.; Giovannini, C.; Veronese, A.; Ferracin, M.; Sabbioni, S.; Calin, G.A.; Grazi, G.L.; Croce, C.M.; Tavolari, S.; et al. MiR-122/cyclin G1 interaction modulates p53 activity and affects doxorubicin sensitivity of human hepatocarcinoma cells. Cancer Res. 2009, 69, 5761–5767. [Google Scholar] [CrossRef] [Green Version]
  220. Wong, C.C.L.; Wong, C.-M.; Tung, E.K.; Au, S.L.; Lee, J.M.; Poon, R.T.; Man, K.; Ng, I.O.-L. The microRNA miR-139 suppresses metastasis and progression of hepatocellular carcinoma by down-regulating Rho-kinase 2. Gastroenterology 2011, 140, 322–331. [Google Scholar] [CrossRef]
  221. Nagy, Á.; Lánczky, A.; Menyhárt, O.; Győrffy, B. Validation of miRNA prognostic power in hepatocellular carcinoma using expression data of independent datasets. Sci. Rep. 2018, 8, 9227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Sartorius, K.; Sartorius, B.; Winkler, C.; Chuturgoon, A.; Makarova, J. The biological and diagnostic role of miRNA rsquo s in hepatocellular carcinoma. Front. Biosci. 2018, 23, 1701–1720. [Google Scholar] [CrossRef] [PubMed]
  223. Grossi, I.; Arici, B.; Portolani, N.; De Petro, G.; Salvi, A. Clinical and biological significance of miR-23b and miR-193a in human hepatocellular carcinoma. Oncotarget 2017, 8, 6955–6969. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Sun, T.-Y.; Xie, H.-J.; Li, Z.; Kong, L.-F.; Gou, X.-N.; Li, D.-J.; Shi, Y.-J.; Ding, Y.-Z. miR-34a regulates HDAC1 expression to affect the proliferation and apoptosis of hepatocellular carcinoma. Am. J. Transl. Res. 2017, 9, 103–114. [Google Scholar]
  225. Xu, X.; Chen, W.; Miao, R.; Zhou, Y.; Wang, Z.; Zhang, L.; Wan, Y.; Dong, Y.; Qu, K.; Liu, C. miR-34a induces cellular senescence via modulation of telomerase activity in human hepatocellular carcinoma by targeting FoxM1/c-Myc pathway. Oncotarget 2015, 6, 3988–4004. [Google Scholar] [CrossRef] [Green Version]
  226. Yang, F.; Li, Q.-J.; Gong, Z.-B.; Zhou, L.; You, N.; Wang, S.; Li, X.-L.; Li, J.-J.; An, J.-Z.; Wang, D.-S.; et al. MicroRNA-34a targets Bcl-2 and sensitizes human hepatocellular carcinoma cells to sorafenib treatment. Technol. Cancer Res. Treat. 2014, 13, 77–86. [Google Scholar] [CrossRef]
  227. Li, Z.; Wang, Y. miR-96 targets SOX6 and promotes proliferation, migration, and invasion of hepatocellular carcinoma. Biochem. Cell Biol. 2018, 96, 365–371. [Google Scholar] [CrossRef]
  228. Shen, G.; Jia, H.; Tai, Q.; Li, Y.; Chen, D. miR-106b downregulates adenomatous polyposis coli and promotes cell proliferation in human hepatocellular carcinoma. Carcinogenesis 2013, 34, 211–219. [Google Scholar] [CrossRef] [Green Version]
  229. Callegari, E.; Gramantieri, L.; Domenicali, M.; D’Abundo, L.; Sabbioni, S.; Negrini, M. MicroRNAs in liver cancer: A model for investigating pathogenesis and novel therapeutic approaches. Cell Death Differ. 2015, 22, 46–57. [Google Scholar] [CrossRef] [Green Version]
  230. Li, X.-N.; Yang, H.; Yang, T. miR-122 Inhibits Hepatocarcinoma Cell Progression by Targeting LMNB2. Oncol. Res. Featur. Preclin. Clin. Cancer Ther. 2020, 28, 41–49. [Google Scholar] [CrossRef]
  231. Majid, A.; Wang, J.; Nawaz, M.; Abdul, S.; Ayesha, M.; Guo, C.; Liu, Q.; Liu, S.; Sun, M.-Z. miR-124-3p Suppresses the Invasiveness and Metastasis of Hepatocarcinoma Cells via Targeting CRKL. Front. Mol. Biosci. 2020, 7, 223. [Google Scholar] [CrossRef]
  232. Ou, C.; Peng, N.-F.; Li, H.; Peng, Y.-C.; Li, L.-Q. The potential mechanism of miR-130b on promotion of the invasion and metastasis of hepatocellular carcinoma by inhibiting Notch-Dll1. J. Recept. Signal Transduct. 2020, 40, 157–165. [Google Scholar] [CrossRef]
  233. Xing, B.; Shen, C.; Yang, Q.; Wang, Z.; Tan, W. miR-144-3p represses hepatocellular carcinoma progression by affecting cell aerobic glycolysis via FOXK1. Int. J. Exp. Pathol. 2023, 104, 117–127. [Google Scholar] [CrossRef]
