Next Article in Journal
Lymphoid Tissue in Teleost Gills: Variations on a Theme
Next Article in Special Issue
5-Fluorouracil Conversion Pathway Mutations in Gastric Cancer
Previous Article in Journal
Phosphoinositide Lipids in Ocular Tissues
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

STAT3 Pathway in Gastric Cancer: Signaling, Therapeutic Targeting and Future Prospects

1
Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz 5166616471, Iran
2
Sabanci University Nanotechnology Research and Application Center (SUNUM), 34956 Tuzla, Istanbul, Turkey
3
Center of Excellence for Functional Surfaces and Interfaces (EFSUN), Faculty of Engineering and Natural Sciences, Sabanci University, 34956 Tuzla, Istanbul, Turkey
4
Department of Genetic Science, Tehran Medical Science Branch, Islamic Azad University, Tehran 1916893813, Iran
5
Laboratory for Stem Cell Research, Shiraz University of Medical Sciences, Shiraz 7134814336, Iran
6
Department of Anatomical sciences, School of Medicine, Student Research Committee, Shiraz University of Medical Sciences, Shiraz 7134814336, Iran
7
Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran 1916893813, Iran
8
General Practitioner, Kerman University of Medical Sciences, Kerman 7616913555, Iran
9
Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran 1417414418, Iran
10
Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz 5166616471, Iran
11
Institute for Polymers, Composites and Biomaterials (IPCB), National Research Council (CNR), 80125 Naples, Italy
12
Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran
13
Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman 7616911319, Iran
*
Authors to whom correspondence should be addressed.
Biology 2020, 9(6), 126; https://doi.org/10.3390/biology9060126
Submission received: 16 May 2020 / Revised: 2 June 2020 / Accepted: 4 June 2020 / Published: 12 June 2020

Abstract

:
Molecular signaling pathways play a significant role in the regulation of biological mechanisms, and their abnormal expression can provide the conditions for cancer development. The signal transducer and activator of transcription 3 (STAT3) is a key member of the STAT proteins and its oncogene role in cancer has been shown. STAT3 is able to promote the proliferation and invasion of cancer cells and induces chemoresistance. Different downstream targets of STAT3 have been identified in cancer and it has also been shown that microRNA (miR), long non-coding RNA (lncRNA) and other molecular pathways are able to function as upstream mediators of STAT3 in cancer. In the present review, we focus on the role and regulation of STAT3 in gastric cancer (GC). miRs and lncRNAs are considered as potential upstream mediators of STAT3 and they are able to affect STAT3 expression in exerting their oncogene or onco-suppressor role in GC cells. Anti-tumor compounds suppress the STAT3 signaling pathway to restrict the proliferation and malignant behavior of GC cells. Other molecular pathways, such as sirtuin, stathmin and so on, can act as upstream mediators of STAT3 in GC. Notably, the components of the tumor microenvironment that are capable of targeting STAT3 in GC, such as fibroblasts and macrophages, are discussed in this review. Finally, we demonstrate that STAT3 can target oncogene factors to enhance the proliferation and metastasis of GC cells.

1. Introduction

Gastric cancer (GC) is one of the leading causes of death worldwide and its incidence rate is different among nations [1,2,3]. It has been reported that Eastern Asia claims the highest incidence rate of GC and after that, Central/Eastern Europe has the second place. It seems that non-cardiac GC is prevalent in the aforementioned places and cardiac GC has a high incidence rate in North America, Australia and the United Kingdom [4]. It is worth mentioning that infections can lead to the emergence of GC. The most well-known example of an infection resulting in GC is Helicobacter pylori (H. pylori) infection. There is information about the involvement of Epstein–Barr virus (EBV) infection in GC development [5]. The alterations in lifestyle play a significant role in GC emergence. Among them, cigarette smoking, obesity, high salt intake and low vegetable consumption are of importance [5,6]. The metastatic nature of GC cells has made them a leading cause of death. In a recent study, it was revealed that patients with GC exhibit metastasis into neighboring and distant tissues, such as the liver, peritoneum, lung and bone [7]. So, it seems that GC is a multifactorial disorder and dealing with this life-threatening disorder requires an understanding of the factors involved in its development and malignancy.
To date, a large body of evidence has been provided about the factors leading to GC progression. It seems that complex molecular pathways are the most important ones in GC malignancy [8]. The identification of these pathways can pave the way to effective GC therapy. Thanks to experiments performed in this field, it has been reported that a high number of molecular pathways and their upstream and downstream mediators, can contribute to GC development [9,10,11]. Due to limitations in space, it is impossible to describe all these underlying pathways, but we briefly discuss them to shed some light on the complexity of these signaling networks. The long non-coding RNAs (lncRNAs) are considered as the most common upstream mediators of molecular signaling pathways, since they are capable of the regulation of different pathways that affect biological mechanisms such as apoptosis, proliferation differentiation and so on [12,13]. In GC cells, lncRNA LINC00511 reduces the expression of microRNA (miR)-124-3p to induce PDK4. This axis leads to an increase in the proliferation and progression of GC cells [14]. Notably, there are interactions between lncRNAs in GC. The lncRNA PTCSC3 is an onco-suppressor factor and its expression undergoes downregulation in GC cells. There is negative feedback between PTCSC3 and HOXA11-AS, so that the upregulation of PTCSC3 provides the conditions for the downregulation of HOXA11-AS, resulting in decreased invasion and proliferation of GC cells [15]. As mentioned earlier, GC cells have a high metastatic ability that is correlated with the poor prognosis of patients. The epithelial-to-mesenchymal transition (EMT) is a signaling pathway capable of enhancing the migration of cancer cells [16,17]. The non-coding RNAs are able to act as upstream mediators of EMT in cancer cells [18]. In GC cells, the Wnt/β-catenin signaling pathway induces EMT to ensure the migration and invasion of cancer cells. The miR-330-3p acts as an onco-suppressor factor, inhibiting EMT through Wnt downregulation, leading to a decrease in the metastasis of GC cells, and an improvement in prognosis [19]. It is suggested that the tumor microenvironment plays a significant role in the progression and immune evasion of GC cells. The secretion of inflammatory cytokines, such as IL-4 and IFN-γ, results in PD-L2 expression that in turn provides immune suppression, leading to the proliferation and survival of cancer cells [20]. The growth of GC cells relies on the activation of molecular pathways that promote glucose metabolism. Due to the uncontrolled proliferation of GC cells, these networks should be in precise regulation. The PI3K/Akt signaling pathway participates in the proliferation of cancer cells and its disruption can pave the way to growth suppression [21]. In GC cells, HBXIP induces the PI3K/Akt pathway to enhance glucose metabolism. The inhibition of the HBXIP/PI3K/Akt axis is correlated with an inhibition in the growth of GC cells [22]. Overall, the studies are in line with the fact that a complicated network of molecular pathways is involved in GC malignancy [23,24]. In addition to the identification of molecular pathways, there have been attempts to target them using anti-tumor compounds. It seems that anti-tumor compounds, such as chrysin, psoralen and tivatinib, are able to induce apoptosis in GC cells and suppress their angiogenesis and migration through the downregulation of vascular endothelial growth factor (VEGF) [25,26,27]. In the current review, we present a comprehensive discussion of the STAT3 signaling pathway in GC malignancy and its regulation by upstream mediators. Furthermore, we represent how anti-tumor drugs can target STAT3 in suppressing GC progression and metastasis.

2. STAT3 Signaling Pathway and Its Role in Pathological Events

Signal transducer and activator of transcription (STAT) proteins are mediators for transferring signals from the extracellular matrix into the nucleus [28,29]. Among transcription factors, the role of STAT proteins has been extensively examined [29,30,31,32]. There are seven members of STAT proteins and STAT3 is of importance due to its role in affecting genes involved in cancer progression and malignancy [33]. As a protein with 770 amino acids, STAT3 has six characteristic domains with special functions. The protein–protein interactions are mediated via the N-terminal domain of the STAT3 protein. The coil–coil domain induces a nuclear localization signal. The interaction of STAT3 with target genes is performed through a DNA-binding domain. Src homology-2 (SH2) participates in STAT3 dimerization by the identification of phospho-tyrosine in the target protein. The phosphorylation of the C-terminal transcription activation domain (TAD) at serine sites provides the assembly of STAT3 with other transcriptional activators [34,35,36]. This shows that the STAT3 structure has been designed for interacting with different molecules, and this has resulted in pleiotropic impacts on STAT3 signaling pathway.
The STAT3 signaling pathway is induced by the binding of an extracellular ligand, such as IL-6. This leads to the receptor dimerization and trans-phosphorylation/activation of Janus kinases (JAKs). The JAK proteins are involved in providing docking sites for STAT3 by the phosphorylation of cytoplasmic receptor tails. Then, JAK proteins activate the STAT3 signaling pathway via phosphorylation at tyrosine 705. After activation, STAT3 separates from the receptor/kinase complex to produce homodimers or heterodimers using its SH2 domain. Further signaling depends on translocation into the nucleus and targeting genes. Upon nuclear translocation, the STAT3 signaling pathway contributes to an enhancement in the proliferation, metastasis and migration of cancer cells. During physiological conditions, suppressor of cytokine signaling (SOCS) proteins, protein inhibitor of activated STAT (PIAS) proteins and phosphatases are able to negatively affect the expression of STAT3 and suppress its nuclear translocation. The SOCS3 interferes with JAK activity, while PIAS3 inhibits the STAT3–DNA interaction. The phosphatases, such as SHP-1, SHP-2 and PTP1B, can dually suppress JAK kinase activity or its nuclear interaction with target genes [37,38,39,40,41,42,43,44]. In cancer cells, the endogenous inhibitors of the STAT3 signaling pathway undergo downregulation, which mediates the enhanced proliferation and malignancy of cancer cells (Figure 1) [45,46,47,48].
Similar to other molecular signaling pathways, STAT3 can be considered as a downstream target. Proviral integration site for Moloney murine leukemia virus (PIM) kinases are a category of serine/threonine kinases and include PIM1, PIM2 and PIM3. PIM1 increases the proliferation and growth of cancer cells via the downregulation of cycle cell suppressor p27 [49]. In lung cancer cells, PIM1 stimulates STAT3 phosphorylation to suppress apoptosis [50]. The molecular pathways allowing the survival of cancer cells target STAT3. Stomatin-like protein 2 (SLP-2) is an oncogene factor that ensures the growth and viability of colorectal cancer cells via the upregulation of JAK2/STAT3 [51]. The lncRNAs are able to act as upstream mediators of STAT3 [52]. The lncRNA LINC01535 reduces the sensitivity of cancer cells to apoptosis and enhances their growth through the induction of the JAK/STAT3 signaling pathway [53]. The anti-tumor compounds can target the STAT3 signaling pathway in cancer therapy. For instance, Centipeda minima is a key member of Chinese traditional medicine (CTM) that suppresses the invasion and malignancy of breast cancer cells via the downregulation of the STAT3 pathway [54]. Brevilin A suppresses the phosphorylation of STAT3 at tyrosine 70 to restrict the growth of lung cancer cells [55]. These studies are in line with the fact that STAT3 is a tumorigenesis factor in cancer cells, and its inhibition is an ideal strategy in overcoming cancer (Table 1) [56,57,58,59].

3. Targeting STAT3 in Clinical Trials: A Focus on Cancer Therapy

In respect to the role of STAT3 in the development and progression of cancer, a variety of clinical trials have focused on targeting STAT3 in the treatment of patients with cancer. Table 2 summarizes the clinical trials related to targeting STAT3 in cancer therapy. In a study, AZD9150, as an inhibitor of STAT3, was administered for the treatment of patients with lymphoma and lung cancers. Before the introduction of AZD9150 in clinical trials, its efficacy was investigated in preclinical models of lymphoma and lung cancer. It effectively inhibited the proliferation and invasion of cancer cells. Then, it was translated into clinical trials. Similarly, in patients with lymphoma and lung cancer, AZD9150 demonstrated high anti-tumor activity [76], demonstrating its potential for being used in clinical trials. Notably, clinical trials have focused on using an expression profile of STAT3 as a prognostic factor. This study demonstrated that the expression of phosphorylated STAT3 (p-STAT3) is associated with the desirable prognosis of patients with luminal breast cancer. This phase III study puts an emphasis on the positive relationship between STAT3 and favorable prognosis [77]. Although this study provides controversial results that are not in line with our aforementioned statements, another study (104 French patients) also confirms that the expression of p-STAT3 is associated with the improved overall survival of patients with rectal cancer [78]. More studies are needed to clarify these discussions.
One of the challenges faced in radiotherapy is the resistance of cancer cells. Clinical trials have examined the relationship between STAT3 and the response of cancer cells to radiotherapy. It seems that STAT3 accumulation in cancer cells and its nuclear translocation can lead to radio resistance. In patients who are resistant into radiotherapy, genes associated with retinoid IFN-induced mortality-19 (GRIM-19) can inhibit STAT3 accumulation, resulting in enhanced sensitivity to radiotherapy [79].
Complicated molecular pathways are involved in ensuring the malignant behavior of cancer cells. In ovarian cancer, IL-6 induces STAT3 expression that in turn activates hypoxia inducible factor (HIF), resulting in the resistance of patients with ovarian cancer to chemotherapy with sunitinib [80]. Another clinical trial considered STAT3 as a factor involved in the migration of prostate cancer cells. In this study, 456 people were enrolled and it was found that STAT3 induction has a reverse correlation with distant metastasis, but it can lead to the local progression of prostate cancer cells [81].

4. Search Strategy

We conducted a literature search in different databases, such as PubMed, Scopus and Google scholar. Articles in English that were published before April 2020 were collected. The keywords included “STAT3 & gastric cancer”, “drug & STAT3 & gastric cancer”, “lncRNA & STAT3 & gastric cancer” and “miR & gastric cancer & STAT3”.

5. STAT3 and Gastric Cancer

5.1. MicroRNA-Mediated Regulation of STAT3

Although the STAT3 signaling pathway is an upstream mediator of a number of transcription factors, it can be regulated by miRs. Briefly, miRs are endogenous non-coding RNA with a low length of 19–23 nucleotides and can affect various biological processes via targeting different molecular pathways [84,85,86,87,88,89]. A high number of studies have demonstrated that miRs are able to regulate the STAT3 signaling pathway by targeting upstream mediators, such as ROCK1 [59], suppressing translation and phosphorylation [90], affecting JAK proteins [91] and influencing the nuclear translocation of STAT3 [92]. A similar phenomenon occurs in GC cells. It seems that miRs are able to target the STAT3 signaling pathway in GC to affect its metastasis and growth. In this way, miRs are divided into two categories, including onco-suppressor miRs, which reduce the expression of STAT3, while oncogene miRs are capable of enhancing the expression of STAT3. The miR-143 is considered as an onco-suppressor factor in GC, and its overexpression is associated with a decrease in survival and proliferation [93]. This miR can negatively affect both the proliferation and metastasis of GC cells via targeting molecular pathways such as DNMT3A and MYO6 [94,95]. In GC cells, miR-143 diminishes the expression of the STAT3 signaling pathway. Although expression of miR-143 undergoes downregulation in GC cells, enhancing the expression of this miR-143 paves the way for the downregulation of STAT3 and suppressing the invasion and proliferation of GC cells [96]. The miR-125a is another onco-suppressor miR whose downregulation mediates the undesirable prognosis of GC cells [97,98]. There is a reverse relationship between miR-125a and STAT3 in GC cells, as miR-125a reduces the expression of STAT3 to suppress the activation of its downstream target, HAS1, leading to a decrease in the migration and metastasis of GC cells [99]. As mentioned earlier, infection with H. pylori is one of the predisposing factors for GC development. This infection can lead to changes in immune responses and the enhanced production of inflammatory factors [100,101]. Accumulating data demonstrates that H. pylori infection results in the abnormal expression of miRs, which provide the conditions for GC development [102,103,104]. The miR-375 undergoes downregulation by H. pylori infection. Enhancing the expression of miR-375 is considered as a promising strategy in suppressing H. pylori-mediated GC development via the downregulation of the STAT3 signaling pathway. By the inhibition of STAT3, miR-375 suppresses the metastasis (Twist1 downregulation) and proliferation (Bcl-2 downregulation) of GC cells [105].
The miR-148a as an onco-suppressor factor that decreases the growth and invasion of GC cells and is capable of enhancing the sensitivity of GC cells to chemotherapy [106,107]. By the downregulation of the STAT3 signaling pathway, miR-148a suppresses cholecystokinin B receptor (CCK-BR) to interfere with the proliferation and migration of GC cells [108]. In respect to the high metastatic capability of GC cells, revealing the molecular pathways involved in their migration is of interest. Angiogenesis is a molecular mechanism that demonstrates overexpression in cancer cells [109,110]. In fact, based on the high growth and proliferation of cancer cells, they need a high amount of energy and oxygen supplies. By the induction of angiogenesis, cancer cells can promote their ability to proliferate. Besides, angiogenesis can enhance the migration of cancer cells [111,112,113]. Various molecular pathways have been considered as upstream mediators of angiogenesis, and STAT3 is one of them [114]. By the inhibition of STAT3, the induction of angiogenesis is inhibited and the growth of cancer cells is inhibited [115,116]. In GC cells, the STAT3 signaling pathway induces angiogenesis via the upregulation of VEGF. The miR-874 as an onco-suppressor factor that diminishes the expression of STAT3 to disrupt the STAT3/VEGF axis, leading to a decrease in the proliferation and migration of GC cells by the inhibition of angiogenesis [117]. In addition to angiogenesis, the STAT3 signaling pathway can regulate EMT in cancer cells. Increasing evidence demonstrates that the STAT3 pathway is capable of the induction of EMT in cancer cells, and in this way, a number of upstream mediators, such as miR-449b-3p and SIX4, act as inducers of STAT3/EMT [118,119]. The anti-tumor compounds inhibit EMT via STAT3 downregulation [120]. These studies exhibit the critical role of STAT3 in the regulation of EMT in cancer cells. The miR-216a has demonstrated great potential in suppressing the invasion and migration of GC cells via targeting the STAT3 signaling pathway. It seems that miR-216a inhibits the JAK2/STAT3 axis to downregulate EMT, resulting in a decrease in the metastasis of GC cells [121].
In addition to onco-suppressor miRs, the role of oncogene miRs in the regulation of STAT3 in GC has been examined. As mentioned earlier, EMT can enhance the migration of GC cells via transforming epithelial cells into mobile mesenchymal ones [122,123]. The matrix metalloproteinase proteins (MMPs) are also capable of promoting the metastasis of cancer cells via the degradation of the extracellular matrix (ECM) [124]. MMP-9 is a key member of this family and its role in GC has been investigated. It seems that MMP-9 upregulation enhances the metastasis of GC cells and is associated with an unfavorable prognosis [125,126]. The STAT3 signaling pathway induces MMP-9 to elevate the progression and metastasis of GC cells. The oncogene miR-93-5p inhibits STAT3 via IFNAR1 downregulation, leading to a decrease in MMP-9 expression and the malignant behavior of GC cells [127]. Notably, miRs are able to target endogenous inhibitors of the STAT3 signaling pathway, such as PIAS3, in cancer cells [128,129]. The miR-18a is suggested to be an oncogene factor in cancer and its downregulation can pave the way to suppressing cancer malignancy [113,130]. In GC cells, miR-18a down-regulates PIAS3 to induce the STAT3 signaling pathway. As a consequence, downstream targets of STAT3 including, c-Myc, Survivin and Bcl-xl, undergo upregulation that ensures the viability and proliferation of cancer cells [131]. Overall, studies are in agreement with the fact that miRs are potential upstream mediators of the STAT3 signaling pathway in GC and the modulation of the miR/STAT3 axis can lead to effective GC therapy (Table 3, Figure 2 and Figure 3).

5.2. Drug-Mediated Regulation of STAT3

Based on the role of the STAT3 signaling pathway in enhancing the progression and malignancy of GC cells, much attention has been directed towards the regulation and targeting of this pathway in GC therapy. Anti-tumor compounds are able to target STAT3 in GC therapy [134,135]. It is worth mentioning that, to date, most of the anti-tumor drugs applied in the treatment of GC cells by targeting STAT3 have been isolated from plants. The plant-derived natural compounds have demonstrated great potential in the regulation of the STAT3 signaling pathway in cancer therapy [136]. In GC therapy, the natural products capable of targeting the STAT3 signaling pathway have been applied. In Table 4, we summarize these anti-tumor compounds, and this section, we describe their ability in suppressing GC malignancy and proliferation. Cucurbitacins are anti-tumor compounds and well known in traditional Chinese medicine [137]. These plant-derived natural compounds have anti-tumor activity against GC cells and are able to suppress the growth and viability of GC cells via the induction of cell cycle arrest [138]. A newly published article has shed some light on the anti-tumor activity of cucurbitacins in GC cells. It seems that the STAT3 signaling pathway ensures the growth and survival of GC cells via the induction of Bcl-xl and c-Myc upregulation. The administration of cucurbitacin B is correlated with the downregulation of STAT3 and its downstream targets. In the inhibition of STAT3 activity, cucurbitacin B attaches to the DNA-binding domain of STAT3. This decrease in STAT3 activity and expression caused by cucurbitacin B paves the way for a reduction in the proliferation of GC cells and their sensitization into the anti-tumor activity of cisplatin as a chemotherapeutic agent [126]. This study highlights the fact that natural products can be used as chemosensitizers in GC therapy. The examination of molecular pathways demonstrates that anti-tumor compounds are able to enhance the generation of reactive oxygen species (ROS) to modulate the expression of STAT3. In fact, phytochemicals induce ROS production to stimulate apoptotic cell death via the downregulation of STAT3 phosphorylation [139]. This strategy is not only beneficial in the induction of apoptosis in GC cells, but it can also mediate the stimulation of cell cycle arrest. After the administration of glycitein as an anti-tumor agent, GC cells undergo cell cycle arrest at the G0/G1 phase. This anti-tumor activity partially emanates from the inhibitory effect of glycitein on the expression of STAT3 [140]. However, natural products with anti-tumor activity can reduce the viability of GC cells via the reduction of ROS levels. In fact, they are not just dependent on the induction of ROS generation. This is due to the dual role of ROS in cancer cells. Although ROS can mediate the mitochondrial dysfunction and stimulation of endoplasmic reticulum (ER) stress [141,142,143], accumulating data demonstrates that ROS can lead to tumorigenesis via the activation of oncogene signaling pathways, such as STAT3 and Wnt/β-catenin [144,145]. In these cases, decreasing ROS production can pave the way to effective cancer therapy. A similar strategy is used by lycopene as a potential anti-tumor agent [146]. In suppressing GC development, lycopene reduces ROS generation that in turn inhibits the STAT3 signaling pathway, as a carcinogenesis factor [147].
In cancer cells, IL-6 functions as an upstream mediator of STAT3 signaling pathway. It has been demonstrated that IL-6 can induce STAT3 to ensure the malignant behavior of cancer cells [148]. Apigetrin (APG) is a flavonoid compound with excellent anti-tumor activity [149]. In GC cells, the administration of APG induces apoptosis and remarkably diminishes their growth and proliferation. The examination of molecular pathways demonstrates that APG inhibits STAT3 through IL-6 downregulation. Besides, APG triggers the dephosphorylation of JAK2/STAT3 [150], making it a suitable compound in GC therapy. NF-kB is an oncogene signaling pathway responsible for the enhanced proliferation and migration of cancer cells [151]. Increasing evidence demonstrates that there is a dual relationship between NF-kB and STAT3 through the p65 and p50 subunits [152,153]. Targeting the STAT3/NF-kB axis is a potential strategy in cancer therapy [154]. Troxerutin (TXN) is a natural flavonoid rutin with different pharmacological activities, such as anti-diabetes, hepatoprotective, neuroprotective, antioxidant and anti-inflammatory [155,156]. Newly recorded studies are in line with the fact that TXN is capable of affecting molecular pathways, such as NF-kB and MDM2, in cancer therapy [157,158]. TXN supplementation is associated with a decrease in the survival of GC cells and their sensitivity to 5-fluorouracil chemotherapy. It is said that TXN is able to suppress STAT3 phosphorylation, which subsequently reduces the expression of NF-KB, leading to the decreased viability and growth of GC cells [159].
In previous section, we mentioned that miRs can function as upstream regulators of the STAT3 signaling pathway in cancer cells. The accumulated data demonstrate that the STAT3 signaling pathway can also regulate miR expression as an upstream mediator [160,161]. This crosstalk is of importance in cancer cells. The miR-373 is an oncogene factor capable of increasing the malignancy and proliferation of cancer cells via targeting molecular pathways. This miR dually promotes metastasis and proliferation, and its inhibition can remarkably suppress cancer malignancy, leading to their sensitivity to chemotherapy [162,163,164]. Exposing GC cells to isoproterenol sensitizes GC cells to cell death. Isoproterenol inhibits STAT3 phosphorylation to suppress miR-373 expression, resulting in the inhibition of drug resistance and metastasis via the upregulation of E-cadherin [165]. As discussed earlier, H. pylori can cause a predisposition to GC. It has been reported that H. pylori stimulates the STAT3 signaling pathway in GC development. The administration of docosahexaenoic acid (DHA) induces peroxisome proliferator-activated receptor gamma (PPAR-γ) to inhibit the phosphorylation of STAT3 at tyrosine 705. Besides, DHA enhances the expression of SOCS3 and suppresses the nuclear translocation of STAT3, resulting in a decrease in the proliferation and invasion of GC cells [166]. Taking everything into account, studies are in agreement with the fact that anti-tumor compounds are able to inhibit STAT3 in different stages, including targeting upstream mediators, the activation of endogenous inhibitors, the downregulation of downstream targets and suppressing STAT3 expression [167,168,169,170,171,172,173,174].

5.3. LncRNA-Mediated Regulation of STAT3

The lncRNAs are key members of non-coding RNAs with lengths more than 200 nucleotides [196]. Similar to miRs, lncRNAs are able to affect and regulate a number of biological mechanisms, such as cell proliferation, differentiation, angiogenesis, migration and so on [197,198]. These modulatory effects of lncRNAs have led to the investigation of their roles in different diseases, particularly cancer [199,200]. Newly published studies have shown that lncRNAs are able to target molecular pathways by the induction of their effects [201,202]. It is worth mentioning that a large body of evidence has examined the relationship between lncRNAs and the STAT3 signaling pathway [203,204]. The oncogene lncRNAs are able to upregulate the expression of STAT3, while onco-suppressor lncRNAs reduce the expression of STAT3 [205,206,207,208]. It is held that targeting the lncRNA/STAT3 axis is of importance in cancer therapy [209,210]. Fortunately, experiments have attempted to provide information about the dual relationship between lncRNAs and STAT3 in GC cells, and it has been shown that not only can lncRNAs function as upstream regulators of STAT3, but also STAT3 can affect the expression of lncRNAs [211,212]. The identification of this feedback is of importance in effective GC therapy. To date, just oncogene lncRNAs targeting STAT3 and their regulation have been investigated in GC. The lncRNA SNHG16 is an oncogene factor that is correlated with the invasion and growth of cancer cells. This lncRNA enhances the migration of cancer cells via the induction of EMT. Besides, the lncRNA SNHG16 regulates miRs in exerting their stimulatory effect on cancer cells [213,214,215]. In GC cells, SNHG16 considerably reduces the expression of the onco-suppressor miR-135a to activate the JAK2/STAT3 signaling pathway. This leads to an increase in colony formation and the growth of GC cells and reduces their sensitivity to apoptotic cell death [195]. Based on the role of oncogene lncRNAs in promoting the malignant behavior of GC cells, their modulation can pave the way into effective cancer therapy. It is held that the downregulation of the lncRNA HOTAIR, as an oncogene factor, enhances the expression of miR-454-3p. This miR is able to negatively affect the survival of GC cells by the induction of apoptosis and cell cycle arrest partially via the inhibition of the STAT3/cyclin D1 axis [216]. As mentioned earlier, STAT3 can induce angiogenesis via targeting VEGF. This results in an increase in the proliferation and viability of cancer cells [217,218]. It is held that lncRNAs can regulate the STAT3/VEGF axis in GC cells to affect angiogenesis and their proliferation. The lncRNA PVT1 is an oncogene lncRNA that induces angiogenesis via the activation of the STAT3/VEGF axis. This axis and the stimulation of angiogenesis are positive factors for the enhanced proliferation and migration of GC cells [219]. These studies demonstrate that lncRNAs indirectly affect the STAT3 signaling pathway by targeting their upstream mediators, such as miR-506 and IL-6. It is worth mentioning that the dual relationship between lncRNAs and the STAT3 signaling pathways can lead to the increased malignant behavior of GC cells, since they can increase their expression in a positive feedback loop [220,221]. Further studies should focus on revealing more oncogene lncRNAs, and also onco-suppressor lncRNAs. Besides, the genetic or pharmacological targeting of lncRNAs can pave the way to effective GC therapy (Table 5, Figure 4).

5.4. Other Molecular Signaling Pathways Regulate STAT3

In previous sections, we provided explanations about the role of upstream mediators, such as miRs and lncRNAs, in the regulation of the STAT3 signaling pathway in GC cells. Besides, we demonstrated that anti-tumor compounds are able to target the STAT3 signaling pathway in suppressing GC proliferation and malignancy. In this section, we discuss the other molecular pathways capable of targeting the STAT3 signaling pathway in GC cells. The identification of these signaling pathways and their upstream and downstream targets can pave the way to effective GC therapy. Sirtuins are a family of histone deacetylases and their function relies on NAD+. This family consists of seven families (SIRT1-7) and they are able to modulate different biological mechanisms, including cell metabolism, cell division and aging [224]. Increasing evidence demonstrates that SIRT1 is a positive factor for the progression and growth of GC cells. It has been reported that SIRT1 upregulation is correlated with reduced survival and undesirable prognosis [225]. However, another study exhibits that SIRT1 suppresses GC growth and stimulates apoptosis and cell cycle arrest in GC cells [226]. In GC cells, the STAT3 signaling pathway increases the invasion and migration of cancer cells via the stimulation of MMP-13. It is held that SIRT1 downregulates STAT3 to inhibit MMP-19 expression, resulting in the decreased invasion and malignancy of GC cells [227]. SIRT6 is another member of the SIRT family and is capable of reducing the proliferation of GC cells. The previous study revealed that SIRT1 exerts an inhibitory effect on the migration and metastasis of GC cells. It appears that SIRT6 negatively affects the proliferation and growth of GC cells by the inhibition of the STAT3 signaling pathway and the subsequent downregulation of cyclin D1 and Bcl-2 [228].
Stathmin (STMN) is a microtubule-regulating protein capable of the regulation of mitosis via targeting the aggregation and depolymerization of spindles. The STMN is an oncogene factor that undergoes upregulation in different tumors to ensure their proliferation and viability. Besides, STMN can be considered as a potential factor for the diagnosis of cancer [229,230,231]. The relationship between STMN and the STAT3 signaling pathway is of importance in GC cells. It has been reported that STMN dually enhances the migration and growth of GC cells. The knock-down of STMN reduces the expression of STAT3, resulting in cell cycle arrest and apoptosis in GC cells [232]. The uncontrolled proliferation of cancer cells requires a vast source of energy. Increasing evidence demonstrates that cancer cells enhance their glucose metabolism to meet their energy needs. This is known as the Warburg effect [233,234]. The examination of molecular pathways reveals interesting pathways involved in the Warburg effect of GC cells. PKM2 and c-Myc are considered as factors involved in the glycolysis of GC cells. These factors provide the mild acidic pH of the tumor microenvironment and promote glucose metabolism to provide for the proliferation of GC cells. STAT3 acts as an upstream mediator of c-Myc, while mTOR is the upstream mediator of PKM2. More importantly, it seems that the STAT3/c-Myc and mTOR/PKM2 signaling pathways have positive feedback and together they can lead to the enhanced proliferation and energy metabolism of GC cells [235]. The downregulation of the aforementioned signaling networks can result in the growth inhibition of GC cells.
RNF6 is a member of the E3 ligase family and its role in cancer has been explored. This oncogene factor is able to promote the growth and viability of cancer cells via the upregulation of pro-survival factors, such as Bcl-xl and Mcl-1 [236]. The clinical trials have also confirmed the role of RNF6 in the malignancy of cancer cells. It has been shown that RNF6 overexpression can mediate the metastasis and migration of colorectal cancer cells [237]. So, recognizing the downstream targets of RNF6 is of importance in cancer therapy. There is a dual relationship between RNF6 and the STAT3 signaling pathway in GC cells. RNF6 induces the STAT3 signaling pathway to upregulate the expression of pro-survival factors, such as XIAP and Mcl-1, resulting in an increase in the growth and survival of GC cells [238]. The signaling networks not only ensure the survival and proliferation of GC cells, but they can also trigger the resistance of GC cells to chemotherapy. Sphingosine-1-phosphate receptor (S1PR1) is a member of the G protein-coupled receptor family that is able to promote the malignant behavior of cancer cells. Onco-suppressor factors, such as miR-125b-1-3p, downregulate the expression of S1PR1 to suppress the migration and invasion of cancer cells and induce apoptosis [239]. It is held that the overexpression of S1PR1 is associated with chemoresistance [240]. The relationship between S1PR1 and STAT3 mediates the resistance of GC cells to chemotherapy. The inhibition of the S1PR1/STAT3 axis is correlated with the sensitization of GC cells to chemotherapy [241]. There is evidence that the inhibition of two signaling pathways is absolutely efficient in suppressing the chemoresistance of GC cells. Y-box binding protein-1 (YB-1) is capable of mediating the chemoresistance of cancer cells [242]. On the other hand, increasing evidence has demonstrated that the STAT3 signaling pathway is involved in chemoresistance [243]. In GC cells, the inhibition of STAT3 and YB-1 can suppress the resistance of cancer cells to chemotherapy. Although there is no dual relationship between STAT3 and YB-1, it has been reported that their simultaneous inhibition can result in synergistic effects in sensitizing GC cells to chemotherapy [244]. It is worth mentioning that, in addition to chemoresistance, the STAT3 signaling pathway may trigger the immune evasion of cancer cells. The PD-1/PD-L1 axis undergoes overexpression in malignant tumors and it can be regulated by different factors, such as miRs, lncRNAs, transcription factors and so on [245,246,247,248]. The enhanced expression of PD-1 provides the conditions for the resistance of cancer cells to chemotherapy and is correlated with undesirable prognosis [249,250,251]. It is said that GC mesenchymal stem cells are able to elevate levels of IL-8. IL-8 functions as an upstream mediator and upregulates the expression of STAT3. The STAT3 signaling pathway induces c-Myc to activate the PD-1/PD-L1 axis, leading to immunosuppression and the increased malignant behavior of GC cells [252].
TMEM119 is a member of the transmembrane proteins with important functions in cancer cells. TMEM119 provides the unfavorable prognosis of a patient with prostate cancer [253]. As a consequence, targeting TMEM119 is a potential strategy in overcoming cancer [254]. A same story occurs in GC cells. It is held that TMEM119 enhances the invasion and metastasis of GC cells via the upregulation of the STAT3 signaling pathway [255]. The inhibition of TMEM119 paves the way to GC treatment. It seems that STAT3 can trigger EMT in GC cells. However, upstream mediators are able to affect the STAT3/EMT axis. Nuclear factor I/B (NFIB) participates in normal somatic development and recent studies have revealed its role in cancer progression and development [256,257]. NFIB is able to induce Akt phosphorylation. Subsequently, an increase occurs in the expression of STAT3, resulting in the activation of EMT by enhancing vimentin levels and decreasing E-cadherin levels [258]. A same phenomenon occurs during inflammation. Increasing evidence shows that chronic inflammation can remarkably promote the progression and malignancy of cancer cells [259,260]. IL-23 is one of the pro-inflammatory cytokines and it has been reported that IL-23 induces the EMT mechanism through STAT3 upregulation to ensure the migration and metastasis of GC cells [261].
The tumor microenvironment (TME) is a complicated structure, consisting of different cells that have been included in the ECM [262]. Fibroblasts, endothelial cells, pericytes, immune cells and inflammatory cells are the main components of the TME. The bidirectional communication among these cells plays a significant role in cancer progression and malignancy [263]. Cancer-associated macrophages (CAMs) are M2 phenotype macrophages and an increase in their number is associated with poor prognosis in different cancers, particularly GC [264,265,266,267]. The CAMs are able to secrete IL-10 that in turn activates the c-Met signaling pathway. The c-Met acts as an upstream mediator of STAT3, resulting in the enhanced proliferation and invasion of GC cells [268]. Tumor-associated macrophages (TAMs) are one of the most important and abundant components of the TME. These macrophages have tumor-promoting effects (M2 phenotype macrophage) and are CD68-marked [269,270,271,272]. The TAMs enhance the levels of pro-inflammatory cytokines, including IL-6 and IL-8, to activate the STAT3 signaling pathway, leading to the metastasis and progression of GC cells [273]. Experiments also demonstrate that pro-inflammatory cytokines in the TME can provide the conditions for the differentiation of macrophages into the M2 phenotype that exerts a stimulatory effect on the proliferation and progression of cancer cells. IL-6 enhances levels of M2 macrophages with a high expression of IL-10 and TGF-β. It is held that the effect of IL-6 on macrophage differentiation is mediated through STAT3 upregulation. This axis leads to a remarkable increase in the progression and malignancy of GC cells [274]. More importantly, the TME can trigger the resistance of cancer cells to chemotherapy and immunotherapy. This has resulted in considerable attention to role of the TME in cancer malignancy [275,276,277]. Cancer-associated fibroblasts (CAFs) are another key member of the TME that can induce the metastasis, growth and malignancy of cancer cells [278]. In GC cells, CAFs secrete IL-11 that in turn activates the JAK/STAT3 signaling pathway. This leads to an increase in expression of anti-apoptotic factor Bcl-2 to mediate the resistance of GC cells to chemotherapy [279].
Taking everything into account, studies are in agreement with the fact that different signaling pathways can function as upstream mediators in the induction of STAT3 in GC cells. The identification of these pathways and further targeting can pave the way to effective GC therapy [280,281,282,283,284]. Besides, we demonstrated that different components of the TME are able to secrete inflammatory cytokines to activate STAT3, ensuring the metastasis and invasion of GC cells (Table 6).

6. STAT3 as an Oncogene Factor in Gastric Cancer

In previous sections, it was revealed that lncRNAs, miRs and other molecular pathways are able to regulate the STAT3 signaling pathway in GC cells. Besides, we described how anti-tumor compounds are able to modulate STAT3. In this section, we demonstrate that STAT3 can function as an upstream mediator to affect other molecular pathways, leading to the malignant behavior of GC cells [304,305]. DNA methylation and histone modification are hallmarks of carcinogenesis and they are able to influence the transcription output of the genome [306,307]. It is held that the presence of p-H3S10 and/or H3S28ph in the promoter regions can accelerate histone modification [308]. In GC cells, the STAT3 signaling pathway stimulates epigenetic kinase mitogen- and stress-activated protein kinase 1 (MSK1) to induce H3S10 phosphorylation, resulting in an increase in the tumorigenesis of GC cells [309]. A recently recorded study shows that the STAT3 signaling pathway can cause the poor differentiation of GC cells and is correlated with their metastasis into distant tissues [310]. Interestingly, downstream targets of the STAT3 signaling pathway can be considered as biomarkers for the early detection of GC. STAT3 is able to inhibit SPG20 expression via hypermethylation and the downregulation of this factor is a potential biomarker for GC detection [311]. It is worth mentioning that polymorphisms in STAT3 are correlated with GC tumorigenesis. It appears that the existence of a minor allele of STAT3 (rs1053023) is associated with a risk of GC development [312]. These studies demonstrate that not only downstreams of STAT3 can be used as biomarkers for GC detection, but also own STAT3 is a potential biomarker in GC identification. The clinical studies are also in line with the benefit of using STAT3 as a diagnostic factor. It has been reported that a positive feedback loop between STAT3 and miR-200 can promote the progression of GC cells, and this feedback is of importance for the diagnosis and prognosis of GC [313]. Overall, the overexpression of STAT3 provides the poor prognosis of patients with GC and its expression undergoes upregulation at the point when the gastric mucosa reaches the tumor stage [314,315]. In enhancing the growth and viability of GC cells, STAT3 targets cell cycle proteins. STAT3 is able to induce cyclin D1 expression, resulting in an increase in the proliferation and malignancy of GC cells [316].
CD163 is called a macrophage-associated antigen and is abundantly expressed in monocytes and macrophages [317]. CD163 has physiological functions, such as iron metabolism and the endocytosis of hemoglobin–haptoglobin complexes [318]. In spite of these vital functions in normal conditions, increasing evidence demonstrates that CD163 may be involved in the emergence of different disorders, particularly cancer [319,320,321]. STAT3 enhances the expression of CD163 on the components of the TME, such as macrophages, to enhance the proliferation of GC cells [322]. Targeting CD163 can be considered as a potential strategy in effective GC therapy. CD44 is another factor that can be targeted in GC cells by the STAT3 signaling pathway. CD44 is a cell adhesion molecule and this trans-membrane glycoprotein increases the proliferation and metastasis of cancer cells by binding to hyaluronic acid [323,324,325]. Accumulating data demonstrates that CD44 is a cell surface marker of cancer stem cells and is of importance for the malignancy of GC cells [324,325,326,327,328,329]. CD44 undergoes upregulation by STAT3 in GC cells and provides the undesirable prognosis of patients with GC [292].
Enhancer of zeste homologue 2 (EZH2) is a key member of the polycomb group genes and is able to promote the malignancy of cancer cells via inhibiting the expression of a variety of tumor suppressor genes (TSGs) [330]. EZH2 dually enhances the migration and proliferation of cancer cells, and targeting this oncogene factor is a promising strategy in cancer therapy [331,332]. In GC cells, STAT3 functions as an upstream mediator of EZH2. By the activation of EZH2, STAT3 elevates the proliferation and metastasis of GC cells, and is correlated with poor prognosis [333]. In respect to the oncogene role of STAT3 in GC, studies have focused on investigating the expression of STAT3 in GC cells. It seems that the expression of STAT3 undergoes overexpression in gastric stromal tumors, and this can be considered as a diagnostic and prognostic factor (Figure 5) [334,335,336,337,338,339,340].