  234. Law, P.T.-Y.; Ching, A.K.-K.; Chan, A.W.-H.; Wong, Q.W.-L.; Wong, C.-K.; To, K.-F.; Wong, N. MiR-145 modulates multiple components of the insulin-like growth factor pathway in hepatocellular carcinoma. Carcinogenesis 2012, 33, 1134–1141. [Google Scholar] [CrossRef] [Green Version]
  235. Ding, J.; Huang, S.; Wu, S.; Zhao, Y.; Liang, L.; Yan, M.; Ge, C.; Yao, J.; Chen, T.; Wan, D.; et al. Gain of miR-151 on chromosome 8q24.3 facilitates tumour cell migration and spreading through downregulating RhoGDIA. Nat. Cell Biol. 2010, 12, 390–399. [Google Scholar] [CrossRef]
  236. Wang, C.; Ren, R.; Hu, H.; Tan, C.; Han, M.; Wang, X.; Zheng, Y. MiR-182 is up-regulated and targeting Cebpa in hepatocellular carcinoma. Chin. J. Cancer Res. 2014, 26, 17–29. [Google Scholar] [CrossRef]
  237. Du, C.; Weng, X.; Hu, W.; Lv, Z.; Xiao, H.; Ding, C.; Gyabaah, O.-A.K.; Xie, H.; Zhou, L.; Wu, J.; et al. Hypoxia-inducible MiR-182 promotes angiogenesis by targeting RASA1 in hepatocellular carcinoma. J. Exp. Clin. Cancer Res. 2015, 34, 67. [Google Scholar] [CrossRef] [Green Version]
  238. Li, R.; Deng, Y.; Liang, J.; Hu, Z.; Li, X.; Liu, H.; Wang, G.; Fu, B.; Zhang, T.; Zhang, Q.; et al. Circular RNA circ-102,166 acts as a sponge of miR-182 and miR-184 to suppress hepatocellular carcinoma proliferation and invasion. Cell. Oncol. 2021, 44, 279–295. [Google Scholar] [CrossRef]
  239. Li, Z.-B.; Li, L.; Chu, H.-T.; Jia, M. MiR-21 and miR-183 can simultaneously target SOCS6 and modulate growth and invasion of hepatocellular carcinoma (HCC) cells. Eur. Rev. Med. Pharmacol. Sci. 2015, 19, 3208–3217. [Google Scholar]
  240. Zhou, L.; Liu, S.; Han, M.; Feng, S.; Liang, J.; Li, Z.; Li, Y.; Lu, H.; Liu, T.; Ma, Y.; et al. MicroRNA-185 induces potent autophagy via AKT signaling in hepatocellular carcinoma. Tumor Biol. 2017, 39, 1010428317694313. [Google Scholar] [CrossRef] [Green Version]
  241. Xu, T.; Zhu, Y.; Xiong, Y.; Ge, Y.-Y.; Yun, J.-P.; Zhuang, S.-M. MicroRNA-195 suppresses tumorigenicity and regulates G1/S transition of human hepatocellular carcinoma cells. Hepatology 2009, 50, 113–121. [Google Scholar] [CrossRef] [PubMed]
  242. Yu, S.; Jing, L.; Yin, X.-R.; Wang, M.-C.; Chen, Y.-M.; Guo, Y.; Nan, K.-J.; Han, L.-L. MiR-195 suppresses the metastasis and epithelial-mesenchymal transition of hepatocellular carcinoma by inhibiting YAP. Oncotarget 2017, 8, 99757–99771. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  243. Fornari, F.; Milazzo, M.; Chieco, P.; Negrini, M.; Calin, G.A.; Grazi, G.L.; Pollutri, D.; Croce, C.M.; Bolondi, L.; Gramantieri, L.; et al. MiR-199a-3p Regulates mTOR and c-Met to Influence the Doxorubicin Sensitivity of Human Hepatocarcinoma Cells. Cancer Res. 2010, 70, 5184–5193. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  244. Xia, H.; Ooi, L.L.P.J.; Hui, K.M. MiR-214 targets β-catenin pathway to suppress invasion, stem-like traits and recurrence of human hepatocellular carcinoma. PLoS ONE 2012, 7, e44206. [Google Scholar] [CrossRef]
  245. Zhang, K.; Fang, T.; Zhao, D.; Cen, F.; Yan, X.; Jin, X. Circular RNA Circ_0008043 promotes the proliferation and metastasis of hepatocellular carcinoma cells by regulating the microRNA (miR)-326/RAB21 axis. Bioengineered 2022, 13, 6600–6614. [Google Scholar] [CrossRef]
  246. Zhang, C.-S.; Lin, Y.; Sun, F.-B.; Gao, J.; Han, B.; Li, S.-J. miR-409 down-regulates Jak-Stat pathway to inhibit progression of liver cancer. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 146–154. [Google Scholar] [CrossRef]
  247. Hui, Y.; Jin, D.; Leng, J.; Liu, D.; Yuan, P.; Tang, C.; Wang, Q. Hsa_circ_0007059 sponges miR-421 to repress cell growth and stemness in hepatocellular carcinoma by the PTEN-AKT/mTOR pathway. Pathol. Res. Pract. 2021, 229, 153692. [Google Scholar] [CrossRef]