7. Conclusions and Remarks

The STAT3 signaling pathway is a well-known oncogene factor in different cancers, and its involvement in the malignancy and growth of GC cells has been extensively investigated. The present review focused on revealing the downstream and upstream mediators of STAT3 in GC cells to pave the way to understanding the oncogene pathways in this malignant tumor. Oncogene miRs and lncRNAs induce the STAT3 pathway, while onco-suppressor ones inhibit the STAT3 signaling pathway. We devoted a section to examining the relationship between anti-tumor compounds and the STAT3 signaling pathway in GC cells. It was revealed that anti-tumor agents are able to suppress STAT3, resulting in a decrease in the proliferation and invasion of GC cells. One of the interesting points about the STAT3 signaling pathway is that inflammatory factors can act as upstream mediators of STAT3. This relationship is of importance in the tumor microenvironment, since present cells are able to secrete interleukins that in turn activate the STAT3 signaling pathway, leading to the elevated progression and malignancy of GC cells. Finally, we demonstrated that STAT3 can be considered as a diagnostic and prognostic factor in GC. Although studies have extensively examined the molecular pathways involved in the STAT3 regulation in GC, and how anti-tumor compounds can be beneficial in suppressing STAT3 in GC therapy, there are a number of drawbacks that should be considered in further studies. Small interfering RNA (siRNA) has been applied in the inhibition of STAT3 and improving the prognosis of GC [341]. However, the off-targeting of siRNA, and also its degradation have limited its efficacy. On the other hand, anti-tumor compounds and their ability in GC therapy should be improved, since they have low bioavailability, and there are impediments to their entrance into cancer cells, such as the blood–tumor barrier. Recently, nanoparticles have gained significant attention in cancer therapy [342,343]. Using nanoparticles for the encapsulation of siRNA and anti-tumor compounds can facilitate the way to effective GC therapy, as the efficacy of siRNA-loaded nanoparticles improves gene silencing and the high cellular uptake of anti-tumor compounds is observed after nanoparticle delivery.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

GCgastric cancer
H. pyloriHelicobacter pylori
EBVEpstein–Barr virus
lncRNAslong non-coding RNAs
miRmicroRNA
EMTepithelial-to-mesenchymal transition
STATsignal transducer and activator of transcription
SH2Src homology-2
TADtranscription activation domain
JAKsJanus kinases
SOCSsuppressor of cytokine signaling
PIASprotein inhibitor of activated STAT
SLP-2stomatin-like protein 2
CTMChinese traditional medicine
VEGFvascular endothelial growth factor
MMPsmatrix metalloproteinases
ECMextracellular matrix
ROSreactive oxygen species
ERendoplasmic reticulum
APGapigetrin
TXNtroxerutin
DHAdocosahexaenoid acid
PPAR-γperoxisome proliferator-activated receptor gamma
SIRTsirtuin
STMNstathmin
S1PR1sphingosine-1-phosphate receptor
YB-1Y-box binding protein-1
NFIBnuclear factor I/B
TMEtumor microenvironment
CAMscancer-associated macrophages
TAMstumor-associated macrophages
CAFscancer-associated fibroblasts
MSK1mitogen- and stress-activated protein kinase 1
EZH2enhancer of zeste homolog 2

References

  1. Arai, H.; Nakajima, T.E. Recent Developments of Systemic Chemotherapy for Gastric Cancer. Cancers 2020, 12, 1100. [Google Scholar] [CrossRef] [PubMed]
  2. Magnelli, L.; Schiavone, N.; Staderini, F.; Biagioni, A.; Papucci, L. MAP Kinases Pathways in Gastric Cancer. Int. J. Mol. Sci. 2020, 21, 2893. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Ashrafizadeh, M.; Rafiei, H.; Mohammadinejad, R.; Farkhondeh, T.; Samarghandian, S. Wnt-regulating microRNAs role in gastric cancer malignancy. Life Sci. 2020, 250, 117547. [Google Scholar] [CrossRef] [PubMed]
  4. Colquhoun, A.; Arnold, M.; Ferlay, J.; Goodman, K.; Forman, D.; Soerjomataram, I. Global patterns of cardia and non-cardia gastric cancer incidence in 2012. Gut 2015, 64, 1881–1888. [Google Scholar] [CrossRef]
  5. De Martel, C.; Forman, D.; Plummer, M. Gastric cancer: Epidemiology and risk factors. Gastroenterol. Clin. 2013, 42, 219–240. [Google Scholar] [CrossRef]
  6. Yoo, J.Y.; Cho, H.J.; Moon, S.; Choi, J.; Lee, S.; Ahn, C.; Yoo, K.-Y.; Kim, I.; Ko, K.-P.; Lee, J.E. Pickled Vegetable and Salted Fish Intake and the Risk of Gastric Cancer: Two Prospective Cohort Studies and a Meta-Analysis. Cancers 2020, 12, 996. [Google Scholar] [CrossRef]
  7. Riihimäki, M.; Hemminki, A.; Sundquist, K.; Sundquist, J.; Hemminki, K. Metastatic spread in patients with gastric cancer. Oncotarget 2016, 7, 52307. [Google Scholar] [CrossRef] [Green Version]
  8. Baj, J.; Brzozowska, K.; Forma, A.; Maani, A.; Sitarz, E.; Portincasa, P. Immunological Aspects of the Tumor Microenvironment and Epithelial-Mesenchymal Transition in Gastric Carcinogenesis. Int. J. Mol. Sci. 2020, 21, 2544. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Huang, H.G.; Tang, X.L.; Huang, X.S.; Zhou, L.; Hao, Y.G.; Zheng, Y.F. Long noncoding RNA LINC00511 promoted cell proliferation and invasion via regulating miR-124-3p/EZH2 pathway in gastric cancer. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 4232–4245. [Google Scholar] [CrossRef]
  10. Chen, P.; Qian, X.K.; Zhang, Y.F.; Sun, X.G.; Shi, X.J.; Gao, Y.S. KLF5 promotes proliferation in gastric cancer via regulating p21 and CDK4. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 4224–4231. [Google Scholar] [CrossRef]
  11. El-Guindy, D.M.; Wasfy, R.E.; Abdel Ghafar, M.T.; Ali, D.A.; Elkady, A.M. Oct4 expression in gastric carcinoma: Association with tumor proliferation, angiogenesis and survival. J. Egypt. Natl. Cancer Inst. 2019, 31, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Zhang, Z.; Hu, X.; Kuang, J.; Liao, J.; Yuan, Q. LncRNA DRAIC inhibits proliferation and metastasis of gastric cancer cells through interfering with NFRKB deubiquitination mediated by UCHL5. Cell. Mol. Biol. Lett. 2020, 25, 29. [Google Scholar] [CrossRef] [PubMed]
  13. Jiang, X.; Wang, J.; Fang, L. LncRNA WT1-AS over-expression inhibits non-small cell lung cancer cell stemness by down-regulating TGF-beta1. BMC Pulm. Med. 2020, 20, 113. [Google Scholar] [CrossRef]
  14. Sun, C.B.; Wang, H.Y.; Han, X.Q.; Liu, Y.N.; Wang, M.C.; Zhang, H.X.; Gu, Y.F.; Leng, X.G. LINC00511 promotes gastric cancer cell growth by acting as a ceRNA. World J. Gastrointest. Oncol. 2020, 12, 394–404. [Google Scholar] [CrossRef]
  15. Xu, J.; Zhang, Y.; You, Q.; Fu, H.; Zhao, X.; Lu, K.; Yan, R.; Yang, D. LncRNA PTCSC3 Alleviates the Postoperative Distant Recurrence of Gastric Cancer by Suppression of lncRNA HOXA11-AS. Cancer Manag. Res. 2020, 12, 2623–2629. [Google Scholar] [CrossRef] [Green Version]
  16. Loh, C.-Y.; Chai, J.Y.; Tang, T.F.; Wong, W.F.; Sethi, G.; Shanmugam, M.K.; Chong, P.P.; Looi, C.Y. The E-Cadherin and N-Cadherin Switch in Epithelial-to-Mesenchymal Transition: Signaling, Therapeutic Implications, and Challenges. Cells 2019, 8, 1118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Mohammadinejad, R.; Biagioni, A.; Arunkumar, G.; Shapiro, R.; Chang, K.-C.; Sedeeq, M.; Taiyab, A.; Hashemabadi, M.; Pardakhty, A.; Mandegary, A.; et al. EMT signaling: Potential contribution of CRISPR/Cas gene editing. Cell. Mol. Life Sci. 2020. [Google Scholar] [CrossRef]
  18. Cheng, J.-T.; Wang, L.; Wang, H.; Tang, F.-R.; Cai, W.-Q.; Sethi, G.; Xin, H.-W.; Ma, Z. Insights into Biological Role of LncRNAs in Epithelial-Mesenchymal Transition. Cells 2019, 8, 1178. [Google Scholar] [CrossRef] [Green Version]
  19. Ma, B.; Ma, J.; Yang, Y.; He, X.; Pan, X.; Wang, Z.; Qian, Y. Effects of miR-330-3p on Invasion, Migration and EMT of Gastric Cancer Cells by Targeting PRRX1-Mediated Wnt/beta-Catenin Signaling Pathway. OncoTargets Ther. 2020, 13, 3411–3423. [Google Scholar] [CrossRef] [Green Version]
  20. Nakayama, Y.; Mimura, K.; Kua, L.F.; Okayama, H.; Min, A.K.T.; Saito, K.; Hanayama, H.; Watanabe, Y.; Saito, M.; Momma, T.; et al. Immune suppression caused by PD-L2 expression on tumor cells in gastric cancer. Gastric Cancer J. Int. Gastric Cancer Assoc. Jpn. Gastric Cancer Assoc. 2020. [Google Scholar] [CrossRef]
  21. Liu, H.T.; Ma, R.R.; Lv, B.B.; Zhang, H.; Shi, D.B.; Guo, X.Y.; Zhang, G.H.; Gao, P. LncRNA-HNF1A-AS1 functions as a competing endogenous RNA to activate PI3K/AKT signalling pathway by sponging miR-30b-3p in gastric cancer. Br. J. Cancer 2020, 122, 1825–1836. [Google Scholar] [CrossRef] [PubMed]
  22. Qiu, L.; Lu, F.; Zhang, L.; Wang, G.; Geng, R.; Miao, Y. HBXIP Regulates Gastric Cancer Glucose Metabolism and Malignancy Through PI3K/AKT and p53 Signaling. Oncotargets Ther. 2020, 13, 3359–3374. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Liu, Y.; Chen, H.; Dong, P.; Xie, G.; Zhou, Y.; Ma, Y.; Yuan, X.; Yang, J.; Han, L.; Chen, L.; et al. KIF23 activated Wnt/beta-catenin signaling pathway through direct interaction with Amer1 in gastric cancer. Aging 2020, 12, 8372–8396. [Google Scholar] [CrossRef]
  24. Zhou, C.; Guo, Z.; Xu, L.; Jiang, H.; Sun, P.; Zhu, X.; Mu, X. PFND1 Predicts Poor Prognosis of Gastric Cancer and Promotes Cell Metastasis by Activating the Wnt/beta-Catenin Pathway. OncoTargets Ther. 2020, 13, 3177–3186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Kim, B.J.; Kim, Y.J.; Sohn, S.H.; Kim, B.; Sul, H.J.; Kim, H.S.; Zang, D.Y. Tivantinib inhibits the VEGF signaling pathway and induces apoptosis in gastric cancer cells with c-MET or VEGFA amplification. Investig. New Drugs 2020. [Google Scholar] [CrossRef] [PubMed]
  26. Zhong, X.; Liu, D.; Jiang, Z.; Li, C.; Chen, L.; Xia, Y.; Liu, D.; Yao, Q.; Wang, D. Chrysin Induced Cell Apoptosis and Inhibited Invasion Through Regulation of TET1 Expression in Gastric Cancer Cells. OncoTargets Ther. 2020, 13, 3277–3287. [Google Scholar] [CrossRef] [Green Version]
  27. Jin, L.; Ma, X.M.; Wang, T.T.; Yang, Y.; Zhang, N.; Zeng, N.; Bai, Z.G.; Yin, J.; Zhang, J.; Ding, G.Q.; et al. Psoralen Suppresses Cisplatin-Mediated Resistance and Induces Apoptosis of Gastric Adenocarcinoma by Disruption of the miR196a-HOXB7-HER2 Axis. Cancer Manag. Res. 2020, 12, 2803–2827. [Google Scholar] [CrossRef] [Green Version]
  28. Mohan, C.D.; Rangappa, S.; Preetham, H.D.; Nayak, S.C.; Gupta, V.K.; Basappa, S.; Sethi, G.; Rangappa, K.S. Targeting STAT3 signaling pathway in cancer by agents derived from Mother Nature. In Seminars in Cancer Biology; Elsevier: Amsterdam, The Netherlands, 2020. [Google Scholar]
  29. Ma, J.H.; Qin, L.; Li, X. Role of STAT3 signaling pathway in breast cancer. Cell Commun. Signal. CCS 2020, 18, 33. [Google Scholar] [CrossRef] [Green Version]
  30. Qin, J.; Shen, X.; Zhang, J.; Jia, D. Allosteric inhibitors of the STAT3 signaling pathway. Eur. J. Med. Chem. 2020, 190, 112122. [Google Scholar] [CrossRef]
  31. Wu, C.J.; Sundararajan, V.; Sheu, B.C.; Huang, R.Y.; Wei, L.H. Activation of STAT3 and STAT5 Signaling in Epithelial Ovarian Cancer Progression: Mechanism and Therapeutic Opportunity. Cancers 2019, 12, 24. [Google Scholar] [CrossRef] [Green Version]
  32. Orlova, A.; Wagner, C.; de Araujo, E.D.; Bajusz, D.; Neubauer, H.A.; Herling, M.; Gunning, P.T.; Keseru, G.M.; Moriggl, R. Direct Targeting Options for STAT3 and STAT5 in Cancer. Cancers 2019, 11, 1930. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Mohrherr, J.; Uras, I.Z.; Moll, H.P.; Casanova, E. STAT3: Versatile Functions in Non-Small Cell Lung Cancer. Cancers 2020, 12, 1107. [Google Scholar] [CrossRef] [PubMed]
  34. Zouein, F.A.; Altara, R.; Chen, Q.; Lesnefsky, E.J.; Kurdi, M.; Booz, G.W. Pivotal importance of STAT3 in protecting the heart from acute and chronic stress: New advancement and unresolved issues. Front. Cardiovasc. Med. 2015, 2, 36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Chai, E.Z.P.; Shanmugam, M.K.; Arfuso, F.; Dharmarajan, A.; Wang, C.; Kumar, A.P.; Samy, R.P.; Lim, L.H.; Wang, L.; Goh, B.C. Targeting transcription factor STAT3 for cancer prevention and therapy. Pharmacol. Ther. 2016, 162, 86–97. [Google Scholar] [CrossRef]
  36. Timofeeva, O.A.; Tarasova, N.I.; Zhang, X.; Chasovskikh, S.; Cheema, A.K.; Wang, H.; Brown, M.L.; Dritschilo, A. STAT3 suppresses transcription of proapoptotic genes in cancer cells with the involvement of its N-terminal domain. Proc. Natl. Acad. Sci. USA 2013, 110, 1267–1272. [Google Scholar] [CrossRef] [Green Version]
  37. Dutta, P.; Sabri, N.; Li, J.; Li, W.X. Role of STAT3 in lung cancer. Jak-Stat 2014, 3, e999503. [Google Scholar] [CrossRef]
  38. Yu, H.; Kortylewski, M.; Pardoll, D. Crosstalk between cancer and immune cells: Role of STAT3 in the tumour microenvironment. Nat. Rev. Immunol. 2007, 7, 41–51. [Google Scholar] [CrossRef]
  39. Levy, D.E.; Darnell, J. Stats: Transcriptional control and biological impact. Nat. Rev. Mol. Cell Biol. 2002, 3, 651–662. [Google Scholar] [CrossRef]
  40. Avalle, L.; Camporeale, A.; Camperi, A.; Poli, V. STAT3 in cancer: A double edged sword. Cytokine 2017, 98, 42–50. [Google Scholar] [CrossRef]
  41. Alexander, W.S. Suppressors of cytokine signalling (SOCS) in the immune system. Nat. Rev. Immunol. 2002, 2, 410–416. [Google Scholar] [CrossRef]
  42. Shuai, K.; Liu, B. Regulation of gene-activation pathways by PIAS proteins in the immune system. Nat. Rev. Immunol. 2005, 5, 593–605. [Google Scholar] [CrossRef] [PubMed]
  43. Xu, D.; Qu, C.K. Protein tyrosine phosphatases in the JAK/STAT pathway. Front. Biosci. A J. Virtual Libr. 2008, 13, 4925–4932. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Krebs, D.L.; Hilton, D.J. SOCS proteins: Negative regulators of cytokine signaling. Stem Cells 2001, 19, 378–387. [Google Scholar] [CrossRef] [PubMed]
  45. Kadye, R.; Stoffels, M.; Fanucci, S.; Mbanxa, S.; Prinsloo, E. A STAT3 of Addiction: Adipose Tissue, Adipocytokine Signalling and STAT3 as Mediators of Metabolic Remodelling in the Tumour Microenvironment. Cells 2020, 9, 1043. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Brachet-Botineau, M.; Polomski, M.; Neubauer, H.A.; Juen, L.; Hédou, D.; Viaud-Massuard, M.-C.; Prié, G.; Gouilleux, F. Pharmacological Inhibition of Oncogenic STAT3 and STAT5 Signaling in Hematopoietic Cancers. Cancers 2020, 12, 240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Liang, R.; Chen, X.; Chen, L.; Wan, F.; Chen, K.; Sun, Y.; Zhu, X. STAT3 signaling in ovarian cancer: A potential therapeutic target. J. Cancer 2020, 11, 837. [Google Scholar] [CrossRef] [Green Version]
  48. Hu, F.; Li, G.; Huang, C.; Hou, Z.; Yang, X.; Luo, X.; Feng, Y.; Wang, G.; Hu, J.; Cao, Z. The autophagy-independent role of BECN1 in colorectal cancer metastasis through regulating STAT3 signaling pathway activation. Cell Death Dis. 2020, 11, 304. [Google Scholar] [CrossRef] [PubMed]
  49. Zhang, X.; Song, M.; Kundu, J.K.; Lee, M.H.; Liu, Z.Z. PIM Kinase as an Executional Target in Cancer. J. Cancer Prev. 2018, 23, 109–116. [Google Scholar] [CrossRef] [Green Version]
  50. Sun, Z.; Zeng, L.; Zhang, M.; Zhang, Y.; Yang, N. PIM1 inhibitor synergizes the anti-tumor effect of osimertinib via STAT3 dephosphorylation in EGFR-mutant non-small cell lung cancer. Ann. Transl. Med. 2020, 8, 366. [Google Scholar] [CrossRef]
  51. Liu, Q.; Li, A.; Wang, L.; He, W.; Zhao, L.; Wu, C.; Lu, S.; Ye, X.; Zhao, H.; Shen, X.; et al. Stomatin-like Protein 2 Promotes Tumor Cell Survival by Activating the JAK2-STAT3-PIM1 Pathway, Suggesting a Novel Therapy in CRC. Mol. Ther. Oncolytics 2020, 17, 169–179. [Google Scholar] [CrossRef]
  52. Shang, R.; Wang, M.; Dai, B.; Du, J.; Wang, J.; Liu, Z.; Qu, S.; Yang, X.; Liu, J.; Xia, C.; et al. Long noncoding RNA SLC2A1-AS1 regulates aerobic glycolysis and progression in hepatocellular carcinoma via inhibiting the STAT3/FOXM1/GLUT1 pathway. Mol. Oncol. 2020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Fang, Y.; Zhang, S.; Yin, J.; Shen, Y.X.; Wang, H.; Chen, X.S.; Tang, H. LINC01535 promotes proliferation and inhibits apoptosis in esophageal squamous cell cancer by activating the JAK/STAT3 pathway. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 3694–3700. [Google Scholar] [CrossRef] [PubMed]
  54. Lee, M.M.; Chan, B.D.; Wong, W.Y.; Qu, Z.; Chan, M.S.; Leung, T.W.; Lin, Y.; Mok, D.K.; Chen, S.; Tai, W.C. Anti-cancer Activity of Centipeda minima Extract in Triple Negative Breast Cancer via Inhibition of AKT, NF-kappaB, and STAT3 Signaling Pathways. Front. Oncol. 2020, 10, 491. [Google Scholar] [CrossRef] [PubMed]
  55. Khan, M.; Maryam, A.; Saleem, M.Z.; Shakir, H.A.; Qazi, J.I.; Li, Y.; Ma, T. Brevilin A induces ROS-dependent apoptosis and suppresses STAT3 activation by direct binding in human lung cancer cells. J. Cancer 2020, 11, 3725–3735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Cao, L.; Ren, Y.; Guo, X.; Wang, L.; Zhang, Q.; Li, X.; Wu, X.; Meng, Z.; Xu, K. Downregulation of SETD7 promotes migration and invasion of lung cancer cells via JAK2/STAT3 pathway. Int. J. Mol. Med. 2020, 45, 1616–1626. [Google Scholar] [CrossRef] [PubMed]
  57. Lv, J.; Yu, W.; Zhang, Y.; Cao, X.; Han, L.; Hu, H.; Wang, C. LNK promotes the growth and metastasis of triple negative breast cancer via activating JAK/STAT3 and ERK1/2 pathway. Cancer Cell Int. 2020, 20, 124. [Google Scholar] [CrossRef] [Green Version]
  58. Feng, J.; Jiang, W.; Liu, Y.; Huang, W.; Hu, K.; Li, K.; Chen, J.; Ma, C.; Sun, Z.; Pang, X. Blocking STAT3 by pyrvinium pamoate causes metabolic lethality in KRAS-mutant lung cancer. Biochem. Pharmacol. 2020, 177, 113960. [Google Scholar] [CrossRef]
  59. Zhang, Z.; Wang, W.; Ma, D.; Xiong, J.; Kuang, X.; Zhang, S.; Fang, Q.; Wang, J. Heme oxygenase-1 inhibition mediates Gas6 to enhance bortezomib-sensitivity in multiple myeloma via ERK/STAT3 axis. Aging 2020, 12, 6611–6629. [Google Scholar] [CrossRef]
  60. DeVaux, R.S.; Ropri, A.S.; Grimm, S.L.; Hall, P.A.; Herrera, E.O.; Chittur, S.V.; Smith, W.P.; Coarfa, C.; Behbod, F.; Herschkowitz, J.I. Long noncoding RNA BHLHE40-AS1 promotes early breast cancer progression through modulating IL-6/STAT3 signaling. J. Cell. Biochem. 2020, 121, 3465–3478. [Google Scholar] [CrossRef] [Green Version]
  61. Arnold, K.M.; Opdenaker, L.M.; Flynn, N.J.; Appeah, D.K.; Sims-Mourtada, J. Radiation induces an inflammatory response that results in STAT3-dependent changes in cellular plasticity and radioresistance of breast cancer stem-like cells. Int. J. Radiat. Biol. 2020, 96, 434–447. [Google Scholar] [CrossRef]
  62. Song, H.; Luo, Q.; Deng, X.; Ji, C.; Li, D.; Munankarmy, A.; Jian, W.; Zhao, J.; Fang, L. VGLL4 interacts with STAT3 to function as a tumor suppressor in triple-negative breast cancer. Exp. Mol. Med. 2019, 51, 1–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Kim, J.H.; Choi, H.S.; Kim, S.L.; Lee, D.S. The PAK1-Stat3 Signaling Pathway Activates IL-6 Gene Transcription and Human Breast Cancer Stem Cell Formation. Cancers 2019, 11, 1527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Xing, J.; Li, J.; Fu, L.; Gai, J.; Guan, J.; Li, Q. SIRT4 enhances the sensitivity of ER-positive breast cancer to tamoxifen by inhibiting the IL-6/STAT3 signal pathway. Cancer Med. 2019, 8, 7086–7097. [Google Scholar] [CrossRef] [Green Version]
  65. Zheng, T.; Ma, G.; Tang, M.; Li, Z.; Xu, R. IL-8 Secreted from M2 Macrophages Promoted Prostate Tumorigenesis via STAT3/MALAT1 Pathway. Int. J. Mol. Sci. 2018, 20, 98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Dai, H.; Wang, C.; Yu, Z.; He, D.; Yu, K.; Liu, Y.; Wang, S. MiR-17 Regulates Prostate Cancer Cell Proliferation and Apoptosis Through Inhibiting JAK-STAT3 Signaling Pathway. Cancer Biother. Radiopharm. 2018, 33, 103–109. [Google Scholar] [CrossRef]
  67. Soutto, M.; Chen, Z.; Bhat, A.A.; Wang, L.; Zhu, S.; Gomaa, A.; Bates, A.; Bhat, N.S.; Peng, D.; Belkhiri, A.; et al. Activation of STAT3 signaling is mediated by TFF1 silencing in gastric neoplasia. Nat. Commun. 2019, 10, 3039. [Google Scholar] [CrossRef]
  68. Anand, V.; Khandelwal, M.; Appunni, S.; Gupta, N.; Seth, A.; Singh, P.; Mathur, S.; Sharma, A. CD44 splice variant (CD44v3) promotes progression of urothelial carcinoma of bladder through Akt/ERK/STAT3 pathways: Novel therapeutic approach. J. Cancer Res. Clin. Oncol. 2019, 145, 2649–2661. [Google Scholar] [CrossRef]
  69. Yuan, L.; Ye, J.; Fan, D. The B7-H4 gene induces immune escape partly via upregulating the PD-1/Stat3 pathway in non-small cell lung cancer. Hum. Immunol. 2020, 81, 254–261. [Google Scholar] [CrossRef]
  70. Lee, J.H.; Lee, H.H.; Ryu, K.D.; Kim, M.; Ko, D.; Chung, K.S.; Hassan, A.H.E.; Lee, S.H.; Lee, J.Y.; Lee, K.T. KCP10043F Represses the Proliferation of Human Non-Small Cell Lung Cancer Cells by Caspase-Mediated Apoptosis via STAT3 Inactivation. J. Clin. Med. 2020, 9, 704. [Google Scholar] [CrossRef] [Green Version]
  71. Yun, H.H.; Kim, S.; Kuh, H.J.; Lee, J.H. Downregulation of BIS sensitizes A549 cells for digoxin-mediated inhibition of invasion and migration by the STAT3-dependent pathway. Biochem. Biophys. Res. Commun. 2020, 524, 643–648. [Google Scholar] [CrossRef]
  72. Matsumoto, Y.; Ichikawa, T.; Kurozumi, K.; Otani, Y.; Fujimura, A.; Fujii, K.; Tomita, Y.; Hattori, Y.; Uneda, A.; Tsuboi, N.; et al. Annexin A2-STAT3-Oncostatin M receptor axis drives phenotypic and mesenchymal changes in glioblastoma. Acta Neuropathol. Commun. 2020, 8, 42. [Google Scholar] [CrossRef] [PubMed]
  73. Liu, H.W.; Lee, P.M.; Bamodu, O.A.; Su, Y.K.; Fong, I.H.; Yeh, C.T.; Chien, M.H.; Kan, I.H.; Lin, C.M. Enhanced Hsa-miR-181d/p-STAT3 and Hsa-miR-181d/p-STAT5A Ratios Mediate the Anticancer Effect of Garcinol in STAT3/5A-Addicted Glioblastoma. Cancers 2019, 11, 1888. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Hou, J.; Lv, A.; Deng, Q.; Zhang, G.; Hu, X.; Cui, H. TROP2 promotes the proliferation and metastasis of glioblastoma cells by activating the JAK2/STAT3 signaling pathway. Oncol. Rep. 2019, 41, 753–764. [Google Scholar] [CrossRef] [Green Version]
  75. Liu, Y.S.; Hsu, J.W.; Lin, H.Y.; Lai, S.W.; Huang, B.R.; Tsai, C.F.; Lu, D.Y. Bradykinin B1 receptor contributes to interleukin-8 production and glioblastoma migration through interaction of STAT3 and SP-1. Neuropharmacology 2019, 144, 143–154. [Google Scholar] [CrossRef] [PubMed]
  76. Hong, D.; Kurzrock, R.; Kim, Y.; Woessner, R.; Younes, A.; Nemunaitis, J.; Fowler, N.; Zhou, T.; Schmidt, J.; Jo, M.; et al. AZD9150, a next-generation antisense oligonucleotide inhibitor of STAT3 with early evidence of clinical activity in lymphoma and lung cancer. Sci. Transl. Med. 2015, 7, 314ra185. [Google Scholar] [CrossRef] [Green Version]
  77. Sonnenblick, A.; Salgado, R.; Brohée, S.; Zahavi, T.; Peretz, T.; Van den Eynden, G.; Rouas, G.; Salmon, A.; Francis, P.A.; Di Leo, A.; et al. p-STAT3 in luminal breast cancer: Integrated RNA-protein pooled analysis and results from the BIG 2-98 phase III trial. Int. J. Oncol. 2018, 52, 424–432. [Google Scholar] [CrossRef]
  78. Monnien, F.; Zaki, H.; Borg, C.; Mougin, C.; Bosset, J.F.; Mercier, M.; Arbez-Gindre, F.; Kantelip, B. Prognostic value of phosphorylated STAT3 in advanced rectal cancer: A study from 104 French patients included in the EORTC 22921 trial. J. Clin. Pathol. 2010, 63, 873–878. [Google Scholar] [CrossRef]
  79. Bu, X.; Zhao, C.; Wang, W.; Zhang, N. GRIM-19 inhibits the STAT3 signaling pathway and sensitizes gastric cancer cells to radiation. Gene 2013, 512, 198–205. [Google Scholar] [CrossRef]
  80. Anglesio, M.S.; George, J.; Kulbe, H.; Friedlander, M.; Rischin, D.; Lemech, C.; Power, J.; Coward, J.; Cowin, P.A.; House, C.M.; et al. IL6-STAT3-HIF signaling and therapeutic response to the angiogenesis inhibitor sunitinib in ovarian clear cell cancer. Clin. Cancer Res. An. J. Am. Assoc. Cancer Res. 2011, 17, 2538–2548. [Google Scholar] [CrossRef] [Green Version]
  81. Torres-Roca, J.F.; DeSilvio, M.; Mora, L.B.; Khor, L.Y.; Hammond, E.; Ahmad, N.; Jove, R.; Forman, J.; Lee, R.J.; Sandler, H.; et al. Activated STAT3 as a correlate of distant metastasis in prostate cancer: A secondary analysis of Radiation Therapy Oncology Group 86-10. Urology 2007, 69, 505–509. [Google Scholar] [CrossRef]