  248. Wang, W.; Li, Y.; Li, X.; Liu, B.; Han, S.; Li, X.; Zhang, B.; Li, J.; Sun, S. Circular RNA circ-FOXP1 induced by SOX9 promotes hepatocellular carcinoma progression via sponging miR-875-3p and miR-421. Biomed. Pharmacother. 2020, 121, 109517. [Google Scholar] [CrossRef]
  249. Zhang, B.; Zhou, J. CircSEC24A (hsa_circ_0003528) interference suppresses epithelial-mesenchymal transition of hepatocellular carcinoma cells via miR-421/MMP3 axis. Bioengineered 2022, 13, 9050–9063. [Google Scholar] [CrossRef]
  250. Liu, Y.; Wang, J.; Chen, J.; Wu, S.; Zeng, X.; Xiong, Q.; Guo, Y.; Sun, J.; Song, F.; Xu, J.; et al. Upregulation of miR-520c-3p via hepatitis B virus drives hepatocellular migration and invasion by the PTEN/AKT/NF-κB axis. Mol. Ther. Nucleic Acids 2022, 29, 47–63. [Google Scholar] [CrossRef]
  251. Cui, X.; Zhang, A.; Liu, J.; Wu, K.; Chen, Z.; Wang, Q. Down-regulation of MAP2K1 by miR-539 inhibits hepatocarcinoma progression. Biochem. Biophys. Res. Commun. 2018, 504, 784–791. [Google Scholar] [CrossRef]
  252. Quintavalle, C.; Meyer-Schaller, N.; Roessler, S.; Calabrese, D.; Marone, R.; Riedl, T.; Picco-Rey, S.; Panagiotou, O.A.; Uzun, S.; Piscuoglio, S.; et al. miR-579-3p Controls Hepatocellular Carcinoma Formation by Regulating the Phosphoinositide 3-Kinase–Protein Kinase B Pathway in Chronically Inflamed Liver. Hepatol. Commun. 2022, 6, 1467–1481. [Google Scholar] [CrossRef]
  253. Ayesha, M.; Majid, A.; Zhao, D.; Greenaway, F.T.; Yan, N.; Liu, Q.; Liu, S.; Sun, M.-Z. MiR-4521 plays a tumor repressive role in growth and metastasis of hepatocarcinoma cells by suppressing phosphorylation of FAK/AKT pathway via targeting FAM129A. J. Adv. Res. 2022, 36, 147–161. [Google Scholar] [CrossRef]
  254. To, K.K.; Tong, C.W.; Wu, M.; Cho, W.C. MicroRNAs in the prognosis and therapy of colorectal cancer: From bench to bedside. World J. Gastroenterol. 2018, 24, 2949–2973. [Google Scholar] [CrossRef]
  255. Sha, D.; Lee, A.M.; Shi, Q.; Alberts, S.R.; Sargent, D.J.; Sinicrope, F.A.; Diasio, R.B. Association study of the let-7 miRNA-complementary site variant in the 3’ untranslated region of the KRAS gene in stage III colon cancer (NCCTG N0147 Clinical Trial). Clin. Cancer Res. 2014, 20, 3319–3327. [Google Scholar] [CrossRef] [Green Version]
  256. Slaby, O.; Svoboda, M.; Fabian, P.; Smerdova, T.; Knoflickova, D.; Bednarikova, M.; Nenutil, R.; Vyzula, R. Altered expression of miR-21, miR-31, miR-143 and miR-145 is related to clinicopathologic features of colorectal cancer. Oncology 2007, 72, 397–402. [Google Scholar] [CrossRef]
  257. Tang, W.; Zhu, Y.; Gao, J.; Fu, J.; Liu, C.; Liu, Y.; Song, C.; Zhu, S.; Leng, Y.; Wang, G.; et al. MicroRNA-29a promotes colorectal cancer metastasis by regulating matrix metalloproteinase 2 and E-cadherin via KLF4. Br. J. Cancer 2014, 110, 450–458. [Google Scholar] [CrossRef] [Green Version]
  258. Yu, G.; Tang, J.-Q.; Tian, M.-L.; Li, H.; Wang, X.; Wu, T.; Zhu, J.; Huang, S.-J.; Wan, Y.-L. Prognostic values of the miR-17-92 cluster and its paralogs in colon cancer. J. Surg. Oncol. 2012, 106, 232–237. [Google Scholar] [CrossRef]
  259. Ast, V.; Kordaß, T.; Oswald, M.; Kolte, A.; Eisel, D.; Osen, W.; Eichmüller, S.B.; Berndt, A.; König, R. MiR-192, miR-200c and miR-17 are fibroblast-mediated inhibitors of colorectal cancer invasion. Oncotarget 2018, 9, 35559–35580. [Google Scholar] [CrossRef]
  260. Sukocheva, O.A.; Liu, J.; Neganova, M.E.; Beeraka, N.M.; Aleksandrova, Y.R.; Manogaran, P.; Grigorevskikh, E.M.; Chubarev, V.N.; Fan, R. Perspectives of using microRNA-loaded nanocarriers for epigenetic reprogramming of drug resistant colorectal cancers. Semin. Cancer Biol. 2022, 86 Pt 2, 358–375. [Google Scholar] [CrossRef] [PubMed]
  261. Chen, Q.; Zhou, L.; Ye, X.; Tao, M.; Wu, J. miR-145-5p suppresses proliferation, metastasis and EMT of colorectal cancer by targeting CDCA3. Pathol. Res. Pract. 2020, 216, 152872. [Google Scholar] [CrossRef] [PubMed]
  262. Ma, Y.; Zhang, P.; Wang, F.; Zhang, H.; Yang, Y.; Shi, C.; Xia, Y.; Peng, J.; Liu, W.; Yang, Z.; et al. Elevated oncofoetal miR-17-5p expression regulates colorectal cancer progression by repressing its target gene P130. Nat. Commun. 2012, 3, 1291. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  263. Cheng, D.; Zhao, S.; Tang, H.; Zhang, D.; Sun, H.; Yu, F.; Jiang, W.; Yue, B.; Wang, J.; Zhang, M.; et al. MicroRNA-20a-5p promotes colorectal cancer invasion and metastasis by downregulating Smad4. Oncotarget 2016, 7, 45199–45213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  264. Alotaibi, A.G.; Li, J.V.; Gooderham, N.J. Tumour Necrosis Factor-Alpha (TNF-α)-Induced Metastatic Phenotype in Colorectal Cancer Epithelial Cells: Mechanistic Support for the Role of MicroRNA-21. Cancers 2023, 15, 627. [Google Scholar] [CrossRef]
  265. Chen, T.; Yao, L.-Q.; Shi, Q.; Ren, Z.; Ye, L.-C.; Xu, J.-M.; Zhou, P.-H.; Zhong, Y.-S. MicroRNA-31 contributes to colorectal cancer development by targeting factor inhibiting HIF-1α (FIH-1). Cancer Biol. Ther. 2014, 15, 516–523. [Google Scholar] [CrossRef] [Green Version]
  266. Hiyoshi, Y.; Schetter, A.J.; Okayama, H.; Inamura, K.; Anami, K.; Nguyen, G.H.; Horikawa, I.; Hawkes, J.E.; Bowman, E.D.; Leung, S.Y.; et al. Increased microRNA-34b and -34c predominantly expressed in stromal tissues is associated with poor prognosis in human colon cancer. PLoS ONE 2015, 10, e0124899. [Google Scholar] [CrossRef] [Green Version]
  267. Liu, Y.; Zhou, Y.; Feng, X.; Yang, P.; Yang, J.; An, P.; Wang, H.; Ye, S.; Yu, C.; He, Y.; et al. Low expression of MicroRNA-126 is associated with poor prognosis in colorectal cancer. Genes Chromosom. Cancer 2014, 53, 358–365. [Google Scholar] [CrossRef]
  268. Nagel, R.; le Sage, C.; Diosdado, B.; van der Waal, M.; Vrielink, J.A.O.; Bolijn, A.; Meijer, G.A.; Agami, R. Regulation of the adenomatous polyposis coli gene by the miR-135 family in colorectal cancer. Cancer Res. 2008, 68, 5795–5802. [Google Scholar] [CrossRef] [Green Version]
  269. Hibino, Y.; Sakamoto, N.; Naito, Y.; Goto, K.; Oo, H.Z.; Sentani, K.; Hinoi, T.; Ohdan, H.; Oue, N.; Yasui, W. Significance of miR-148a in Colorectal Neoplasia: Downregulation of miR-148a Contributes to the Carcinogenesis and Cell Invasion of Colorectal Cancer. Pathobiology 2015, 82, 233–241. [Google Scholar] [CrossRef]
  270. Lu, Y.-X.; Yuan, L.; Xue, X.-L.; Zhou, M.; Liu, Y.; Zhang, C.; Li, J.-P.; Zheng, L.; Hong, M.; Li, X.-N. Regulation of colorectal carcinoma stemness, growth, and metastasis by an miR-200c-Sox2-negative feedback loop mechanism. Clin. Cancer Res. 2014, 20, 2631–2642. [Google Scholar] [CrossRef] [Green Version]
  271. Zhou, W.; Yang, W.; Yang, J.; Zhu, H.; Duan, L.; Wang, X.; Li, Y.; Niu, L.; Xiao, S.; Zhang, R.; et al. miR-483 promotes the development of colorectal cancer by inhibiting the expression level of EI24. Mol. Med. Rep. 2021, 24, 567. [Google Scholar] [CrossRef]
  272. Tong, H.-X.; Zhou, Y.-H.; Hou, Y.-Y.; Zhang, Y.; Huang, Y.; Xie, B.; Wang, J.-Y.; Jiang, Q.; He, J.-Y.; Shao, Y.-B.; et al. Expression profile of microRNAs in gastrointestinal stromal tumors revealed by high throughput quantitative RT-PCR microarray. World J. Gastroenterol. 2015, 21, 5843–5855. [Google Scholar] [CrossRef]
  273. Niinuma, T.; Suzuki, H.; Nojima, M.; Nosho, K.; Yamamoto, H.; Takamaru, H.; Yamamoto, E.; Maruyama, R.; Nobuoka, T.; Miyazaki, Y.; et al. Upregulation of miR-196a and HOTAIR drive malignant character in gastrointestinal stromal tumors. Cancer Res. 2012, 72, 1126–1136. [Google Scholar] [CrossRef] [Green Version]