  82. Delyon, J.; Chevret, S.; Jouary, T.; Dalac, S.; Dalle, S.; Guillot, B.; Arnault, J.P.; Avril, M.F.; Bedane, C.; Bens, G.; et al. STAT3 Mediates Nilotinib Response in KIT-Altered Melanoma: A Phase II Multicenter Trial of the French Skin Cancer Network. J. Investig. Dermatol. 2018, 138, 58–67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Oh, D.Y.; Lee, S.H.; Han, S.W.; Kim, M.J.; Kim, T.M.; Kim, T.Y.; Heo, D.S.; Yuasa, M.; Yanagihara, Y.; Bang, Y.J. Phase I Study of OPB-31121, an Oral STAT3 Inhibitor, in Patients with Advanced Solid Tumors. Cancer Res. Treat. J. Korean Cancer Assoc. 2015, 47, 607–615. [Google Scholar] [CrossRef] [PubMed]
  84. Jamali, Z.; Taheri-Anganeh, M.; Shabaninejad, Z.; Keshavarzi, A.; Taghizadeh, H.; Razavi, Z.S.; Mottaghi, R.; Abolhassan, M.; Movahedpour, A.; Mirzaei, H. Autophagy regulation by microRNAs: Novel insights into osteosarcoma therapy. IUBMB Life 2020. [Google Scholar] [CrossRef] [PubMed]
  85. Pourhanifeh, M.H.; Mahjoubin-Tehran, M.; Shafiee, A.; Hajighadimi, S.; Moradizarmehri, S.; Mirzaei, H.; Asemi, Z. MicroRNAs and exosomes: Small molecules with big actions in multiple myeloma pathogenesis. IUBMB Life 2020, 72, 314–333. [Google Scholar] [CrossRef] [PubMed]
  86. Nahand, J.S.; Karimzadeh, M.R.; Nezamnia, M.; Fatemipour, M.; Khatami, A.; Jamshidi, S.; Moghoofei, M.; Taghizadieh, M.; Hajighadimi, S.; Shafiee, A.; et al. The role of miR-146a in viral infection. IUBMB Life 2020, 72, 343–360. [Google Scholar] [CrossRef]
  87. Javandoost, E.; Firoozi-Majd, E.; Rostamian, H.; Khakpoor-Koosheh, M.; Mirzaei, H.R. Role of microRNAs in Chronic Lymphocytic Leukemia Pathogenesis. Curr. Med. Chem. 2020, 27, 282–297. [Google Scholar] [CrossRef]
  88. Savardashtaki, A.; Shabaninejad, Z.; Movahedpour, A.; Sahebnasagh, R.; Mirzaei, H.; Hamblin, M.R. miRNAs derived from cancer-associated fibroblasts in colorectal cancer. Epigenomics 2019, 11, 1627–1645. [Google Scholar] [CrossRef]
  89. Nahand, J.S.; Taghizadeh-boroujeni, S.; Karimzadeh, M.; Borran, S.; Pourhanifeh, M.H.; Moghoofei, M.; Bokharaei-Salim, F.; Karampoor, S.; Jafari, A.; Asemi, Z.; et al. microRNAs: New prognostic, diagnostic, and therapeutic biomarkers in cervical cancer. J. Cell. Physiol. 2019, 234, 17064–17099. [Google Scholar] [CrossRef]
  90. Zhang, L.; Wang, Z.; Li, B.; Xia, Z.; Wang, X.; Xiu, Y.; Zhang, Z.; Chen, C.; Song, H.; Li, W.; et al. The inhibition of miR-17-5p promotes cortical neuron neurite growth via STAT3/GAP-43 pathway. Mol. Biol. Rep. 2020, 47, 1795–1802. [Google Scholar] [CrossRef]
  91. Zhou, Y.; Xu, X.M.; Feng, Y. MiR-769-5p inhibits cancer progression in oral squamous cell carcinoma by directly targeting JAK1/STAT3 pathway. Neoplasma 2020, 67, 528–536. [Google Scholar] [CrossRef]
  92. Quero, L.; Tiaden, A.N.; Hanser, E.; Roux, J.; Laski, A.; Hall, J.; Kyburz, D. miR-221-3p Drives the Shift of M2-Macrophages to a Pro-Inflammatory Function by Suppressing JAK3/STAT3 Activation. Front. Immunol. 2019, 10, 3087. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Guoping, M.; Ran, L.; Yanru, Q. miR-143 Inhibits Cell Proliferation of Gastric Cancer Cells Through Targeting GATA6. Oncol. Res. 2018, 26, 1023–1029. [Google Scholar] [CrossRef] [PubMed]
  94. Lei, C.; Du, F.; Sun, L.; Li, T.; Li, T.; Min, Y.; Nie, A.; Wang, X.; Geng, L.; Lu, Y.; et al. miR-143 and miR-145 inhibit gastric cancer cell migration and metastasis by suppressing MYO6. Cell Death Dis. 2017, 8, e3101. [Google Scholar] [CrossRef] [Green Version]
  95. Zhang, Q.; Feng, Y.; Liu, P.; Yang, J. MiR-143 inhibits cell proliferation and invasion by targeting DNMT3A in gastric cancer. Tumour Biol. J. Int. Soc. Oncodev. Biol. Med. 2017, 39, 1010428317711312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Wu, Y.; Wan, X.; Zhao, X.; Song, Z.; Xu, Z.; Tao, Y.; Sun, C. MicroRNA-143 suppresses the proliferation and metastasis of human gastric cancer cells via modulation of STAT3 expression. Am. J. Transl. Res. 2020, 12, 867–874. [Google Scholar] [PubMed]
  97. Xiong, J.; Tu, Y.; Feng, Z.; Li, D.; Yang, Z.; Huang, Q.; Li, Z.; Cao, Y.; Jie, Z. Epigenetics mechanisms mediate the miR-125a/BRMS1 axis to regulate invasion and metastasis in gastric cancer. OncoTargets Ther. 2019, 12, 7513–7525. [Google Scholar] [CrossRef] [Green Version]
  98. Li, G.; Ao, S.; Hou, J.; Lyu, G. Low expression of miR-125a-5p is associated with poor prognosis in patients with gastric cancer. Oncol. Lett. 2019, 18, 1483–1490. [Google Scholar] [CrossRef]
  99. Yang, L.; Zhang, S.; Guo, K.; Huang, H.; Qi, S.; Yao, J.; Zhang, Z. miR-125a restrains cell migration and invasion by targeting STAT3 in gastric cancer cells. OncoTargets Ther. 2019, 12, 205–215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Zabaleta, J. MicroRNA: A bridge from H. pylori infection to gastritis and gastric cancer development. Front. Genet. 2012, 3, 294. [Google Scholar] [CrossRef] [Green Version]
  101. Isomoto, H.; Matsushima, K.; Inoue, N.; Hayashi, T.; Nakayama, T.; Kunizaki, M.; Hidaka, S.; Nakayama, M.; Hisatsune, J.; Nakashima, M. Interweaving microRNAs and proinflammatory cytokines in gastric mucosa with reference to H. pylori infection. J. Clin. Immunol. 2012, 32, 290–299. [Google Scholar] [CrossRef]
  102. Belair, C.; Darfeuille, F.; Staedel, C. Helicobacter pylori and gastric cancer: Possible role of microRNAs in this intimate relationship. Clin. Microbiol. Infect. 2009, 15, 806–812. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Link, A.; Kupcinskas, J.; Wex, T.; Malfertheiner, P. Macro-role of microRNA in gastric cancer. Dig. Dis. 2012, 30, 255–267. [Google Scholar] [CrossRef] [PubMed]
  104. 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] [PubMed]
  105. Miao, L.; Liu, K.; Xie, M.; Xing, Y.; Xi, T. miR-375 inhibits Helicobacter pylori-induced gastric carcinogenesis by blocking JAK2-STAT3 signaling. Cancer Immunol. Immunother. 2014, 63, 699–711. [Google Scholar] [CrossRef]
  106. Shi, H.; Chen, X.; Jiang, H.; Wang, X.; Yu, H.; Sun, P.; Sui, X. miR-148a suppresses cell invasion and migration in gastric cancer by targeting DNA methyltransferase 1. Oncol. Lett. 2018, 15, 4944–4950. [Google Scholar] [CrossRef] [Green Version]
  107. Li, B.; Wang, W.; Li, Z.; Chen, Z.; Zhi, X.; Xu, J.; Li, Q.; Wang, L.; Huang, X.; Wang, L.; et al. MicroRNA-148a-3p enhances cisplatin cytotoxicity in gastric cancer through mitochondrial fission induction and cyto-protective autophagy suppression. Cancer Lett. 2017, 410, 212–227. [Google Scholar] [CrossRef]
  108. Yu, B.; Lv, X.; Su, L.; Li, J.; Yu, Y.; Gu, Q.; Yan, M.; Zhu, Z.; Liu, B. MiR-148a Functions as a Tumor Suppressor by Targeting CCK-BR via Inactivating STAT3 and Akt in Human Gastric Cancer. PLoS ONE 2016, 11, e0158961. [Google Scholar] [CrossRef]
  109. Hashemi Goradel, N.; Ghiyami-Hour, F.; Jahangiri, S.; Negahdari, B.; Sahebkar, A.; Masoudifar, A.; Mirzaei, H. Nanoparticles as new tools for inhibition of cancer angiogenesis. J. Cell. Physiol. 2018, 233, 2902–2910. [Google Scholar] [CrossRef]
  110. Mashreghi, M.; Azarpara, H.; Bazaz, M.R.; Jafari, A.; Masoudifar, A.; Mirzaei, H.; Jaafari, M.R. Angiogenesis biomarkers and their targeting ligands as potential targets for tumor angiogenesis. J. Cell. Physiol. 2018, 233, 2949–2965. [Google Scholar] [CrossRef]
  111. Wang, M.; Zeng, Q.; Li, Y.; Imani, S.; Xie, D.; Li, Y.; Han, Y.; Fan, J. Bevacizumab combined with apatinib enhances antitumor and anti-angiogenesis effects in a lung cancer model in vitro and in vivo. J. Drug Target. 2020, 1–29. [Google Scholar] [CrossRef]
  112. Yadav, A.S.; Radharani, N.N.V.; Gorain, M.; Bulbule, A.; Shetti, D.; Roy, G.; Baby, T.; Kundu, G.C. RGD functionalized chitosan nanoparticle mediated targeted delivery of raloxifene selectively suppresses angiogenesis and tumor growth in breast cancer. Nanoscale 2020, 12, 10664–10684. [Google Scholar] [CrossRef] [PubMed]
  113. Zheng, X.; Ma, Y.F.; Zhang, X.R.; Li, Y.; Zhao, H.H.; Han, S.G. Circ_0056618 promoted cell proliferation, migration and angiogenesis through sponging with miR-206 and upregulating CXCR4 and VEGF-A in colorectal cancer. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 4190–4202. [Google Scholar] [CrossRef] [PubMed]
  114. Cao, X.; Li, B.; Han, X.; Zhang, X.; Dang, M.; Wang, H.; Du, F.; Zeng, X.; Guo, C. Soluble receptor for advanced glycation end-products promotes angiogenesis through activation of STAT3 in myocardial ischemia/reperfusion injury. Apoptosis Int. J. Program. Cell Death 2020, 25, 341–353. [Google Scholar] [CrossRef] [PubMed]
  115. Yuan, Q.; Yao, F.; Zhou, L.; Liang, G.; Song, X.; Jiang, G.; Zhou, M.; Zhang, L. Yu Ping Feng San Exert Anti-Angiogenesis Effects through the Inhibition of TSLP-STAT3 Signaling Pathways in Hepatocellular Carcinoma. Evid. Based Complement. Altern. Med. ECAM 2019, 2019, 1947156. [Google Scholar] [CrossRef] [Green Version]
  116. Zhu, F.; Wang, K.B.; Rui, L. STAT3 Activation and Oncogenesis in Lymphoma. Cancers 2019, 12, 19. [Google Scholar] [CrossRef] [Green Version]
  117. Zhang, X.; Tang, J.; Zhi, X.; Xie, K.; Wang, W.; Li, Z.; Zhu, Y.; Yang, L.; Xu, H.; Xu, Z. miR-874 functions as a tumor suppressor by inhibiting angiogenesis through STAT3/VEGF-A pathway in gastric cancer. Oncotarget 2015, 6, 1605–1617. [Google Scholar] [CrossRef] [PubMed]
  118. Cai, K.; Li, H.X.; Li, P.P.; Guo, Z.J.; Yang, Y. MicroRNA-449b-3p inhibits epithelial-mesenchymal transition by targeting IL-6 and through the JAK2/STAT3 signaling pathway in non-small cell lung cancer. Exp. Ther. Med. 2020, 19, 2527–2534. [Google Scholar] [CrossRef] [Green Version]
  119. Sun, X.; Ma, J.; Chen, Q.; Hou, Z.; Luo, X.; Wang, G.; Wang, J.; Hu, J.; Cao, Z. SIX4 promotes metastasis through STAT3 activation in breast cancer. Am. J. Cancer Res. 2020, 10, 224–236. [Google Scholar]
  120. Sun, H.; Zhang, Z.; Zhang, T.; Geng, H.; Xie, D.; Wang, Y.; Ding, D.; Zhang, T.; Yu, D. Resveratrol Reverses Cigarette Smoke-Induced Urocystic Epithelial-Mesenchymal Transition via Suppression of STAT3 Phosphorylation in SV-HUC-1-Immortalized Human Urothelial Cells. Oncotargets Ther. 2019, 12, 10227–10237. [Google Scholar] [CrossRef] [Green Version]
  121. Tao, Y.; Yang, S.; Wu, Y.; Fang, X.; Wang, Y.; Song, Y.; Han, T. MicroRNA-216a inhibits the metastasis of gastric cancer cells by targeting JAK2/STAT3-mediated EMT process. Oncotarget 2017, 8, 88870–88881. [Google Scholar] [CrossRef] [Green Version]
  122. Leng, X.; Liu, G.; Wang, S.; Song, J.; Zhang, W.; Zhang, X.; Rong, L.; Ma, Y.; Song, F. LINC01272 Promotes Migration and Invasion of Gastric Cancer Cells via EMT. Oncotargets Ther. 2020, 13, 3401–3410. [Google Scholar] [CrossRef] [Green Version]
  123. Jin, G.H.; Shi, Y.; Tian, Y.; Cao, T.T.; Mao, Y.; Tang, T.Y. HMGA1 accelerates the malignant progression of gastric cancer through stimulating EMT. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 3642–3647. [Google Scholar] [CrossRef]
  124. Zhao, L.; Niu, H.; Liu, Y.; Wang, L.; Zhang, N.; Zhang, G.; Liu, R.; Han, M. LOX inhibition downregulates MMP-2 and MMP-9 in gastric cancer tissues and cells. J. Cancer 2019, 10, 6481–6490. [Google Scholar] [CrossRef] [PubMed]
  125. Fang, X.; Dong, Y.; Yang, R.; Wei, L. LINC00619 restricts gastric cancer progression by preventing microRNA-224-5p-mediated inhibition of OPCML. Arch. Biochem. Biophys. 2020, 108390. [Google Scholar] [CrossRef] [PubMed]
  126. Xu, J.; Wu, J.; Tang, C. Effect of isofebrifugine on the proliferation and invasion of human gastric cancer cells via MMP. Cell. Mol. Biol. (Noisy-Le-Grandfrance) 2020, 66, 27–31. [Google Scholar] [CrossRef]
  127. Ma, D.H.; Li, B.S.; Liu, J.J.; Xiao, Y.F.; Yong, X.; Wang, S.M.; Wu, Y.Y.; Zhu, H.B.; Wang, D.X.; Yang, S.M. miR-93-5p/IFNAR1 axis promotes gastric cancer metastasis through activating the STAT3 signaling pathway. Cancer Lett. 2017, 408, 23–32. [Google Scholar] [CrossRef] [PubMed]
  128. Yoon, C.J.; Chang, M.S.; Kim, D.H.; Kim, W.; Koo, B.K.; Yun, S.C.; Kim, S.H.; Kim, Y.S.; Woo, J.H. Epstein-Barr virus-encoded miR-BART5-5p upregulates PD-L1 through PIAS3/pSTAT3 modulation, worsening clinical outcomes of PD-L1-positive gastric carcinomas. Gastric Cancer J. Int. Gastric Cancer Assoc. Jpn. Gastric Cancer Assoc. 2020, 1–16. [Google Scholar] [CrossRef] [PubMed]
  129. Qu, D.; Yang, Y.; Huang, X. miR-199a-5p promotes proliferation and metastasis and epithelial-mesenchymal transition through targeting PIAS3 in cervical carcinoma. J. Cell. Biochem. 2019, 120, 13562–13572. [Google Scholar] [CrossRef] [PubMed]
  130. Dai, Y.X.; Qiu, M.K.; Wang, S.Q.; Pan, C.; Wang, Y.; Ou, J.M. lncRNA CASC2 suppresses the growth of hemangioma cells by regulating miR-18a-5p/FBXL3 axis. J. Biol. Regul. Homeost. Agents 2020, 34, 49–56. [Google Scholar] [CrossRef]
  131. Wu, W.; Takanashi, M.; Borjigin, N.; Ohno, S.I.; Fujita, K.; Hoshino, S.; Osaka, Y.; Tsuchida, A.; Kuroda, M. MicroRNA-18a modulates STAT3 activity through negative regulation of PIAS3 during gastric adenocarcinogenesis. Br. J. Cancer 2013, 108, 653–661. [Google Scholar] [CrossRef] [PubMed]
  132. Wei, H.; Li, Y.; Ning, Q.; Suo, Z.M. Regulation of miR-155 affects the invasion and migration of gastric carcinoma cells by modulating the STAT3 signaling pathway. Oncol. Lett. 2018, 16, 4137–4142. [Google Scholar] [CrossRef] [Green Version]
  133. Guo, W.; Li, W.; Yuan, L.; Mei, X.; Hu, W. MicroRNA-106a-3p Induces Apatinib Resistance and Activates Janus-Activated Kinase 2 (JAK2)/Signal Transducer and Activator of Transcription 3 (STAT3) by Targeting the SOCS System in Gastric Cancer. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2019, 25, 10122–10128. [Google Scholar] [CrossRef] [PubMed]
  134. Koh, J.S.; Joo, M.K.; Park, J.J.; Yoo, H.S.; Choi, B.I.; Lee, B.J.; Chun, H.J.; Lee, S.W. Inhibition of STAT3 in gastric cancer: Role of pantoprazole as SHP-1 inducer. Cell Biosci. 2018, 8, 50. [Google Scholar] [CrossRef] [PubMed]
  135. Zhang, Y.; Wang, Q. Sunitinib reverse multidrug resistance in gastric cancer cells by modulating Stat3 and inhibiting P-gp function. Cell Biochem. Biophys. 2013, 67, 575–581. [Google Scholar] [CrossRef] [PubMed]
  136. Tian, M.; Tian, D.; Qiao, X.; Li, J.; Zhang, L. Modulation of Myb-induced NF-kB -STAT3 signaling and resulting cisplatin resistance in ovarian cancer by dietary factors. J. Cell. Physiol. 2019, 234, 21126–21134. [Google Scholar] [CrossRef] [PubMed]
  137. Hussain, H.; Green, I.R.; Saleem, M.; Khattak, K.F.; Irshad, M.; Ali, M. Cucurbitacins as Anticancer Agents: A Patent Review. Recent Pat. Anti-Cancer Drug Discov. 2019, 14, 133–143. [Google Scholar] [CrossRef]
  138. Jafargholizadeh, N.; Zargar, S.J.; Aftabi, Y. The cucurbitacins D, E, and I from Ecballium elaterium (L.) upregulate the LC3 gene and induce cell-cycle arrest in human gastric cancer cell line AGS. Iran. J. Basic Med. Sci. 2018, 21, 253–259. [Google Scholar] [CrossRef]
  139. Wang, J.-R.; Shen, G.-N.; Luo, Y.-H.; Piao, X.-J.; Zhang, Y.; Wang, H.; Li, J.-Q.; Xu, W.-T.; Zhang, Y.; Wang, S.-N. 2-(4-methoxyphenylthio)-5, 8-dimethoxy-1, 4-naphthoquinone induces apoptosis via ROS-mediated MAPK and STAT3 signaling pathway in human gastric cancer cells. J. Chemother. 2019, 31, 214–226. [Google Scholar] [CrossRef]
  140. Zang, Y.Q.; Feng, Y.Y.; Luo, Y.H.; Zhai, Y.Q.; Ju, X.Y.; Feng, Y.C.; Wang, J.R.; Yu, C.Q.; Jin, C.H. Glycitein induces reactive oxygen species-dependent apoptosis and G0/G1 cell cycle arrest through the MAPK/STAT3/NF-κB pathway in human gastric cancer cells. Drug Dev. Res. 2019, 80, 573–584. [Google Scholar] [CrossRef]
  141. Chuang, K.C.; Chang, C.R.; Chang, S.H.; Huang, S.W.; Chuang, S.M.; Li, Z.Y.; Wang, S.T.; Kao, J.K.; Chen, Y.J.; Shieh, J.J. Imiquimod-induced ROS production disrupts the balance of mitochondrial dynamics and increases mitophagy in skin cancer cells. J. Dermatol. Sci. 2020. [Google Scholar] [CrossRef]
  142. Li, D.; Zhao, D.; Du, J.; Dong, S.; Aldhamin, Z.; Yuan, X.; Li, W.; Du, H.; Zhao, W.; Cui, L.; et al. Heme oxygenase-1 alleviated non-alcoholic fatty liver disease via suppressing ROS-dependent endoplasmic reticulum stress. Life Sci. 2020, 253, 117678. [Google Scholar] [CrossRef]
  143. Saleem, M.Z.; Nisar, M.A.; Alshwmi, M.; Din, S.R.U.; Gamallat, Y.; Khan, M.; Ma, T. Brevilin A Inhibits STAT3 Signaling and Induces ROS-Dependent Apoptosis, Mitochondrial Stress and Endoplasmic Reticulum Stress in MCF-7 Breast Cancer Cells. OncoTargets Ther. 2020, 13, 435–450. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Sun, N.; Liang, Y.; Chen, Y.; Wang, L.; Li, D.; Liang, Z.; Sun, L.; Wang, Y.; Niu, H. Glutamine affects T24 bladder cancer cell proliferation by activating STAT3 through ROS and glutaminolysis. Int. J. Mol. Med. 2019, 44, 2189–2200. [Google Scholar] [CrossRef] [PubMed]
  145. Deng, W.; Wang, Y.; Zhao, S.; Zhang, Y.; Chen, Y.; Zhao, X.; Liu, L.; Sun, S.; Zhang, L.; Ye, B.; et al. MICAL1 facilitates breast cancer cell proliferation via ROS-sensitive ERK/cyclin D pathway. J. Cell. Mol. Med. 2018, 22, 3108–3118. [Google Scholar] [CrossRef] [PubMed]
  146. Wang, S.; Wu, Y.Y.; Wang, X.; Shen, P.; Jia, Q.; Yu, S.; Wang, Y.; Li, X.; Chen, W.; Wang, A.; et al. Lycopene prevents carcinogen-induced cutaneous tumor by enhancing activation of the Nrf2 pathway through p62-triggered autophagic Keap1 degradation. Aging 2020, 12, 8167. [Google Scholar] [CrossRef]
  147. Park, B.; Lim, J.W.; Kim, H. Lycopene treatment inhibits activation of Jak1/Stat3 and Wnt/β-catenin signaling and attenuates hyperproliferation in gastric epithelial cells. Nutr. Res. 2019, 70, 70–81. [Google Scholar] [CrossRef]
  148. Lin, L.; Huang, K.; Guo, W.; Zhou, C.; Wang, G.; Zhao, Q. Conditioned Medium of the Osteosarcoma Cell Line U2OS Induces hBMSCs to Exhibit Characteristics of Carcinoma-Associated Fibroblasts via Activation of IL-6/STAT3 Signaling. J. Biochem. 2020. [Google Scholar] [CrossRef]
  149. Lim, H.S.; Kim, O.S.; Kim, B.Y.; Jeong, S.J. Apigetrin from Scutellaria baicalensis Georgi Inhibits Neuroinflammation in BV-2 Microglia and Exerts Neuroprotective Effect in HT22 Hippocampal Cells. J. Med. Food 2016, 19, 1032–1040. [Google Scholar] [CrossRef]
  150. Sun, Q.; Lu, N.-N.; Feng, L. Apigetrin inhibits gastric cancer progression through inducing apoptosis and regulating ROS-modulated STAT3/JAK2 pathway. Biochem. Biophys. Res. Commun. 2018, 498, 164–170. [Google Scholar] [CrossRef]
  151. Puar, Y.R.; Shanmugam, M.K.; Fan, L.; Arfuso, F.; Sethi, G.; Tergaonkar, V. Evidence for the Involvement of the Master Transcription Factor NF-κB in Cancer Initiation and Progression. Biomedicines 2018, 6, 82. [Google Scholar] [CrossRef] [Green Version]
  152. Fan, Y.; Mao, R.; Yang, J. NF-κB and STAT3 signaling pathways collaboratively link inflammation to cancer. Protein Cell 2013, 4, 176–185. [Google Scholar] [CrossRef] [Green Version]
  153. Hoesel, B.; Schmid, J.A. The complexity of NF-κB signaling in inflammation and cancer. Mol. Cancer 2013, 12, 86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Esparza-Lopez, J.; Alvarado-Munoz, J.F.; Escobar-Arriaga, E.; Ulloa-Aguirre, A.; de Jesus Ibarra-Sanchez, M. Metformin reverses mesenchymal phenotype of primary breast cancer cells through STAT3/NF-kappaB pathways. BMC Cancer 2019, 19, 728. [Google Scholar] [CrossRef] [PubMed]
  155. Malinska, H.; Huttl, M.; Oliyarnyk, O.; Markova, I.; Poruba, M.; Racova, Z.; Kazdova, L.; Vecera, R. Beneficial effects of troxerutin on metabolic disorders in non-obese model of metabolic syndrome. PLoS ONE 2019, 14, e0220377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Hoseindoost, M.; Alipour, M.R.; Farajdokht, F.; Diba, R.; Bayandor, P.; Mehri, K.; Nayebi Rad, S.; Babri, S. Effects of troxerutin on inflammatory cytokines and BDNF levels in male offspring of high-fat diet fed rats. Avicenna J. Phytomed. 2019, 9, 597–605. [Google Scholar] [CrossRef] [PubMed]
  157. Thomas, N.S.; George, K.; Selvam, A.A.A. Anticancer mechanism of troxerutin via targeting Nrf2 and NF-kappaB signalling pathways in hepatocarcinoma cell line. Toxicol. in vitro Int. J. Publ. Assoc. BIBRA 2019, 54, 317–329. [Google Scholar] [CrossRef]
  158. Thomas, N.S.; George, K.; Selvam, A.A.A. Troxerutin subdues hepatic tumorigenesis via disrupting the MDM2-p53 interaction. Food Funct. 2018, 9, 5336–5349. [Google Scholar] [CrossRef]
  159. Xu, G.-Y.; Tang, X.-J. Troxerutin (TXN) potentiated 5-Fluorouracil (5-Fu) treatment of human gastric cancer through suppressing STAT3/NF-κB and Bcl-2 signaling pathways. Biomed. Pharmacother. 2017, 92, 95–107. [Google Scholar] [CrossRef]