  274. Niinuma, T.; Kai, M.; Kitajima, H.; Yamamoto, E.; Harada, T.; Maruyama, R.; Nobuoka, T.; Nishida, T.; Kanda, T.; Hasegawa, T.; et al. Downregulation of miR-186 is associated with metastatic recurrence of gastrointestinal stromal tumors. Oncol. Lett. 2017, 14, 5703–5710. [Google Scholar] [CrossRef]
  275. Yamamoto, H.; Kohashi, K.; Fujita, A.; Oda, Y. Fascin-1 overexpression and miR-133b downregulation in the progression of gastrointestinal stromal tumor. Mod. Pathol. 2013, 26, 563–571. [Google Scholar] [CrossRef] [Green Version]
  276. Kupcinskas, J. Small Molecules in Rare Tumors: Emerging Role of MicroRNAs in GIST. Int. J. Mol. Sci. 2018, 19, 397. [Google Scholar] [CrossRef] [Green Version]
  277. Liu, X.; Li, J.; Qin, F.; Dai, S. miR-152 as a tumor suppressor microRNA: Target recognition and regulation in cancer. Oncol. Lett. 2016, 11, 3911–3916. [Google Scholar] [CrossRef] [Green Version]
  278. Akçakaya, P.; Lui, W.-O. MicroRNAs and Gastrointestinal Stromal Tumor. Adv. Exp. Med. Biol. 2015, 889, 51–70. [Google Scholar] [CrossRef]
  279. Gyvyte, U.; Lukosevicius, R.; Inciuraite, R.; Streleckiene, G.; Gudoityte, G.; Bekampyte, J.; Valentini, S.; Salteniene, V.; Ruzgys, P.; Satkauskas, S.; et al. The Role of miR-375-3p and miR-200b-3p in Gastrointestinal Stromal Tumors. Int. J. Mol. Sci. 2020, 21, 5151. [Google Scholar] [CrossRef]
  280. Turriziani, M.; Fantini, M.; Benvenuto, M.; Izzi, V.; Masuelli, L.; Sacchetti, P.; Modesti, A.; Bei, R. Carcinoembryonic antigen (CEA)-based cancer vaccines: Recent patents and antitumor effects from experimental models to clinical trials. Recent Patents Anti-Cancer Drug Discov. 2012, 7, 265–296. [Google Scholar] [CrossRef]
  281. Bei, R.; Guptill, V.; Masuelli, L.; Kashmiri, S.V.S.; Muraro, R.; Frati, L.; Schlom, J.; Kantor, J. The use of a cationic liposome formulation (DOTAP) mixed with a recombinant tumor-associated antigen to induce immune responses and protective immunity in mice. J. Immunother. 1998, 21, 159–169. [Google Scholar] [CrossRef] [PubMed]
  282. You, W.; Yan, L.; Cai, Z.; Xie, L.; Sheng, N.; Wang, G.; Wu, X.; Wang, Z. Clinical Significances of Positive Postoperative Serum CEA and Post-preoperative CEA Increment in Stage II and III Colorectal Cancer: A Multicenter Retrospective Study. Front. Oncol. 2020, 10, 671. [Google Scholar] [CrossRef]
  283. Shibata, C.; Nakano, T.; Yasumoto, A.; Mitamura, A.; Sawada, K.; Ogawa, H.; Miura, T.; Ise, I.; Takami, K.; Yamamoto, K.; et al. Comparison of CEA and CA19-9 as a predictive factor for recurrence after curative gastrectomy in gastric cancer. BMC Surg. 2022, 22, 213. [Google Scholar] [CrossRef] [PubMed]
  284. Feng, F.; Tian, Y.; Xu, G.; Liu, Z.; Liu, S.; Zheng, G.; Guo, M.; Lian, X.; Fan, D.; Zhang, H. Diagnostic and prognostic value of CEA, CA19–9, AFP and CA125 for early gastric cancer. BMC Cancer 2017, 17, 737. [Google Scholar] [CrossRef] [PubMed]
  285. Lee, J.W.; Lee, J.H.; Park, Y.; Kwon, J.; Lee, W.; Song, K.B.; Hwang, D.W.; Kim, S.C. Prognostic Impact of Perioperative CA19-9 Levels in Patients with Resected Perihilar Cholangiocarcinoma. J. Clin. Med. 2021, 10, 1345. [Google Scholar] [CrossRef]
  286. Shan, M.; Tian, Q.; Zhang, L. Serum CA50 levels in patients with cancers and other diseases. Prog. Mol. Biol. Transl. Sci. 2019, 162, 187–198. [Google Scholar] [CrossRef]
  287. Pan, Q.; Law, C.O.K.; Yung, M.M.H.; Han, K.C.; Pon, Y.L.; Lau, T.C.K. Novel RNA aptamers targeting gastrointestinal cancer biomarkers CEA, CA50 and CA72-4 with superior affinity and specificity. PLoS ONE 2018, 13, e0198980. [Google Scholar] [CrossRef] [Green Version]
  288. Hu, P.-J.; Chen, M.-Y.; Wu, M.-S.; Lin, Y.-C.; Shih, P.-H.; Lai, C.-H.; Lin, H.-J. Clinical Evaluation of CA72-4 for Screening Gastric Cancer in a Healthy Population: A Multicenter Retrospective Study. Cancers 2019, 11, 733. [Google Scholar] [CrossRef] [Green Version]
  289. Bei, R. Alpha-fetoprotein is an autoantigen in hepatocellular carcinoma and juvenile Batten disease. Front. Biosci. 2020, 25, 912–929. [Google Scholar] [CrossRef]
  290. Guo, W.; Cao, P.; Wang, X.; Hu, M.; Feng, Y. Medicinal Plants for the Treatment of Gastrointestinal Cancers from the Metabolomics Perspective. Front. Pharmacol. 2022, 13, 909755. [Google Scholar] [CrossRef]
  291. Masuelli, L.; Benvenuto, M.; Focaccetti, C.; Ciuffa, S.; Fazi, S.; Bei, A.; Miele, M.T.; Piredda, L.; Manzari, V.; Modesti, A.; et al. Targeting the tumor immune microenvironment with “nutraceuticals”: From bench to clinical trials. Pharmacol. Ther. 2020, 219, 107700. [Google Scholar] [CrossRef]
  292. Das, L.; Bhaumik, E.; Raychaudhuri, U.; Chakraborty, R. Role of nutraceuticals in human health. J. Food Sci. Technol. 2012, 49, 173–183. [Google Scholar] [CrossRef] [Green Version]
  293. AlAli, M.; Alqubaisy, M.; Aljaafari, M.N.; AlAli, A.O.; Baqais, L.; Molouki, A.; Abushelaibi, A.; Lai, K.-S.; Lim, S.-H.E. Nutraceuticals: Transformation of Conventional Foods into Health Promoters/Disease Preventers and Safety Considerations. Molecules 2021, 26, 2540. [Google Scholar] [CrossRef]
  294. Chen, C.; Ma, T.; Zhang, C.; Bai, L.; Kong, L.; Luo, J. Down-regulation of aquaporin 5-mediated epithelial-mesenchymal transition and anti-metastatic effect by natural product Cairicoside E in colorectal cancer. Mol. Carcinog. 2017, 56, 2692–2705. [Google Scholar] [CrossRef]
  295. He, L.; Zhong, Z.; Chen, M.; Liang, Q.; Wang, Y.; Tan, W. Current Advances in Coptidis Rhizoma for Gastrointestinal and Other Cancers. Front. Pharmacol. 2021, 12, 775084. [Google Scholar] [CrossRef]
  296. Liu, Q.; Tang, J.; Chen, S.; Hu, S.; Shen, C.; Xiang, J.; Chen, N.; Wang, J.; Ma, X.; Zhang, Y.; et al. Berberine for gastric cancer prevention and treatment: Multi-step actions on the Correa’s cascade underlie its therapeutic effects. Pharmacol. Res. 2022, 184, 106440. [Google Scholar] [CrossRef]
  297. Ortiz, L.M.G.; Lombardi, P.; Tillhon, M.; Scovassi, A.I. Berberine, an epiphany against cancer. Molecules 2014, 19, 12349–12367. [Google Scholar] [CrossRef] [Green Version]
  298. You, H.-Y.; Xie, X.-M.; Zhang, W.-J.; Zhu, H.-L.; Jiang, F.-Z. Berberine modulates cisplatin sensitivity of human gastric cancer cells by upregulation of miR-203. In Vitro Cell. Dev. Biol. Anim. 2016, 52, 857–863. [Google Scholar] [CrossRef]
  299. Pollier, J.; Goossens, A. Oleanolic acid. Phytochemistry 2012, 77, 10–15. [Google Scholar] [CrossRef]
  300. Xu, Q.-F.; Peng, H.-P.; Lu, X.-R.; Hu, Y.; Xu, Z.-H.; Xu, J.-K. Oleanolic acid regulates the Treg/Th17 imbalance in gastric cancer by targeting IL-6 with miR-98-5p. Cytokine 2021, 148, 155656. [Google Scholar] [CrossRef]
  301. Wang, Z.-F.; Ma, D.-G.; Wang, L.; Feng, L.; Fu, J.-W.; Li, Y.; Wang, D.-T.; Jia, Y.-F. Paeoniflorin Inhibits Migration- and Invasion-Promoting Capacities of Gastric Cancer Associated Fibroblasts. Chin. J. Integr. Med. 2019, 25, 837–844. [Google Scholar] [CrossRef] [PubMed]
  302. Elkashty, O.A.; Tran, S.D. Sulforaphane as a Promising Natural Molecule for Cancer Prevention and Treatment. Curr. Med. Sci. 2021, 41, 250–269. [Google Scholar] [CrossRef] [PubMed]
  303. Kim, D.H.; Sung, B.; Kang, Y.J.; Hwang, S.Y.; Kim, M.J.; Yoon, J.-H.; Im, E.; Kim, N.D. Sulforaphane inhibits hypoxia-induced HIF-1α and VEGF expression and migration of human colon cancer cells. Int. J. Oncol. 2015, 47, 2226–2232. [Google Scholar] [CrossRef] [Green Version]