  160. Samidurai, A.; Roh, S.K.; Prakash, M.; Durrant, D.; Salloum, F.N.; Kukreja, R.C.; Das, A. STAT3-miR-17/20 Signaling Axis Plays a Critical Role in Attenuating Myocardial Infarction following Rapamycin Treatment in Diabetic mice. Cardiovasc. Res. 2019. [Google Scholar] [CrossRef]
  161. Tang, Z.; Xu, T.; Li, Y.; Fei, W.; Yang, G.; Hong, Y. Inhibition of CRY2 by STAT3/miRNA-7-5p Promotes Osteoblast Differentiation through Upregulation of CLOCK/BMAL1/P300 Expression. Mol. Therapy. Nucleic Acids 2020, 19, 865–876. [Google Scholar] [CrossRef]
  162. Wang, Y.; Xu, Z.; Wang, X. miRNA-373 promotes urinary bladder cancer cell proliferation, migration and invasion through upregulating epidermal growth factor receptor. Exp. Ther. Med. 2019, 17, 1190–1195. [Google Scholar] [CrossRef] [Green Version]
  163. Zhang, X.J.; Jin, Y.; Song, J.L.; Deng, F. MiR-373 promotes proliferation and metastasis of oral squamous cell carcinoma by targeting SPOP. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 5270–5276. [Google Scholar] [CrossRef] [PubMed]
  164. Yuan, X.L.; Wen, F.Q.; Chen, X.W.; Jiang, X.P.; Liu, S.X. miR-373 promotes neuroblastoma cell proliferation, migration, and invasion by targeting SRCIN1. OncoTargets Ther. 2019, 12, 4927–4936. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Wei, B.; Sun, X.; Geng, Z.; Shi, M.; Chen, Z.; Chen, L.; Wang, Y.; Fu, X. Isoproterenol regulates CD44 expression in gastric cancer cells through STAT3/MicroRNA373 cascade. Biomaterials 2016, 105, 89–101. [Google Scholar] [CrossRef]
  166. Ji, H.G.; Piao, J.Y.; Kim, S.J.; Kim, D.H.; Lee, H.N.; Na, H.K.; Surh, Y.J. Docosahexaenoic acid inhibits Helicobacter pylori-induced STAT3 phosphorylation through activation of PPARγ. Mol. Nutr. Food Res. 2016, 60, 1448–1457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Song, B.; Zhan, H.; Bian, Q.; Gu, J. Piperlongumine inhibits gastric cancer cells via suppression of the JAK1, 2/STAT3 signaling pathway. Mol. Med. Rep. 2016, 13, 4475–4480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Pandey, A.; Vishnoi, K.; Mahata, S.; Tripathi, S.C.; Misra, S.P.; Misra, V.; Mehrotra, R.; Dwivedi, M.; Bharti, A.C. Berberine and curcumin target survivin and STAT3 in gastric cancer cells and synergize actions of standard chemotherapeutic 5-fluorouracil. Nutr. Cancer 2015, 67, 1295–1306. [Google Scholar] [CrossRef] [PubMed]
  169. Xiao, W.; Wu, K.; Yin, M.; Han, S.; Ding, Y.; Qiao, A.; Lu, G.; Deng, B.; Bo, P.; Gong, W. Wogonin inhibits tumor-derived regulatory molecules by suppressing STAT3 signaling to promote tumor immunity. J. Immunother. 2015, 38, 167–184. [Google Scholar] [CrossRef]
  170. Dai, C.; Liu, P.; Wang, X.; Yin, Y.; Jin, W.; Shen, L.; Chen, Y.; Chen, Z.; Wang, Y. The Antipsychotic Agent Sertindole Exhibited Antiproliferative Activities by Inhibiting the STAT3 Signaling Pathway in Human Gastric Cancer Cells. J. Cancer 2020, 11, 849–857. [Google Scholar] [CrossRef] [Green Version]
  171. Yang, Y.; Fang, E.; Luo, J.; Wu, H.; Jiang, Y.; Liu, Y.; Tong, S.; Wang, Z.; Zhou, R.; Tong, Q. The Antioxidant Alpha-Lipoic Acid Inhibits Proliferation and Invasion of Human Gastric Cancer Cells via Suppression of STAT3-Mediated MUC4 Gene Expression. Oxidative Med. Cell. Longev. 2019, 2019, 3643715. [Google Scholar] [CrossRef]
  172. Jo, M.J.; Jeong, S.; Yun, H.K.; Kim, D.Y.; Kim, B.R.; Kim, J.L.; Na, Y.J.; Park, S.H.; Jeong, Y.A.; Kim, B.G.; et al. Genipin induces mitochondrial dysfunction and apoptosis via downregulation of Stat3/mcl-1 pathway in gastric cancer. BMC Cancer 2019, 19, 739. [Google Scholar] [CrossRef] [Green Version]
  173. Wang, H.; Luo, Y.H.; Shen, G.N.; Piao, X.J.; Xu, W.T.; Zhang, Y.; Wang, J.R.; Feng, Y.C.; Li, J.Q.; Zhang, Y.; et al. Two novel 1,4naphthoquinone derivatives induce human gastric cancer cell apoptosis and cell cycle arrest by regulating reactive oxygen speciesmediated MAPK/Akt/STAT3 signaling pathways. Mol. Med. Rep. 2019, 20, 2571–2582. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Wang, Z.; Tang, X.; Wu, X.; Yang, M.; Wang, W.; Wang, L.; Tang, D.; Wang, D. Cardamonin exerts anti-gastric cancer activity via inhibiting LncRNA-PVT1-STAT3 axis. Biosci. Rep. 2019, 39, BSR20190357. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Xia, Y.; Khoi, P.N.; Yoon, H.J.; Lian, S.; Joo, Y.E.; Chay, K.O.; Kim, K.K.; Do Jung, Y. Piperine inhibits IL-1β-induced IL-6 expression by suppressing p38 MAPK and STAT3 activation in gastric cancer cells. Mol. Cell. Biochem. 2015, 398, 147–156. [Google Scholar] [CrossRef] [PubMed]
  176. Zhang, Y.; Guo, S.; Fang, J.; Peng, B.; Zhang, Y.; Cao, T. Tanshinone IIA inhibits cell proliferation and tumor growth by downregulating STAT3 in human gastric cancer. Exp. Ther. Med. 2018, 16, 2931–2937. [Google Scholar] [CrossRef] [Green Version]
  177. Huang, Y.; Zhang, J.; Wang, G.; Chen, X.; Zhang, R.; Liu, H.; Zhu, J. Oxymatrine exhibits anti-tumor activity in gastric cancer through inhibition of IL-21R-mediated JAK2/STAT3 pathway. Int. J. Immunopathol. Pharmacol. 2018, 32. [Google Scholar] [CrossRef]
  178. Song, S.; Su, Z.; Xu, H.; Niu, M.; Chen, X.; Min, H.; Zhang, B.; Sun, G.; Xie, S.; Wang, H.; et al. Luteolin selectively kills STAT3 highly activated gastric cancer cells through enhancing the binding of STAT3 to SHP-1. Cell Death Dis. 2017, 8, e2612. [Google Scholar] [CrossRef]
  179. Li, H.; Lu, H.; Lv, M.; Wang, Q.; Sun, Y. Parthenolide facilitates apoptosis and reverses drug-resistance of human gastric carcinoma cells by inhibiting the STAT3 signaling pathway. Oncol. Lett. 2018, 15, 3572–3579. [Google Scholar] [CrossRef] [Green Version]
  180. Rajamanickam, V.; Zhu, H.; Feng, C.; Chen, X.; Zheng, H.; Xu, X.; Zhang, Q.; Zou, P.; He, G.; Dai, X.; et al. Novel allylated monocarbonyl analogs of curcumin induce mitotic arrest and apoptosis by reactive oxygen species-mediated endoplasmic reticulum stress and inhibition of STAT3. Oncotarget 2017, 8, 101112–101129. [Google Scholar] [CrossRef] [Green Version]
  181. Zheng, H.; Hong, H.; Zhang, L.; Cai, X.; Hu, M.; Cai, Y.; Zhou, B.; Lin, J.; Zhao, C.; Hu, W. Nifuratel, a novel STAT3 inhibitor with potent activity against human gastric cancer cells. Cancer Manag. Res. 2017, 9, 565–572. [Google Scholar] [CrossRef] [Green Version]
  182. Wang, J.; Zhang, G.; Dai, C.; Gao, X.; Wu, J.; Shen, L.; Chen, Z.; Liu, P. Cryptotanshinone potentiates the antitumor effects of doxorubicin on gastric cancer cells via inhibition of STAT3 activity. J. Int. Med. Res. 2017, 45, 220–230. [Google Scholar] [CrossRef]
  183. Wang, G.; Jing, Y.; Cao, L.; Gong, C.; Gong, Z.; Cao, X. A novel synthetic Asiatic acid derivative induces apoptosis and inhibits proliferation and mobility of gastric cancer cells by suppressing STAT3 signaling pathway. OncoTargets Ther. 2017, 10, 55–66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Wang, X.; Li, Y.; Dai, Y.; Liu, Q.; Ning, S.; Liu, J.; Shen, Z.; Zhu, D.; Jiang, F.; Zhang, J.; et al. Sulforaphane improves chemotherapy efficacy by targeting cancer stem cell-like properties via the miR-124/IL-6R/STAT3 axis. Sci. Rep. 2016, 6, 36796. [Google Scholar] [CrossRef] [PubMed]
  185. Zhu, W.Q.; Wang, J.; Guo, X.F.; Liu, Z.; Dong, W.G. Thymoquinone inhibits proliferation in gastric cancer via the STAT3 pathway in vivo and in vitro. World J. Gastroenterol. 2016, 22, 4149–4159. [Google Scholar] [CrossRef]
  186. Zheng, Y.B.; Xiao, G.C.; Tong, S.L.; Ding, Y.; Wang, Q.S.; Li, S.B.; Hao, Z.N. Paeoniflorin inhibits human gastric carcinoma cell proliferation through up-regulation of microRNA-124 and suppression of PI3K/Akt and STAT3 signaling. World J. Gastroenterol. 2015, 21, 7197–7207. [Google Scholar] [CrossRef] [PubMed]
  187. Cheong, J.H.; Hong, S.Y.; Zheng, Y.; Noh, S.H. Eupatilin Inhibits Gastric Cancer Cell Growth by Blocking STAT3-Mediated VEGF Expression. J. Gastric Cancer 2011, 11, 16–22. [Google Scholar] [CrossRef] [Green Version]
  188. Zhu, B.H.; Chen, H.Y.; Zhan, W.H.; Wang, C.Y.; Cai, S.R.; Wang, Z.; Zhang, C.H.; He, Y.L. (-)-Epigallocatechin-3-gallate inhibits VEGF expression induced by IL-6 via Stat3 in gastric cancer. World J. Gastroenterol. 2011, 17, 2315–2325. [Google Scholar] [CrossRef]
  189. Xie, Y.L.; Tao, W.H.; Yang, T.X.; Qiao, J.G. Anticancer effect of cucurbitacin B on MKN-45 cells via inhibition of the JAK2/STAT3 signaling pathway. Exp. Ther. Med. 2016, 12, 2709–2715. [Google Scholar] [CrossRef] [Green Version]
  190. Liu, Y.F.; Lu, Y.M.; Qu, G.Q.; Liu, Y.; Chen, W.X.; Liao, X.H.; Kong, W.M. Ponicidin induces apoptosis via JAK2 and STAT3 signaling pathways in gastric carcinoma. Int. J. Mol. Sci. 2015, 16, 1576–1589. [Google Scholar] [CrossRef] [Green Version]
  191. Hwang, S.T.; Kim, C.; Lee, J.H.; Chinnathambi, A.; Alharbi, S.A.; Shair, O.H.; Sethi, G.; Ahn, K.S. Cycloastragenol can negate constitutive STAT3 activation and promote paclitaxel-induced apoptosis in human gastric cancer cells. Phytomedicine 2019, 59, 152907. [Google Scholar] [CrossRef]
  192. Yu, R.X.; Yu, R.T.; Liu, Z. Inhibition of two gastric cancer cell lines induced by fucoxanthin involves downregulation of Mcl-1 and STAT3. Hum. Cell 2018, 31, 50–63. [Google Scholar] [CrossRef]
  193. Jiang, X.; Wu, M.; Xu, Z.; Wang, H.; Wang, H.; Yu, X.; Li, Z.; Teng, L. HJC0152, a novel STAT3 inhibitor with promising anti-tumor effect in gastric cancer. Cancer Manag. Res. 2018, 10, 6857–6867. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Su, C.; Wang, W.; Wang, C. IGF-1-induced MMP-11 expression promotes the proliferation and invasion of gastric cancer cells through the JAK1/STAT3 signaling pathway. Oncol. Lett. 2018, 15, 7000–7006. [Google Scholar] [CrossRef] [PubMed]
  195. Jiang, X.; Tang, J.; Wu, M.; Chen, S.; Xu, Z.; Wang, H.; Wang, H.; Yu, X.; Li, Z.; Teng, L. BP1102 exerts an antitumor effect on the AGS human gastric cancer cell line through modulating the STAT3 and MAPK signaling pathways. Mol. Med. Rep. 2019, 19, 2698–2706. [Google Scholar] [CrossRef] [PubMed]
  196. Vafadar, A.; Shabaninejad, Z.; Movahedpour, A.; Mohammadi, S.; Fathullahzadeh, S.; Mirzaei, H.R.; Namdar, A.; Savardashtaki, A.; Mirzaei, H. Long Non-Coding RNAs As Epigenetic Regulators in Cancer. Curr. Pharm. Des. 2019, 25, 3563–3577. [Google Scholar] [CrossRef]
  197. Liang, Y.; Song, X.; Li, Y.; Chen, B.; Zhao, W.; Wang, L.; Zhang, H.; Liu, Y.; Han, D.; Zhang, N.; et al. LncRNA BCRT1 promotes breast cancer progression by targeting miR-1303/PTBP3 axis. Mol. Cancer 2020, 19, 85. [Google Scholar] [CrossRef]
  198. Zhao, Y.; Dong, X.; Hou, R. lncRNA PICART1 alleviates progression of cervical cancer by upregulating TCF21. Oncol. Lett. 2020, 19, 3719–3724. [Google Scholar] [CrossRef] [Green Version]
  199. Shang, A.; Wang, W.; Gu, C.; Chen, W.; Lu, W.; Sun, Z.; Li, D. Long non-coding RNA CCAT1 promotes colorectal cancer progression by regulating miR-181a-5p expression. Aging 2020, 12, 8301. [Google Scholar] [CrossRef]
  200. Luo, L.; Zhang, J.; Tang, H.; Zhai, D.; Huang, D.; Ling, L.; Wang, X.; Liu, T.; Zhang, Q.; Zhang, Z.; et al. LncRNA SNORD3A specifically sensitizes breast cancer cells to 5-FU by sponging miR-185-5p to enhance UMPS expression. Cell Death Dis. 2020, 11, 329. [Google Scholar] [CrossRef]
  201. Wu, Y.; Bi, Q.J.; Han, R.; Zhang, Y. Long noncoding RNA KCNQ1OT1 is correlated with human breast cancer cell development through inverse regulation of hsa-miR-107. Biochem. Cell Biol. Biochim. Et Biol. Cell 2020, 999, 1–7. [Google Scholar] [CrossRef]
  202. Guo, J.; Ding, Y.; Yang, H.; Guo, H.; Zhou, X.; Chen, X. Aberrant expression of lncRNA MALAT1 modulates radioresistance in colorectal cancer in vitro via miR-101-3p sponging. Exp. Mol. Pathol. 2020, 115, 104448. [Google Scholar] [CrossRef]
  203. Li, J.; Tian, J.; Lu, J.; Wang, Z.; Ling, J.; Wu, X.; Yang, F.; Xia, Y. LncRNA GAS5 inhibits Th17 differentiation and alleviates immune thrombocytopenia via promoting the ubiquitination of STAT3. Int. Immunopharmacol. 2020, 80, 106127. [Google Scholar] [CrossRef] [PubMed]
  204. Xia, D.; Yao, R.; Zhou, P.; Wang, C.; Xia, Y.; Xu, S. LncRNA NEAT1 reversed the hindering effects of miR-495-3p/STAT3 axis and miR-211/PI3K/AKT axis on sepsis-relevant inflammation. Mol. Immunol. 2020, 117, 168–179. [Google Scholar] [CrossRef] [PubMed]
  205. Du, C.; Wang, H.X.; Chen, P.; Chen, C.H. STAT3-induced upregulation of lncRNA DUXAP8 functions as ceRNA for miR-577 to promote the migration and invasion in colorectal cancer through the regulation of RAB14. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 6105–6118. [Google Scholar] [CrossRef] [PubMed]
  206. Gao, H.; Liu, R.; Sun, X. STAT3-induced upregulation of lncRNA SNHG17 predicts a poor prognosis of melanoma and promotes cell proliferation and metastasis through regulating PI3K-AKT pathway. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 8000–8010. [Google Scholar] [CrossRef]
  207. Zeng, X.Y.; Jiang, X.Y.; Yong, J.H.; Xie, H.; Yuan, J.; Zeng, D.; Dou, Y.Y.; Xiao, S.S. lncRNA ABHD11-AS1, regulated by the EGFR pathway, contributes to the ovarian cancer tumorigenesis by epigenetically suppressing TIMP2. Cancer Med. 2019, 8, 7074–7085. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  208. Sun, Q.; Li, Q.; Xie, F. LncRNA-MALAT1 regulates proliferation and apoptosis of ovarian cancer cells by targeting miR-503-5p. OncoTargets Ther. 2019, 12, 6297–6307. [Google Scholar] [CrossRef] [Green Version]
  209. Luo, J.; Wang, K.; Yeh, S.; Sun, Y.; Liang, L.; Xiao, Y.; Xu, W.; Niu, Y.; Cheng, L.; Maity, S.N.; et al. LncRNA-p21 alters the antiandrogen enzalutamide-induced prostate cancer neuroendocrine differentiation via modulating the EZH2/STAT3 signaling. Nat. Commun. 2019, 10, 2571. [Google Scholar] [CrossRef] [Green Version]
  210. Su, W.; Guo, C.; Wang, L.; Wang, Z.; Yang, X.; Niu, F.; Tzou, D.; Yang, X.; Huang, X.; Wu, J.; et al. LncRNA MIR22HG abrogation inhibits proliferation and induces apoptosis in esophageal adenocarcinoma cells via activation of the STAT3/c-Myc/FAK signaling. Aging 2019, 11, 4587–4596. [Google Scholar] [CrossRef]
  211. Chen, J.F.; Wu, P.; Xia, R.; Yang, J.; Huo, X.Y.; Gu, D.Y.; Tang, C.J.; De, W.; Yang, F. STAT3-induced lncRNA HAGLROS overexpression contributes to the malignant progression of gastric cancer cells via mTOR signal-mediated inhibition of autophagy. Mol. Cancer 2018, 17, 6. [Google Scholar] [CrossRef] [Green Version]
  212. Jin, Y.; Wu, P.; Zhao, W.; Wang, X.; Yang, J.; Huo, X.; Chen, J.; De, W.; Yang, F. Long noncoding RNA LINC00165-induced by STAT3 exerts oncogenic properties via interaction with Polycomb Repressive Complex 2 to promote EMT in gastric cancer. Biochem. Biophys. Res. Commun. 2018, 507, 223–230. [Google Scholar] [CrossRef]
  213. Guo, J.Q.; Yang, Z.J.; Wang, S.; Wu, Z.Z.; Yin, L.L.; Wang, D.C. LncRNA SNHG16 functions as an oncogene by sponging miR-200a-3p in pancreatic cancer. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 1718–1724. [Google Scholar] [CrossRef] [PubMed]
  214. Chen, W.; Jiang, T.; Mao, H.; Gao, R.; Zhang, H.; He, Y.; Liu, C.; Chen, Q. SNHG16 regulates invasion and migration of bladder cancer through induction of epithelial-to-mesenchymal transition. Hum. Cell 2020. [Google Scholar] [CrossRef] [PubMed]
  215. Shao, M.; Yu, Z.; Zou, J. LncRNA-SNHG16 Silencing Inhibits Prostate Carcinoma Cell Growth, Downregulate GLUT1 Expression and Reduce Glucose Uptake. Cancer Manag. Res. 2020, 12, 1751–1757. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. Jiang, D.; Li, H.; Xiang, H.; Gao, M.; Yin, C.; Wang, H.; Sun, Y.; Xiong, M. Long Chain Non-Coding RNA (lncRNA) HOTAIR Knockdown Increases miR-454-3p to Suppress Gastric Cancer Growth by Targeting STAT3/Cyclin D1. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2019, 25, 1537–1548. [Google Scholar] [CrossRef]
  217. Zhan, G.; Hu, J.; Xiao, B.; Wang, X.; Yang, Z.; Yang, G.; Lu, L. Trillin prevents proliferation and induces apoptosis through inhibiting STAT3 nuclear translocation in hepatoma carcinoma cells. Med. Oncol. (Northwoodlondonengland) 2020, 37, 44. [Google Scholar] [CrossRef]
  218. Zhang, N.; Zhang, M.; Wang, Z.; Gao, W.; Sun, Z.G. Activated STAT3 Could Reduce Survival in Patients with Esophageal Squamous Cell Carcinoma by Up-regulating VEGF and Cyclin D1 Expression. J. Cancer 2020, 11, 1859–1868. [Google Scholar] [CrossRef] [Green Version]
  219. Zhao, J.; Du, P.; Cui, P.; Qin, Y.; Hu, C.; Wu, J.; Zhou, Z.; Zhang, W.; Qin, L.; Huang, G. LncRNA PVT1 promotes angiogenesis via activating the STAT3/VEGFA axis in gastric cancer. Oncogene 2018, 37, 4094–4109. [Google Scholar] [CrossRef]
  220. Tan, H.Y.; Wang, C.; Liu, G.; Zhou, X. Long noncoding RNA NEAT1-modulated miR-506 regulates gastric cancer development through targeting STAT3. J. Cell. Biochem. 2019, 120, 4827–4836. [Google Scholar] [CrossRef]
  221. Shen, W.; Yuan, Y.; Zhao, M.; Li, J.; Xu, J.; Lou, G.; Zheng, J.; Bu, S.; Guo, J.; Xi, Y. Novel long non-coding RNA GACAT3 promotes gastric cancer cell proliferation through the IL-6/STAT3 signaling pathway. Tumor Biol. 2016, 37, 14895–14902. [Google Scholar] [CrossRef]
  222. Wang, X.; Kan, J.; Han, J.; Zhang, W.; Bai, L.; Wu, H. LncRNA SNHG16 Functions as an Oncogene by Sponging MiR-135a and Promotes JAK2/STAT3 Signal Pathway in Gastric Cancer. J. Cancer 2019, 10, 1013–1022. [Google Scholar] [CrossRef] [Green Version]
  223. Zheng, L.; Chen, J.; Zhou, Z.; He, Z. Knockdown of long non-coding RNA HOXD-AS1 inhibits gastric cancer cell growth via inactivating the JAK2/STAT3 pathway. Tumour Biol. J. Int. Soc. Oncodev. Biol. Med. 2017, 39, 1010428317705335. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Bartosch, C.; Monteiro-Reis, S.; Almeida-Rios, D.; Vieira, R.; Castro, A.; Moutinho, M.; Rodrigues, M.; Graça, I.; Lopes, J.M.; Jerónimo, C. Assessing sirtuin expression in endometrial carcinoma and non-neoplastic endometrium. Oncotarget 2016, 7, 1144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Cha, E.J.; Noh, S.J.; Kwon, K.S.; Kim, C.Y.; Park, B.-H.; Park, H.S.; Lee, H.; Chung, M.J.; Kang, M.J.; Lee, D.G. Expression of DBC1 and SIRT1 is associated with poor prognosis of gastric carcinoma. Clin. Cancer Res. 2009, 15, 4453–4459. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. Yang, Q.; Wang, B.; Gao, W.; Huang, S.; Liu, Z.; Li, W.; Jia, J. SIRT1 is downregulated in gastric cancer and leads to G1-phase arrest via NF-κB/Cyclin D1 signaling. Mol. Cancer Res. 2013, 11, 1497–1507. [Google Scholar] [CrossRef] [Green Version]
  227. Zhang, S.; Yang, Y.; Huang, S.; Deng, C.; Zhou, S.; Yang, J.; Cao, Y.; Xu, L.; Yuan, Y.; Yang, J.; et al. SIRT1 inhibits gastric cancer proliferation and metastasis via STAT3/MMP-13 signaling. J. Cell. Physiol. 2019, 234, 15395–15406. [Google Scholar] [CrossRef]
  228. Zhou, J.; Wu, A.; Yu, X.; Zhu, J.; Dai, H. SIRT6 inhibits growth of gastric cancer by inhibiting JAK2/STAT3 pathway. Oncol. Rep. 2017, 38, 1059–1066. [Google Scholar] [CrossRef] [Green Version]
  229. Janacova, L.; Faktor, J.; Capkova, L.; Paralova, V.; Pospisilova, A.; Podhorec, J.; Ebhardt, H.A.; Hrstka, R.; Nenutil, R.; Aebersold, R.; et al. SWATH-MS Analysis of FFPE Tissues Identifies Stathmin as a Potential Marker of Endometrial Cancer in Patients Exposed to Tamoxifen. J. Proteome Res. 2020. [Google Scholar] [CrossRef]
  230. Xu, J.; Wu, W.; Tang, Y.; Lin, Y.; Xue, Y.; Hu, J.; Lin, D. PRL-3 exerts oncogenic functions in myeloid leukemia cells via aberrant dephosphorylation of stathmin and activation of STAT3 signaling. Aging 2019, 11, 7817–7829. [Google Scholar] [CrossRef]
  231. Dos Santos Passaia, B.; Lima, K.; Kremer, J.L.; da Conceicao, B.B.; de Paula Mariani, B.M.; da Silva, J.C.L.; Zerbini, M.C.N.; Fragoso, M.; Machado-Neto, J.A.; Lotfi, C.F.P. Stathmin 1 is highly expressed and associated with survival outcome in malignant adrenocortical tumours. Investig. New Drugs 2020, 38, 899–908. [Google Scholar] [CrossRef]
  232. Shu, F.; Zou, X.; Tuo, H.; She, S.; Huang, J.; Ren, H.; Hu, H.; Peng, S.; Wang, J.; Yang, Y. Stathmin gene silencing suppresses proliferation, migration and invasion of gastric cancer cells via AKT/sCLU and STAT3 signaling. Int. J. Oncol. 2019, 54, 1086–1098. [Google Scholar] [CrossRef] [Green Version]
  233. Cassim, S.; Vucetic, M.; Zdralevic, M.; Pouyssegur, J. Warburg and Beyond: The Power of Mitochondrial Metabolism to Collaborate or Replace Fermentative Glycolysis in Cancer. Cancers 2020, 12, 1119. [Google Scholar] [CrossRef] [PubMed]
  234. Yao, Z.; Zhang, Q.; Guo, F.; Guo, S.; Yang, B.; Liu, B.; Li, P.; Li, J.; Guan, S.; Liu, X. Long Noncoding RNA PCED1B-AS1 Promotes the Warburg Effect and Tumorigenesis by Upregulating HIF-1alpha in Glioblastoma. Cell Transplant. 2020, 29, 963689720906777. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  235. Gao, S.; Chen, M.; Wei, W.; Zhang, X.; Zhang, M.; Yao, Y.; Lv, Y.; Ling, T.; Wang, L.; Zou, X. Crosstalk of mTOR/PKM2 and STAT3/c-Myc signaling pathways regulate the energy metabolism and acidic microenvironment of gastric cancer. J. Cell. Biochem. 2018. [Google Scholar] [CrossRef] [PubMed]
  236. Ren, Y.; Xu, X.; Mao, C.-Y.; Han, K.-K.; Xu, Y.-J.; Cao, B.-Y.; Zhang, Z.-B.; Sethi, G.; Tang, X.-W.; Mao, X.-L. RNF6 promotes myeloma cell proliferation and survival by inducing glucocorticoid receptor polyubiquitination. Acta Pharmacol. Sin. 2020, 41, 394–403. [Google Scholar] [CrossRef] [PubMed]
  237. Zhu, H.; Wang, C. Potential Influences of RNF6 on Prognosis and Metastasis of Colorectal Cancer: A Clinical Analysis. OncoTargets Ther. 2020, 13, 2031–2036. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  238. Huang, Z.; Cai, Y.; Yang, C.; Chen, Z.; Sun, H.; Xu, Y.; Chen, W.; Xu, D.; Tian, W.; Wang, H. Knockdown of RNF6 inhibits gastric cancer cell growth by suppressing STAT3 signaling. OncoTargets Ther. 2018, 11, 6579–6587. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  239. Zhang, X.; Liu, Y.; Huang, W.C.; Zheng, L.C. MiR-125b-1-3p Exerts Antitumor Functions in Lung Carcinoma Cells by Targeting S1PR1. Chin. Med. J. 2018, 131, 1909–1916. [Google Scholar] [CrossRef]
  240. Lankadasari, M.B.; Aparna, J.S.; Mohammed, S.; James, S.; Aoki, K.; Binu, V.S.; Nair, S.; Harikumar, K.B. Targeting S1PR1/STAT3 loop abrogates desmoplasia and chemosensitizes pancreatic cancer to gemcitabine. Theranostics 2018, 8, 3824–3840. [Google Scholar] [CrossRef]
  241. Song, S.; Min, H.; Niu, M.; Wang, L.; Wu, Y.; Zhang, B.; Chen, X.; Liang, Q.; Wen, Y.; Wang, Y.; et al. S1PR1 predicts patient survival and promotes chemotherapy drug resistance in gastric cancer cells through STAT3 constitutive activation. EBioMedicine 2018, 37, 168–176. [Google Scholar] [CrossRef] [Green Version]
  242. Tao, Z.; Ruan, H.; Sun, L.; Kuang, D.; Song, Y.; Wang, Q.; Wang, T.; Hao, Y.; Chen, K. Targeting the YB-1/PD-L1 Axis to Enhance Chemotherapy and Antitumor Immunity. Cancer Immunol. Res. 2019, 7, 1135–1147. [Google Scholar] [CrossRef]
  243. Liu, Y.L.; Yan, Z.X.; Xia, Y.; Xie, X.Y.; Zhou, K.; Xu, L.L.; Shi, Y.L.; Wang, Q.; Bi, J.W. Ligustrazine reverts anthracycline chemotherapy resistance of human breast cancer by inhibiting JAK2/STAT3 signaling and decreasing fibrinogen gamma chain (FGG) expression. Am. J. Cancer Res. 2020, 10, 939–952. [Google Scholar] [PubMed]
  244. Chua, P.J.; Lim, J.P.; Guo, T.T.; Khanna, P.; Hu, Q.; Bay, B.H.; Baeg, G.H. Y-box binding protein-1 and STAT3 independently regulate ATP-binding cassette transporters in the chemoresistance of gastric cancer cells. Int. J. Oncol. 2018, 53, 2579–2589. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  245. Wang, Y.; Wang, H.; Yao, H.; Li, C.; Fang, J.Y.; Xu, J. Regulation of PD-L1: Emerging Routes for Targeting Tumor Immune Evasion. Front. Pharmacol. 2018, 9, 536. [Google Scholar] [CrossRef] [PubMed]
  246. Sun, C.; Mezzadra, R.; Schumacher, T.N. Regulation and Function of the PD-L1 Checkpoint. Immunity 2018, 48, 434–452. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  247. Zhao, L.; Liu, Y.; Zhang, J.; Liu, Y.; Qi, Q. LncRNA SNHG14/miR-5590-3p/ZEB1 positive feedback loop promoted diffuse large B cell lymphoma progression and immune evasion through regulating PD-1/PD-L1 checkpoint. Cell Death Dis. 2019, 10, 731. [Google Scholar] [CrossRef] [PubMed]
  248. Zheng, X.; Dong, L.; Wang, K.; Zou, H.; Zhao, S.; Wang, Y.; Wang, G. MiR-21 Participates in the PD-1/PD-L1 Pathway-Mediated Imbalance of Th17/Treg Cells in Patients After Gastric Cancer Resection. Ann. Surg. Oncol. 2019, 26, 884–893. [Google Scholar] [CrossRef]
  249. Derer, A.; Frey, B.; Fietkau, R.; Gaipl, U.S. Immune-modulating properties of ionizing radiation: Rationale for the treatment of cancer by combination radiotherapy and immune checkpoint inhibitors. Cancer Immunol. Immunother. 2016, 65, 779–786. [Google Scholar] [CrossRef]
  250. Yan, Y.; Kumar, A.B.; Finnes, H.; Markovic, S.N.; Park, S.; Dronca, R.S.; Dong, H. Combining Immune Checkpoint Inhibitors with Conventional Cancer Therapy. Front. Immunol. 2018, 9, 1739. [Google Scholar] [CrossRef] [Green Version]
  251. Chen, M.F.; Chen, P.T.; Chen, W.C.; Lu, M.S.; Lin, P.Y.; Lee, K.D. The role of PD-L1 in the radiation response and prognosis for esophageal squamous cell carcinoma related to IL-6 and T-cell immunosuppression. Oncotarget 2016, 7, 7913–7924. [Google Scholar] [CrossRef]
  252. Sun, L.; Wang, Q.; Chen, B.; Zhao, Y.; Shen, B.; Wang, H.; Xu, J.; Zhu, M.; Zhao, X.; Xu, C.; et al. Gastric cancer mesenchymal stem cells derived IL-8 induces PD-L1 expression in gastric cancer cells via STAT3/mTOR-c-Myc signal axis. Cell Death Dis. 2018, 9, 928. [Google Scholar] [CrossRef]
  253. Gasi Tandefelt, D.; Boormans, J.L.; van der Korput, H.A.; Jenster, G.W.; Trapman, J. A 36-gene signature predicts clinical progression in a subgroup of ERG-positive prostate cancers. Eur. Urol. 2013, 64, 941–950. [Google Scholar] [CrossRef] [PubMed]
  254. Zheng, P.; Wang, W.; Ji, M.; Zhu, Q.; Feng, Y.; Zhou, F.; He, Q. TMEM119 silencing inhibits cell viability and causes the apoptosis of gastric cancer SGC-7901 cells. Oncol. Lett. 2018, 15, 8281–8286. [Google Scholar] [CrossRef] [PubMed]
  255. Zheng, P.; Wang, W.; Ji, M.; Zhu, Q.; Feng, Y.; Zhou, F.; He, Q. TMEM119 promotes gastric cancer cell migration and invasion through STAT3 signaling pathway. OncoTargets Ther. 2018, 11, 5835–5844. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  256. Yu, H.; Ma, J.; Chen, J.; Yang, Y.; Liang, J.; Liang, Y. LncRNA LINC00461 Promotes Colorectal Cancer Progression via miRNA-323b-3p/NFIB Axis. OncoTargets Ther. 2019, 12, 11119–11129. [Google Scholar] [CrossRef] [Green Version]
  257. Zhang, W.; Zhan, F.; Li, D.; Wang, T.; Huang, H. RGMB-AS1/miR-22-3p/NFIB axis contributes to the progression of gastric cancer. Neoplasma 2020, 67, 484–491. [Google Scholar] [CrossRef]
  258. Wu, C.; Zhu, X.; Liu, W.; Ruan, T.; Wan, W.; Tao, K. NFIB promotes cell growth, aggressiveness, metastasis and EMT of gastric cancer through the Akt/Stat3 signaling pathway. Oncol. Rep. 2018, 40, 1565–1573. [Google Scholar] [CrossRef]
  259. Brown, L.C.; Murphy, A.R.; Lalonde, C.S.; Subhedar, P.D.; Miller, A.H.; Stevens, J.S. Posttraumatic stress disorder and breast cancer: Risk factors and the role of inflammation and endocrine function. Cancer 2020. [Google Scholar] [CrossRef]
  260. Piotrowski, I.; Kulcenty, K.; Suchorska, W. Interplay between inflammation and cancer. Rep. Pract. Oncol. Radiother. J. Greatpoland Cancer Cent. Pozn. Pol. Soc. Radiat. Oncol. 2020, 25, 422–427. [Google Scholar] [CrossRef]
  261. 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]
  262. Brassart-Pasco, S.; Brézillon, S.; Brassart, B.; Ramont, L.; Oudart, J.-B.; Monboisse, J.C. Tumor Microenvironment: Extracellular Matrix Alterations Influence Tumor Progression. Front. Oncol. 2020, 10, 397. [Google Scholar] [CrossRef] [Green Version]
  263. Joyce, J.A.; Pollard, J.W. Microenvironmental regulation of metastasis. Nat. Reviews. Cancer 2009, 9, 239–252. [Google Scholar] [CrossRef] [PubMed]
  264. Lan, C.; Huang, X.; Lin, S.; Huang, H.; Cai, Q.; Wan, T.; Lu, J.; Liu, J. Expression of M2-polarized macrophages is associated with poor prognosis for advanced epithelial ovarian cancer. Technol. Cancer Res. Treat. 2013, 12, 259–267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  265. Herrera, M.; Herrera, A.; Dominguez, G.; Silva, J.; Garcia, V.; Garcia, J.M.; Gomez, I.; Soldevilla, B.; Munoz, C.; Provencio, M.; et al. Cancer-associated fibroblast and M2 macrophage markers together predict outcome in colorectal cancer patients. Cancer Sci. 2013, 104, 437–444. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  266. Liu, J.Y.; Peng, C.W.; Yang, G.F.; Hu, W.Q.; Yang, X.J.; Huang, C.Q.; Xiong, B.; Li, Y. Distribution pattern of tumor associated macrophages predicts the prognosis of gastric cancer. Oncotarget 2017, 8, 92757–92769. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  267. Yin, S.; Huang, J.; Li, Z.; Zhang, J.; Luo, J.; Lu, C.; Xu, H.; Xu, H. The Prognostic and Clinicopathological Significance of Tumor-Associated Macrophages in Patients with Gastric Cancer: A Meta-Analysis. PLoS ONE 2017, 12, e0170042. [Google Scholar] [CrossRef]
  268. Chen, L.; Shi, Y.; Zhu, X.; Guo, W.; Zhang, M.; Che, Y.; Tang, L.; Yang, X.; You, Q.; Liu, Z. IL10 secreted by cancerassociated macrophages regulates proliferation and invasion in gastric cancer cells via cMet/STAT3 signaling. Oncol. Rep. 2019, 42, 595–604. [Google Scholar] [CrossRef]
  269. Sawa-Wejksza, K.; Kandefer-Szerszeń, M. Tumor-associated macrophages as target for antitumor therapy. Arch. Immunol. Et Ther. Exp. 2018, 66, 97–111. [Google Scholar] [CrossRef] [Green Version]
  270. Mantovani, A.; Germano, G.; Marchesi, F.; Locatelli, M.; Biswas, S.K. Cancer-promoting tumor-associated macrophages: New vistas and open questions. Eur. J. Immunol. 2011, 41, 2522–2525. [Google Scholar] [CrossRef]
  271. Zheng, X.; Turkowski, K.; Mora, J.; Brüne, B.; Seeger, W.; Weigert, A.; Savai, R. Redirecting tumor-associated macrophages to become tumoricidal effectors as a novel strategy for cancer therapy. Oncotarget 2017, 8, 48436. [Google Scholar] [CrossRef] [Green Version]
  272. Yang, M.; Liu, J.; Shao, J.; Qin, Y.; Ji, Q.; Zhang, X.; Du, J. Cathepsin S-mediated autophagic flux in tumor-associated macrophages accelerate tumor development by promoting M2 polarization. Mol. Cancer 2014, 13, 43. [Google Scholar] [CrossRef] [Green Version]
  273. Zhou, Y.; Xia, L.; Liu, Q.; Wang, H.; Lin, J.; Oyang, L.; Chen, X.; Luo, X.; Tan, S.; Tian, Y.; et al. Induction of Pro-Inflammatory Response via Activated Macrophage-Mediated NF-kappaB and STAT3 Pathways in Gastric Cancer Cells. Cell. Physiol. Biochem. Int. J. Exp. Cell. Physiol. Biochem. Pharmacol. 2018, 47, 1399–1410. [Google Scholar] [CrossRef] [PubMed]
  274. Fu, X.L.; Duan, W.; Su, C.Y.; Mao, F.Y.; Lv, Y.P.; Teng, Y.S.; Yu, P.W.; Zhuang, Y.; Zhao, Y.L. Interleukin 6 induces M2 macrophage differentiation by STAT3 activation that correlates with gastric cancer progression. Cancer Immunol. Immunother. 2017, 66, 1597–1608. [Google Scholar] [CrossRef] [PubMed]
  275. Holthof, L.C.; Mutis, T. Challenges for Immunotherapy in Multiple Myeloma: Bone Marrow Microenvironment-Mediated Immune Suppression and Immune Resistance. Cancers 2020, 12, 988. [Google Scholar] [CrossRef] [PubMed]
  276. Licarete, E.; Rauca, V.F.; Luput, L.; Drotar, D.; Stejerean, I.; Patras, L.; Dume, B.; Toma, V.A.; Porfire, A.; Gherman, C.; et al. Overcoming Intrinsic Doxorubicin Resistance in Melanoma by Anti-Angiogenic and Anti-Metastatic Effects of Liposomal Prednisolone Phosphate on Tumor Microenvironment. Int. J. Mol. Sci. 2020, 21, 2968. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  277. Calabretta, E.; Carlo-Stella, C. The Many Facets of CD38 in Lymphoma: From Tumor-Microenvironment Cell Interactions to Acquired Resistance to Immunotherapy. Cells 2020, 9, 802. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  278. Quail, D.F.; Joyce, J.A. Microenvironmental regulation of tumor progression and metastasis. Nat. Med. 2013, 19, 1423–1437. [Google Scholar] [CrossRef]
  279. Ma, J.; Song, X.; Xu, X.; Mou, Y. Cancer-Associated Fibroblasts Promote the Chemo-resistance in Gastric Cancer through Secreting IL-11 Targeting JAK/STAT3/Bcl2 Pathway. Cancer Res. Treat. J. Korean Cancer Assoc. 2019, 51, 194–210. [Google Scholar] [CrossRef] [Green Version]
  280. Zuo, E.; Lu, Y.; Yan, M.; Pan, X.; Cheng, X. Increased expression of hepcidin and associated upregulation of JAK/STAT3 signaling in human gastric cancer. Oncol. Lett. 2018, 15, 2236–2244. [Google Scholar] [CrossRef] [Green Version]
  281. Hill, D.G.; Yu, L.; Gao, H.; Balic, J.J.; West, A.; Oshima, H.; McLeod, L.; Oshima, M.; Gallimore, A.; D’Costa, K.; et al. Hyperactive gp130/STAT3-driven gastric tumourigenesis promotes submucosal tertiary lymphoid structure development. Int. J. Cancer 2018, 143, 167–178. [Google Scholar] [CrossRef]
  282. Tuo, H.; Shu, F.; She, S.; Yang, M.; Zou, X.Q.; Huang, J.; Hu, H.D.; Hu, P.; Ren, H.; Peng, S.F.; et al. Sorcin induces gastric cancer cell migration and invasion contributing to STAT3 activation. Oncotarget 2017, 8, 104258–104271. [Google Scholar] [CrossRef]
  283. Uen, Y.H.; Fang, C.L.; Lin, C.C.; Hseu, Y.C.; Hung, S.T.; Sun, D.P.; Lin, K.Y. Ceramide synthase 6 predicts the prognosis of human gastric cancer: It functions as an oncoprotein by dysregulating the SOCS2/JAK2/STAT3 pathway. Mol. Carcinog. 2018, 57, 1675–1689. [Google Scholar] [CrossRef] [PubMed]
  284. Ollila, S.; Domenech-Moreno, E.; Laajanen, K.; Wong, I.P.; Tripathi, S.; Pentinmikko, N.; Gao, Y.; Yan, Y.; Niemela, E.H.; Wang, T.C.; et al. Stromal Lkb1 deficiency leads to gastrointestinal tumorigenesis involving the IL-11-JAK/STAT3 pathway. J. Clin. Investig. 2018, 128, 402–414. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  285. Zhang, Q.; Xu, F.; Shi, Y.; Chen, Y.W.; Wang, H.P.; Yu, X.; Li, Y. C-X-C motif chemokine receptor 4 promotes tumor angiogenesis in gastric cancer via activation of JAK2/STAT3. Cell Biol. Int. 2017, 41, 854–862. [Google Scholar] [CrossRef] [PubMed]
  286. Gao, X.; Sun, J.; Huang, C.; Hu, X.; Jiang, N.; Lu, C. RNAi-mediated silencing of NOX4 inhibited the invasion of gastric cancer cells through JAK2/STAT3 signaling. Am. J. Transl. Res. 2017, 9, 4440–4449. [Google Scholar] [PubMed]
  287. Li, X.; Na, H.; Xu, L.; Zhang, X.; Feng, Z.; Zhou, X.; Cui, J.; Zhang, J.; Lin, F.; Yang, S.; et al. DC-SIGN mediates gastric cancer progression by regulating the JAK2/STAT3 signaling pathway and affecting LncRNA RP11-181G12.2 expression. Biomed. Pharmacother. Biomed. Pharmacother. 2020, 121, 109644. [Google Scholar] [CrossRef] [PubMed]
  288. Wang, C.; Shi, M.; Ji, J.; Cai, Q.; Jiang, J.; Zhang, H.; Zhu, Z.; Zhang, J. A self-enforcing HOXA11/Stat3 feedback loop promotes stemness properties and peritoneal metastasis in gastric cancer cells. Theranostics 2019, 9, 7628–7647. [Google Scholar] [CrossRef]
  289. Zhu, S.; Soutto, M.; Chen, Z.; Blanca Piazuelo, M.; Kay Washington, M.; Belkhiri, A.; Zaika, A.; Peng, D.; El-Rifai, W. Activation of IGF1R by DARPP-32 promotes STAT3 signaling in gastric cancer cells. Oncogene 2019, 38, 5805–5816. [Google Scholar] [CrossRef]
  290. Ikeda, T.; Nishita, M.; Hoshi, K.; Honda, T.; Kakeji, Y.; Minami, Y. Mesenchymal stem cell-derived CXCL16 promotes progression of gastric cancer cells by STAT3-mediated expression of Ror1. Cancer Sci. 2020, 111, 1254–1265. [Google Scholar] [CrossRef] [Green Version]
  291. Yuan, K.; Ye, J.; Liu, Z.; Ren, Y.; He, W.; Xu, J.; He, Y.; Yuan, Y. Complement C3 overexpression activates JAK2/STAT3 pathway and correlates with gastric cancer progression. J. Exp. Clin. Cancer Res. 2020, 39, 9. [Google Scholar] [CrossRef] [Green Version]
  292. Xu, Y.Y.; Guo, M.; Yang, L.Q.; Zhou, F.; Yu, C.; Wang, A.; Pang, T.H.; Wu, H.Y.; Zou, X.P.; Zhang, W.J.; et al. Regulation of CD44v6 expression in gastric carcinoma by the IL-6/STAT3 signaling pathway and its clinical significance. Oncotarget 2017, 8, 45848–45861. [Google Scholar] [CrossRef]
  293. Xu, L.; Zhou, R.; Yuan, L.; Wang, S.; Li, X.; Ma, H.; Zhou, M.; Pan, C.; Zhang, J.; Huang, N.; et al. IGF1/IGF1R/STAT3 signaling-inducible IFITM2 promotes gastric cancer growth and metastasis. Cancer Lett. 2017, 393, 76–85. [Google Scholar] [CrossRef] [PubMed]
  294. 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] [PubMed] [Green Version]
  295. Chen, G.; Tang, N.; Wang, C.; Xiao, L.; Yu, M.; Zhao, L.; Cai, H.; Han, L.; Xie, C.; Zhang, Y. TNF-alpha-inducing protein of Helicobacter pylori induces epithelial-mesenchymal transition (EMT) in gastric cancer cells through activation of IL-6/STAT3 signaling pathway. Biochem. Biophys. Res. Commun. 2017, 484, 311–317. [Google Scholar] [CrossRef] [PubMed]
  296. Mu, X.; Zhao, T.; Xu, C.; Shi, W.; Geng, B.; Shen, J.; Zhang, C.; Pan, J.; Yang, J.; Hu, S.; et al. Oncometabolite succinate promotes angiogenesis by upregulating VEGF expression through GPR91-mediated STAT3 and ERK activation. Oncotarget 2017, 8, 13174–13185. [Google Scholar] [CrossRef]
  297. Li, T.; Guo, H.; Zhao, X.; Jin, J.; Zhang, L.; Li, H.; Lu, Y.; Nie, Y.; Wu, K.; Shi, Y.; et al. Gastric Cancer Cell Proliferation and Survival Is Enabled by a Cyclophilin B/STAT3/miR-520d-5p Signaling Feedback Loop. Cancer Res. 2017, 77, 1227–1240. [Google Scholar] [CrossRef] [Green Version]
  298. Yuan, W.; Li, T.; Mo, X.; Wang, X.; Liu, B.; Wang, W.; Su, Y.; Xu, L.; Han, W. Knockdown of CMTM3 promotes metastasis of gastric cancer via the STAT3/Twist1/EMT signaling pathway. Oncotarget 2016, 7, 29507–29519. [Google Scholar] [CrossRef] [Green Version]
  299. Zhang, J.L.; Liu, X.Z.; Wang, P.Y.; Chen, G.W.; Jiang, Y.; Qiao, S.K.; Zhu, J.; Wang, X.; Pan, Y.S.; Liu, Y.C. Targeting HCCR expression resensitizes gastric cancer cells to chemotherapy via down-regulating the activation of STAT3. Sci. Rep. 2016, 6, 24196. [Google Scholar] [CrossRef] [Green Version]
  300. Huang, Y.; Yang, M.; Hu, H.; Zhao, X.; Bao, L.; Huang, D.; Song, L.; Li, Y. Mitochondrial GRIM-19 as a potential therapeutic target for STAT3-dependent carcinogenesis of gastric cancer. Oncotarget 2016, 7, 41404–41420. [Google Scholar] [CrossRef] [Green Version]
  301. Piao, J.Y.; Lee, H.G.; Kim, S.J.; Kim, D.H.; Han, H.J.; Ngo, H.K.; Park, S.A.; Woo, J.H.; Lee, J.S.; Na, H.K.; et al. Helicobacter pylori Activates IL-6-STAT3 Signaling in Human Gastric Cancer Cells: Potential Roles for Reactive Oxygen Species. Helicobacter 2016, 21, 405–416. [Google Scholar] [CrossRef]
  302. Zhao, G.; Zhu, G.; Huang, Y.; Zheng, W.; Hua, J.; Yang, S.; Zhuang, J.; Ye, J. IL-6 mediates the signal pathway of JAK-STAT3-VEGF-C promoting growth, invasion and lymphangiogenesis in gastric cancer. Oncol. Rep. 2016, 35, 1787–1795. [Google Scholar] [CrossRef] [Green Version]
  303. Wu, X.; Yang, T.; Liu, X.; Guo, J.N.; Xie, T.; Ding, Y.; Lin, M.; Yang, H. IL-17 promotes tumor angiogenesis through Stat3 pathway mediated upregulation of VEGF in gastric cancer. Tumour Biol. J. Int. Soc. Oncodev. Biol. Med. 2016, 37, 5493–5501. [Google Scholar] [CrossRef] [PubMed]
  304. Fukui, H.; Watari, J.; Zhang, X.; Ran, Y.; Tomita, T.; Oshima, T.; Hirota, S.; Miwa, H. Phosphorylated STAT3 expression linked to SOCS3 methylation is associated with proliferative ability of gastric mucosa in patients with early gastric cancer. Oncol. Lett. 2020, 19, 3542–3550. [Google Scholar] [CrossRef] [PubMed]
  305. Balic, J.J.; Saad, M.I.; Dawson, R.; West, A.J.; McLeod, L.; West, A.C.; D’Costa, K.; Deswaerte, V.; Dev, A.; Sievert, W.; et al. Constitutive STAT3 Serine Phosphorylation Promotes Helicobacter-Mediated Gastric Disease. Am. J. Pathol. 2020. [Google Scholar] [CrossRef] [PubMed]
  306. Padmanabhan, N.; Ushijima, T.; Tan, P. How to stomach an epigenetic insult: The gastric cancer epigenome. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 467–478. [Google Scholar] [CrossRef]
  307. Gordon, K.; Clouaire, T.; Bao, X.X.; Kemp, S.E.; Xenophontos, M.; de Las Heras, J.I.; Stancheva, I. Immortality, but not oncogenic transformation, of primary human cells leads to epigenetic reprogramming of DNA methylation and gene expression. Nucleic Acids Res. 2014, 42, 3529–3541. [Google Scholar] [CrossRef]
  308. Cai, W.; Wang, C.; Li, Y.; Yao, C.; Shen, L.; Liu, S.; Bao, X.; Schnable, P.S.; Girton, J.; Johansen, J.; et al. Genome-wide analysis of regulation of gene expression and H3K9me2 distribution by JIL-1 kinase mediated histone H3S10 phosphorylation in Drosophila. Nucleic Acids Res. 2014, 42, 5456–5467. [Google Scholar] [CrossRef] [Green Version]
  309. Qi, H.; Yang, Z.; Dai, C.; Wang, R.; Ke, X.; Zhang, S.; Xiang, X.; Chen, K.; Li, C.; Luo, J.; et al. STAT3 activates MSK1-mediated histone H3 phosphorylation to promote NFAT signaling in gastric carcinogenesis. Oncogenesis 2020, 9, 15. [Google Scholar] [CrossRef] [Green Version]
  310. Krstic, M.; Stojanovic, N.M.; Stojnev, S.; Radenkovic, G.; Cukuranovic Kokoris, J.; Mladenovic, B.; Jankovic Velickovic, L. Interplay between STAT3, Cell Adhesion Molecules and Angiogenesis-Related Parameters in Gastric Carcinoma. Does STAT3 Really Have a Prognostic Value? Medicina (Kaunaslithuania) 2019, 55, 300. [Google Scholar] [CrossRef] [Green Version]
  311. Wei, K.L.; Chou, J.L.; Chen, Y.C.; Jin, H.; Chuang, Y.M.; Wu, C.S.; Chan, M.W.Y. Methylomics analysis identifies a putative STAT3 target, SPG20, as a noninvasive epigenetic biomarker for early detection of gastric cancer. PLoS ONE 2019, 14, e0218338. [Google Scholar] [CrossRef]