  304. Masuelli, L.; Marzocchella, L.; Focaccetti, C.; Tresoldi, I.; Palumbo, C.; Izzi, V.; Benvenuto, M.; Fantini, M.; Lista, F.; Tarantino, U.; et al. Resveratrol and diallyl disulfide enhance curcumin-induced sarcoma cell apoptosis. Front. Biosci. 2012, 17, 498–508. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  305. Thongchot, S.; Ferraresi, A.; Vidoni, C.; Loilome, W.; Yongvanit, P.; Namwat, N.; Isidoro, C. Resveratrol interrupts the pro-invasive communication between cancer associated fibroblasts and cholangiocarcinoma cells. Cancer Lett. 2018, 430, 160–171. [Google Scholar] [CrossRef]
  306. Masuelli, L.; Granato, M.; Benvenuto, M.; Mattera, R.; Bernardini, R.; Mattei, M.; D’Amati, G.; D’Orazi, G.; Faggioni, A.; Bei, R.; et al. Chloroquine supplementation increases the cytotoxic effect of curcumin against Her2/neu overexpressing breast cancer cells in vitro and in vivo in nude mice while counteracts it in immune competent mice. Oncoimmunology 2017, 6, e1356151. [Google Scholar] [CrossRef] [Green Version]
  307. Focaccetti, C.; Benvenuto, M.; Ciuffa, S.; Fazi, S.; Scimeca, M.; Nardi, A.; Miele, M.T.; Battisti, A.; Bonanno, E.; Modesti, A.; et al. Curcumin Enhances the Antitumoral Effect Induced by the Recombinant Vaccinia Neu Vaccine (rV-neuT) in Mice with Transplanted Salivary Gland Carcinoma Cells. Nutrients 2020, 12, 1417. [Google Scholar] [CrossRef]
  308. Prakobwong, S.; Khoontawad, J.; Yongvanit, P.; Pairojkul, C.; Hiraku, Y.; Sithithaworn, P.; Pinlaor, P.; Aggarwal, B.B.; Pinlaor, S. Curcumin decreases cholangiocarcinogenesis in hamsters by suppressing inflammation-mediated molecular events related to multistep carcinogenesis. Int. J. Cancer 2010, 129, 88–100. [Google Scholar] [CrossRef]
  309. Chen, Q.; Guo, H.; Zong, Y.; Zhao, X. Curcumin restrains hepatocellular carcinoma progression depending on the regulation of the circ_0078710/miR-378b/PRIM2 axis. J. Recept. Signal Transduct. Res. 2021, 42, 313–324. [Google Scholar] [CrossRef]
  310. Akbari, A.; Sedaghat, M.; Heshmati, J.; Tabaeian, S.P.; Dehghani, S.; Pizarro, A.B.; Rostami, Z.; Agah, S. Molecular mechanisms underlying curcumin-mediated microRNA regulation in carcinogenesis; Focused on gastrointestinal cancers. Biomed. Pharmacother. 2021, 141, 111849. [Google Scholar] [CrossRef]
  311. Avila-Carrasco, L.; Majano, P.; Sánchez-Toméro, J.A.; Selgas, R.; López-Cabrera, M.; Aguilera, A.; Mateo, G.G. Natural Plants Compounds as Modulators of Epithelial-to-Mesenchymal Transition. Front. Pharmacol. 2019, 10, 715. [Google Scholar] [CrossRef] [Green Version]
  312. Zhang, S.; Cheng, Z.; Wang, Y.; Han, T. The Risks of miRNA Therapeutics: In a Drug Target Perspective. Drug Des. Dev. Ther. 2021, 15, 721–733. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of gastrointestinal TME and biological events leading to cell migration and metastasis formation. A pro-tumoral environment is established in the TME of GI tumors given by the presence of activated CAFs and immunosuppressive immune cells, leading to the production of cytokines, chemokines, and soluble factors. These are responsible for ECM remodeling and promotion of EMT that enhances tumor cell motility and subsequent metastasis formation. The figure was created with BioRender.com.
Figure 1. Schematic representation of gastrointestinal TME and biological events leading to cell migration and metastasis formation. A pro-tumoral environment is established in the TME of GI tumors given by the presence of activated CAFs and immunosuppressive immune cells, leading to the production of cytokines, chemokines, and soluble factors. These are responsible for ECM remodeling and promotion of EMT that enhances tumor cell motility and subsequent metastasis formation. The figure was created with BioRender.com.
Biomedicines 11 01761 g001
Table 1. Up- or down-regulated miRNAs in various gastrointestinal tumors compared to normal tissues.
Table 1. Up- or down-regulated miRNAs in various gastrointestinal tumors compared to normal tissues.
miRNAUp/Down-RegulatedType of CancerRef.
let-7UpColon cancer[118]
DownHepatocellular carcinoma[119]
DownGastric cancer[120]
UpHepatocellular carcinoma[121]
DownCholangiocarcinoma[122]
miR-9UpHepatocellular carcinoma[123]
UpGastric cancer[124]
miR-10a-5pUpCholangiocarcinoma[125]
miR-21UpColorectal cancer[126]
UpHepatocellular carcinoma[127]
UpCholangiocarcinoma [128]
miR-23UpHepatocellular carcinoma[121]
miR-23a-3pUpColorectal cancer[129]
miR-27aUpGastric cancer[130]
miR-29sDownCholangiocarcinoma[131]
miR-31DownColorectal cancer[132]
miR-93-5pUpColorectal cancer[133]
miR-103a-3pUpColorectal cancer[134]
miR-152-3pUpColorectal cancer[135]
miR-199a-5pDownHepatocellular carcinoma[136]
miR-210UpGastric cancer[137]
miR-922UpGastric cancer[138]
Table 2. Aberrantly expressed miRNAs in Gastric cancer, with their specific targets.
Table 2. Aberrantly expressed miRNAs in Gastric cancer, with their specific targets.
miRNATargetEffectsRefs.
let-7RAB40C
HMGA2
CCR7
Induces proliferation
Promotes invasion
[166]
miR-9NKFB1
CCND1
CDX2
Induces proliferation
Metastasis
[170,171]
miR-10bHOXD10Promotes cell migration and invasion
Metastasis
[172]
miR-21RECK
PTEN
PDCD4
VIMENTIN
SNAIL TIMP3
Promotes cell migration
Induces EMT
Metastasis
Drug resistance
[159,160,173]
miR-106bPTEN
RB1
TIMP2
Induces cell proliferation
Promotes cell migration and invasion
[174]
miR-107DICER1Promotes cell migration and invasion[175]
miR-124ROCK1Induces cell proliferation
Promotes cell invasion
[176]
miR-126CRK
PI3KR2
Induces cell proliferation
Promotes cell migration and invasion
Metastasis
[177,178]
miR-130aRUNX3Induces metastasis[179]
miR-135bn.d.n.d.[180]
miR-148aCDKN1BInduces cell proliferation
Regulates cell cycle
Metastasis
[181]
miR-153SNAILPromotes cell migration and invasion
Metastasis
[169]
miR-186TWIST1Promotes cell migration[156]
miR-192-1-3pPDCD2Induces cell proliferation[182]
miR-192-5pSMG-1Induces cell proliferation and EMT[158]
miR-196RADIXIN
MXD1
Promotes cell migration and invasion
Metastasis
[161,162]
miR-200DLC-1
ZEB1
ZEB2
BCL-2
XIAP
Induces cell proliferation and EMT
Promotes cell migration and invasion
[167,183]
miR-210DRD5Promotes cell migration and invasion[137]
miR-215FOXO1Promotes cell migration[155]
miR-218ROBO1Promotes cell invasion
Metastasis
[165]
miR-223STMN1Promotes cell invasion
Metastasis
[184,185]
miR-324-5pPTENInduces cell proliferation
Promotes apoptosis
[186]
miR-335BCL-wMetastasis[168]
miR-370TGF-β-RII
UQCRC2
Induces EMT
Metastasis
[163,164]
miR-452EPB41L3Promotes cell migration and invasion[187]
miR-532NKD1Promotes cell migration and invasion[154]
miR-633n.d.n.d.[188]
miR-922SOCS1Induces cell invasion
Promotes cell migration
[138]
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

Lucarini, V.; Nardozi, D.; Angiolini, V.; Benvenuto, M.; Focaccetti, C.; Carrano, R.; Besharat, Z.M.; Bei, R.; Masuelli, L. Tumor Microenvironment Remodeling in Gastrointestinal Cancer: Role of miRNAs as Biomarkers of Tumor Invasion. Biomedicines 2023, 11, 1761. https://doi.org/10.3390/biomedicines11061761

AMA Style

Lucarini V, Nardozi D, Angiolini V, Benvenuto M, Focaccetti C, Carrano R, Besharat ZM, Bei R, Masuelli L. Tumor Microenvironment Remodeling in Gastrointestinal Cancer: Role of miRNAs as Biomarkers of Tumor Invasion. Biomedicines. 2023; 11(6):1761. https://doi.org/10.3390/biomedicines11061761

Chicago/Turabian Style

Lucarini, Valeria, Daniela Nardozi, Valentina Angiolini, Monica Benvenuto, Chiara Focaccetti, Raffaele Carrano, Zein Mersini Besharat, Roberto Bei, and Laura Masuelli. 2023. "Tumor Microenvironment Remodeling in Gastrointestinal Cancer: Role of miRNAs as Biomarkers of Tumor Invasion" Biomedicines 11, no. 6: 1761. https://doi.org/10.3390/biomedicines11061761

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