  312. Mirnoori, S.M.; Shahangian, S.S.; Salehi, Z.; Mashayekhi, F.; Talesh Sasani, S.; Saedi, H.S. Influence of single nucleotide polymorphisms in pri-miR-124-1 and STAT3 genes on gastric cancer susceptibility. Br. J. Biomed. Sci. 2018, 75, 182–186. [Google Scholar] [CrossRef]
  313. Yu, L.; Wu, D.; Gao, H.; Balic, J.J.; Tsykin, A.; Han, T.S.; Liu, Y.D.; Kennedy, C.L.; Li, J.K.; Mao, J.Q.; et al. Clinical Utility of a STAT3-Regulated miRNA-200 Family Signature with Prognostic Potential in Early Gastric Cancer. Clin. Cancer Res. J. Am. Assoc. Cancer Res. 2018, 24, 1459–1472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  314. Zhang, S.; Huang, S.; Deng, C.; Cao, Y.; Yang, J.; Chen, G.; Zhang, B.; Duan, C.; Shi, J.; Kong, B.; et al. Co-ordinated overexpression of SIRT1 and STAT3 is associated with poor survival outcome in gastric cancer patients. Oncotarget 2017, 8, 18848–18860. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  315. Zhou, F.; Cheng, L.; Qiu, L.X.; Wang, M.Y.; Li, J.; Sun, M.H.; Yang, Y.J.; Wang, J.C.; Jin, L.; Wang, Y.N.; et al. Associations of potentially functional variants in IL-6, JAKs and STAT3 with gastric cancer risk in an eastern Chinese population. Oncotarget 2016, 7, 28112–28123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  316. Luo, J.; Yan, R.; He, X.; He, J. Constitutive activation of STAT3 and cyclin D1 overexpression contribute to proliferation, migration and invasion in gastric cancer cells. Am. J. Transl. Res. 2017, 9, 5671–5677. [Google Scholar] [PubMed]
  317. Law, S.K.; Micklem, K.J.; Shaw, J.M.; Zhang, X.P.; Dong, Y.; Willis, A.C.; Mason, D.Y. A new macrophage differentiation antigen which is a member of the scavenger receptor superfamily. Eur. J. Immunol. 1993, 23, 2320–2325. [Google Scholar] [CrossRef]
  318. Schaer, D.J.; Schaer, C.A.; Buehler, P.W.; Boykins, R.A.; Schoedon, G.; Alayash, A.I.; Schaffner, A. CD163 is the macrophage scavenger receptor for native and chemically modified hemoglobins in the absence of haptoglobin. Blood 2006, 107, 373–380. [Google Scholar] [CrossRef] [Green Version]
  319. Tarin, C.; Carril, M.; Martin-Ventura, J.L.; Markuerkiaga, I.; Padro, D.; Llamas-Granda, P.; Moreno, J.A.; Garcia, I.; Genicio, N.; Plaza-Garcia, S.; et al. Targeted gold-coated iron oxide nanoparticles for CD163 detection in atherosclerosis by MRI. Sci. Rep. 2015, 5, 17135. [Google Scholar] [CrossRef] [Green Version]
  320. Karrasch, T.; Brunnler, T.; Hamer, O.W.; Schmid, K.; Voelk, M.; Herfarth, H.; Buechler, C. Soluble CD163 is increased in patients with acute pancreatitis independent of disease severity. Exp. Mol. Pathol. 2015, 99, 236–239. [Google Scholar] [CrossRef]
  321. Rojo-Martinez, G.; Maymo-Masip, E.; Rodriguez, M.M.; Solano, E.; Goday, A.; Soriguer, F.; Valdes, S.; Chaves, F.J.; Delgado, E.; Colomo, N.; et al. Serum sCD163 levels are associated with type 2 diabetes mellitus and are influenced by coffee and wine consumption: Results of the [email protected] study. PLoS ONE 2014, 9, e101250. [Google Scholar] [CrossRef] [Green Version]
  322. Cheng, Z.; Zhang, D.; Gong, B.; Wang, P.; Liu, F. CD163 as a novel target gene of STAT3 is a potential therapeutic target for gastric cancer. Oncotarget 2017, 8, 87244–87262. [Google Scholar] [CrossRef] [Green Version]
  323. Nagano, O.; Saya, H. Mechanism and biological significance of CD44 cleavage. Cancer Sci. 2004, 95, 930–935. [Google Scholar] [CrossRef] [PubMed]
  324. Orian-Rousseau, V. CD44, a therapeutic target for metastasising tumours. Eur. J. Cancer (Oxf. Engl. 1990) 2010, 46, 1271–1277. [Google Scholar] [CrossRef] [PubMed]
  325. Zoller, M. CD44: Can a cancer-initiating cell profit from an abundantly expressed molecule? Nat. Rev. Cancer 2011, 11, 254–267. [Google Scholar] [CrossRef] [PubMed]
  326. Ishimoto, T.; Nagano, O.; Yae, T.; Tamada, M.; Motohara, T.; Oshima, H.; Oshima, M.; Ikeda, T.; Asaba, R.; Yagi, H.; et al. CD44 variant regulates redox status in cancer cells by stabilizing the xCT subunit of system xc(-) and thereby promotes tumor growth. Cancer Cell 2011, 19, 387–400. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  327. Nagano, O.; Okazaki, S.; Saya, H. Redox regulation in stem-like cancer cells by CD44 variant isoforms. Oncogene 2013, 32, 5191–5198. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  328. Zhang, S.; Balch, C.; Chan, M.W.; Lai, H.C.; Matei, D.; Schilder, J.M.; Yan, P.S.; Huang, T.H.; Nephew, K.P. Identification and characterization of ovarian cancer-initiating cells from primary human tumors. Cancer Res. 2008, 68, 4311–4320. [Google Scholar] [CrossRef] [Green Version]
  329. Yae, T.; Tsuchihashi, K.; Ishimoto, T.; Motohara, T.; Yoshikawa, M.; Yoshida, G.J.; Wada, T.; Masuko, T.; Mogushi, K.; Tanaka, H.; et al. Alternative splicing of CD44 mRNA by ESRP1 enhances lung colonization of metastatic cancer cell. Nat. Commun. 2012, 3, 883. [Google Scholar] [CrossRef] [Green Version]
  330. Fujii, S.; Ito, K.; Ito, Y.; Ochiai, A. Enhancer of zeste homologue 2 (EZH2) down-regulates RUNX3 by increasing histone H3 methylation. J. Biol. Chem. 2008, 283, 17324–17332. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  331. Kong, W.Q.; Liang, J.J.; Du, J.; Ye, Z.X.; Gao, P.; Liang, Y.L. Long Noncoding RNA DLX6-AS1 Regulates the Growth and Aggressiveness of Colorectal Cancer Cells Via Mediating miR-26a/EZH2 Axis. Cancer Biother. Radiopharm. 2020. [Google Scholar] [CrossRef]
  332. Chen, H.; Hou, G.; Yang, J.; Chen, W.; Guo, L.; Mao, Q.; Ge, J.; Zhang, X. SOX9-activated PXN-AS1 promotes the tumorigenesis of glioblastoma by EZH2-mediated methylation of DKK1. J. Cell. Mol. Med. 2020. [Google Scholar] [CrossRef] [Green Version]
  333. Pan, Y.M.; Wang, C.G.; Zhu, M.; Xing, R.; Cui, J.T.; Li, W.M.; Yu, D.D.; Wang, S.B.; Zhu, W.; Ye, Y.J.; et al. STAT3 signaling drives EZH2 transcriptional activation and mediates poor prognosis in gastric cancer. Mol. Cancer 2016, 15, 79. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  334. Wang, X.Y.; Wang, L.L.; Zheng, X.; Meng, L.N.; Lyu, B.; Jin, H.F. Expression of p-STAT3 and vascular endothelial growth factor in MNNG-induced precancerous lesions and gastric tumors in rats. World J. Gastrointest. Oncol. 2016, 8, 305–313. [Google Scholar] [CrossRef] [PubMed]
  335. Wu, P.; Wu, D.; Zhao, L.; Huang, L.; Shen, G.; Huang, J.; Chai, Y. Prognostic role of STAT3 in solid tumors: A systematic review and meta-analysis. Oncotarget 2016, 7, 19863–19883. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  336. He, S.; Liao, G.; Liu, Y.; Huang, L.; Kang, M.; Chen, L. Overexpression of STAT3/pSTAT3 was associated with poor prognosis in gastric cancer: A meta-analysis. Int. J. Clin. Exp. Med. 2015, 8, 20014–20023. [Google Scholar] [PubMed]
  337. Hajimoradi, M.; Mohammad Hassan, Z.; Ebrahimi, M.; Soleimani, M.; Bakhshi, M.; Firouzi, J.; Samani, F.S. STAT3 is Overactivated in Gastric Cancer Stem-Like Cells. Cell J. 2016, 17, 617–628. [Google Scholar] [CrossRef] [PubMed]
  338. Thiem, S.; Eissmann, M.F.; Elzer, J.; Jonas, A.; Putoczki, T.L.; Poh, A.; Nguyen, P.; Preaudet, A.; Flanagan, D.; Vincan, E.; et al. Stomach-Specific Activation of Oncogenic KRAS and STAT3-Dependent Inflammation Cooperatively Promote Gastric Tumorigenesis in a Preclinical Model. Cancer Res. 2016, 76, 2277–2287. [Google Scholar] [CrossRef] [Green Version]
  339. Balic, J.J.; Garama, D.J.; Saad, M.I.; Yu, L.; West, A.C.; West, A.J.; Livis, T.; Bhathal, P.S.; Gough, D.J.; Jenkins, B.J. Serine-Phosphorylated STAT3 Promotes Tumorigenesis via Modulation of RNA Polymerase Transcriptional Activity. Cancer Res. 2019, 79, 5272–5287. [Google Scholar] [CrossRef] [Green Version]
  340. Ji, K.; Zhang, L.; Zhang, M.; Chu, Q.; Li, X.; Wang, W. Prognostic Value and Clinicopathological Significance of p-stat3 Among Gastric Carcinoma Patients: A Systematic Review and Meta-Analysis. Medicine 2016, 95, e2641. [Google Scholar] [CrossRef]
  341. Sun, Y.; Guo, B.F.; Xu, L.B.; Zhong, J.T.; Liu, Z.W.; Liang, H.; Wen, N.Y.; Yun, W.J.; Zhang, L.; Zhao, X.J. Stat3-siRNA inhibits the growth of gastric cancer in vitro and in vivo. Cell Biochem. Funct. 2015, 33, 495–502. [Google Scholar] [CrossRef]
  342. Kashyap, D.; Tuli, H.S.; Yerer, M.B.; Sharma, A.; Sak, K.; Srivastava, S.; Pandey, A.; Garg, V.K.; Sethi, G.; Bishayee, A. Natural product-based nanoformulations for cancer therapy: Opportunities and challenges. Semin. Cancer Biol. 2019. [Google Scholar] [CrossRef]
  343. Hosseini, S.H.; Zohreh, N.; Karimi, N.; Gaeini, N.; Alipour, S.; Seidi, F.; Gholipour, N. Magnetic nanoparticles double wrapped into cross-linked salep/PEGylated carboxymethyl cellulose; a biocompatible nanocarrier for pH-triggered release of doxorubicin. Int. J. Biol. Macromol. 2020, 158, 994–1006. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The schematic representation of the STAT3 signaling pathway.
Figure 1. The schematic representation of the STAT3 signaling pathway.
Biology 09 00126 g001
Figure 2. Onco-suppressor microRNAs in the inhibition of STAT3 and the malignant behavior of gastric cancer cells.
Figure 2. Onco-suppressor microRNAs in the inhibition of STAT3 and the malignant behavior of gastric cancer cells.
Biology 09 00126 g002
Figure 3. Oncogene miRs that activate the STAT3 signaling pathway and promote the proliferation and invasion of GC cells.
Figure 3. Oncogene miRs that activate the STAT3 signaling pathway and promote the proliferation and invasion of GC cells.
Biology 09 00126 g003
Figure 4. Long non-coding RNAs and their downstream targets in the regulation of the STAT3 signaling pathway in GC cells.
Figure 4. Long non-coding RNAs and their downstream targets in the regulation of the STAT3 signaling pathway in GC cells.
Biology 09 00126 g004
Figure 5. STAT3 signaling pathway as an oncogene factor in GC cells.
Figure 5. STAT3 signaling pathway as an oncogene factor in GC cells.
Biology 09 00126 g005
Table 1. The role and regulation of STAT3 in different cancers.
Table 1. The role and regulation of STAT3 in different cancers.
Cancer TypeSignaling NetworkEffect on STAT3ResultsRefs
Breast cancerBHLH40-AS1/IL-6/STAT3InductionPromoting progression and proliferation[60]
IL-6/STAT3InductionRadiation induces STAT3-mediated inflammation and radio resistance[61]
MiR-454/VGLL4/STAT3InductionMiR-454 induces the STAT3 signaling pathway via VGLL4 downregulation, leading to cancer malignancy[62]
PAK1/STAT3InductionStimulation of the nuclear translocation of STAT3 and enhancing breast cancer stem cell proliferation[63]
SIRT4/IL-6/STAT3InhibitionSensitizing cancer cells to tamoxifen chemotherapy[64]
Prostate cancerIL-8/STAT3/MALAT1InductionSTAT3 upregulates the expression of MALAT1, leading to progression and proliferation[65]
MiR-17/JAK/STAT3InhibitionMiR-17 reduces the expression of pro-survival factors, such as Bcl-2, and induces apoptosis via STAT3 downregulation[66]
Bladder cancerMiR-4500/STAT3/CCR7InhibitionSuppressing migration and proliferation[67]
CD44/Akt/ERK/STAT3InductionInhibition of apoptosis and cell cycle arrest[68]
Lung cancerB7-H4/PD-1/STAT3InductionPromoting proliferation and invasion via immune evasion[69]
KCP10043F/STAT3InhibitionInduction of apoptotic cell death[70]
BIS/STAT3InductionReducing sensitivity of cancer cells to digoxin-mediated migration and growth inhibition[71]
GlioblastomaAnnexin-A2/STAT3/oncostatin M receptorInductionPromoting the proliferation and invasion of cancer cells[72]
Hsa-miR-181d/STAT3InhibitionGarcinol upregulates the expression of hsa-miR-181d to inhibit STAT3 and the malignancy of cancer cells[73]
TROP2/JAK2/STAT3InductionPromoting proliferation and migration[74]
Bradykinin B1 receptor/STAT3/IL-8InductionEnhancing malignant behavior[75]
Table 2. Targeting STAT3 in cancer therapy in clinical trials.
Table 2. Targeting STAT3 in cancer therapy in clinical trials.
Drug/Molecular PathwayEffect on STAT3Clinical Trial PhaseMajor OutcomesRefs
AZD9150InhibitionPhase IAnti-tumor activity in pre-clinical
models and clinical trial
[76]
GRIM19Inhibition-Sensitizing into radiotherapy[79]
--Phase ISTAT3 provides local progression[81]
NilotinibInhibitionPhase IIDiminution in cancer growth[82]
OPB-31121InhibitionPhase IHigh tolerance
Inhibition of tumor growth
[83]
Table 3. The regulation of STAT3 by miRs in gastric cancer.
Table 3. The regulation of STAT3 by miRs in gastric cancer.
MiiRMiR TypeCancer Cell LineEffect on STAT3Major OutcomesRefs
MiR-143Onco-suppressorGC cell lines (AGS, SNU-1, SNU-5, SNU-16, NCIN87 and KATOIII)DownregulationDisrupting the proliferation and invasion of cancer cells[96]
MiR-125aOnco-suppressorHuman GC cell lines MKN45, SGC7901 and NCI-N87DownregulationReducing the expression of HAS1 and interfering with the migration of cancer cells[99]
MiR-375Onco-suppressorHuman GC cell lines BGC-823, AGS, SGC-7901 and MKN-45DownregulationInhibiting proliferation and migration via STAT3 downregulation[105]
MiR-148aOnco-suppressorGC cell lines SNU-1 (ATCC: CRL-5971), SNU-16 (ATCC: CRL-5974), AGS (ATCC: CRL-1739), NCI-N87 (ATCC: CRL-5822) and KATOIII (ATCC: HTB-103)DownregulationSuppressing growth and metastasis by the downregulation of CCK-BR via STAT3 downregulation[108]
MiR-874Onco-suppressorHuman GC cell lines AGS and BGC823, MKN28 and SGC-7901, as well as the human normal gastric epithelial cell line GES-1DownregulationDisrupting the STAT3/VEGF axis
Inhibition of angiogenesis and the malignancy of cancer cells
[117]
MiR-216aOnco-suppressorNormal human gastric epithelium cell line (GES-1) and GC cell lines (SGC-7901, MGC-803, MKN-28 and BGC-823)DownregulationSuppressing the metastasis of cancer cells via disrupting the STAT3/EMT axis[121]
MiR-93-5pOncogeneAGS and HEK293 cellsUpregulationDownregulation of the STAT3/MMP-9 axis
Inhibition of metastasis and invasion
[127]
MiR-18aOncogeneHuman GAC cell lines MKN28 and MKN1UpregulationSTAT3 induction
Promoting the malignant behavior of cancer cells
[131]
MiR-155OncogeneHuman GC cell lines BGC-823, NCI-N87, SGC-7901, AGS, MKN-45 and immortalized gastric mucosa epithelial cell line GES-1UpregulationStimulation of STAT3
Activation of MMP-2 and MMP-9
Enhancing invasion and migration of cancer cells
[132]
MiR-106a-3pOncogeneHuman GC cell line including SGC-7901 and BGC-823UpregulationStimulation of aptinib resistance
Activation of JAK2/STAT3 signaling
SOCS2, SOCS4 and SOCS5 downregulation
[133]
Table 4. Natural products as anti-tumor compounds in GC therapy via targeting STAT3.
Table 4. Natural products as anti-tumor compounds in GC therapy via targeting STAT3.
Anti-Tumor CompoundCell LineDoseDuration of ExperimentResultsRefs
PiperineTMK-1 human GC cell line25, 50 and 100 μM1 hDownregulation of STAT3
Inhibition of IL-1β and IL-6
Decreasing viability and proliferation of cancer cells
[175]
Tanshinone IIAHuman GC cell lines (SNU-638, MKN1 and AGS)2.5, 5 and 10 μg/mL12, 24, 48 and 72 hInhibition of STAT3
Reduction in the progression and malignancy of cancer cells
[176]
OxymatrineHuman GC cell lines SGC-7901, MGC-803, BGC-823, HGC-27, AGS and GES-10.5, 1, 2, 4 and 8 mg/mL24, 48 and 72 hDiminishing proliferation and malignancy of cancer cells
Inhibition of IL-21R-mediated STAT3
[177]
LuteolinGastric tumor cell lines of SGC7901, SGC7901/DDP, HGC27, MGC803, BGC803 and BGC82310 μM-Selective eradication of STAT3 overexpression-GC cells
Increasing the binding of STAT3 to SHP-1
[178]
ParthenolideHuman GC drug-resistant SGC-7901/DDP cell line1.25, 2.5, 5 and 10 μmol/L24, 48 and 72 hInduction of apoptosis
Inhibition of drug resistance via the downregulation of STAT3
[179]
Curcumin analogueHuman GC cell lines (BGC-823, SGC-7901)0.5, 1, 5, 10, 20, 50, 80 and 100 μM24 and 48 hInduction of apoptosis and mitotic arrest
Downregulation of STAT3
[180]
NifuratelHuman GC cell lines SGC-7901 and BGC-82375, 150 and 300 μM24 hInhibition of IL-6-mediated STAT3 activation[181]
CryptotanshinoneHuman GC cell lines SGC-7901 and HGC-272.5, 5, 7.5, 10, 15 and 20 μM4 hEnhancing anti-tumor activity of doxorubicin
Inhibition of STAT3 phosphorylation
[182]
Asiatic acidSGC7901 (metastatic carcinoma of lymph node)1, 5, 10, 25 and 50 μM12 hStimulation of apoptosis
Inhibition of proliferation and migration
Downregulation of STAT3
[183]
SulforaphaneHuman GC cell lines MGC803 and BGC8232.5, 5 and 10 μM72 hSensitizing cancer cells to chemotherapy
Suppressing cancer stem cell-like properties
Upregulation of miR-124 and subsequent downregulation of IL-6/STAT3
[184]
ThymoquinoneThree human GC cells (HGC27, BGC823 and SGC7901)25, 50 and 75 μmol/L24 hSuppressing STAT3 phosphorylation
Downregulation of survival factors such as Bcl-2 and cyclin-D
[185]
PaeoniflorinHuman gastric carcinoma MGC-803 cells and human normal gastric mucosa GES-1 cell lines5, 10 and 20 μmol/L48 hDownregulation of STAT3
Interfering with the proliferation and invasion of cancer cells
[186]
EupatilinHuman GC cell line MKN4550 and 100 μM16 hInhibiting the STAT3 signaling pathway
Suppressing VEGF and the growth of cancer cells
[187]
Epigallocatechin-6-gallateHuman gastric cancer (AGS) cells5, 10, 25 and 50 μmol/L24 hSuppressing IL-6/STAT3/VEGF results in growth inhibition[188]
Cucurbitacin BGC MKN-45 cells0.1, 1 and 10 μM12, 24 and 48 hSensitizing cancer cells to cell death
Downregulation of JAK2/STAT3
[189]
PonicidinHuman MKN28 cell line10, 25 and 50 μmol/L48 hInduction of apoptosis
Downregulation of JAK2/STAT3
[190]
CycloastragenolHuman gastric adenocarcinoma SNU-1 and SNU-16 cells1, 5, 10, 30 and 50 μM24 hInhibition of STAT3 phosphorylation at tyrosine 705 via suppressing Src and JAK1/2 activation
Induction of apoptosis
[191]
FucoxanthinSGC-7901 cells25, 50 and 75 μM24 hDownregulation of STAT3
Induction of apoptosis and cell cycle arrest
[192]
HJC0152 (niclosamide)Six GC cell lines (AGS, HGC-27, MKN28, MKN45, SGC7901 and BGC-823)5, 10 and 20 μM1, 2 and 4 hSuppressing the STAT3 signaling pathway and subsequent decrease in the expression of survival factors such as Survivin and Mcl-1[193]
PiceatannolHuman GC SGC-7901 cell line10 and 20 μM-Inhibiting STAT3 phosphorylation[194]
BP-1-102Five human GC cell lines (AGS, HGC-27, MKN28, MGC803 and SGC7901)2, 4 and 6 μM72 hSuppressing the invasion and proliferation of cancer cells in a dose- and time-dependent manner
Downregulation of STAT3
[195]
Table 5. lncRNAs as upstream modulators of STAT3 signaling pathway in GC cells.
Table 5. lncRNAs as upstream modulators of STAT3 signaling pathway in GC cells.
lncRNAType of lncRNADownstream SignalingCell LineEffect on STAT3Major ResultsRefs
SNHG16OncogeneMiR-135a/JAK2/STAT3Four GC cell lines (BGC823, MGC803, MKN45, SGC7901) and normal GC cell line GES-1InductionPromoting colony formation and the proliferation of cancer cells
Inhibition of apoptosis
[222]
HOTAIROncogeneMiR-454-3p/STAT3/cyclin D1AGS and SGC7901 cellsInductionKnock-down of HOTAIR
Sensitizing cancer cells to apoptosis
Upregulation of miR-454-3p and the subsequent inhibition of the STAT3/cyclin D1 axis
[216]
PVT1OncogeneSTAT3/VEGFGES-1, SGC-7901, BGC-823, MNK-45, AGS, SUN-638, HGC-27 and HUVECInductionPromoting angiogenesis and the growth of cancer cells[219]
NEAT1OncogeneMiR-506/STAT3BGC823, SGC-7901, AGS, MGC803, MKN28 cells, GES-1 and HEK-293T cellsInductionSponging miR-506
Enhancing expression of STAT3
Increasing the malignancy of cancer cells
[220]
GACAT3OncogeneIL-6/STAT3Human GC cell lines HGC-27 and SGC-7901InductionEnhancing the proliferation of cancer cells in an inflammatory response behavior[221]
HOXD-AS1OncogeneJAK2/STAT3Human GC cell lines (SGC-7901, BGC-823, MGC803 and MKN-45)InductionSilencing of HOXD-AS1 is correlated with the downregulation of STAT3 and growth inhibition[223]
Table 6. Molecular pathways targeting STAT3 in GC cells.
Table 6. Molecular pathways targeting STAT3 in GC cells.
Signaling NetworkCell LineEffect on STAT3ResultsRefs
CXCR4/JAK2/STAT3/VEGFHuman SGC-7901 and MKN45 cellsInductionInduction of VEGF by CXCR4
Subsequent activation of JAK2/STAT3
Enhancing the migration and proliferation of cancer cells
[285]
NOX4/JAK2/STAT3/EMTSix human GC cell lines (MKN-45, SGC-7901, MGC-803, BGC-823, MKN-28 and AGS)InductionInduction of JAK2/STAT3 by NOX4
Stimulation of EMT
Promoting invasion
[286]
DC-SIGNRHuman GC cell lines, SGC-7901, MGC-803, BGC-823 and AGS, and the control gastric epithelial cell line GES-1InductionEnsuring the growth and viability of cancer cells by the induction of the JAK2/STAT3 pathway[287]
HOXA11GC cell lines (KATO III, NCI-N87, SNU-16, AGS and SNU-16) and HEK 293TInductionPromoting the stemness and migration of cancer cells by the stimulation of STAT3[288]
TFF1/STAT3AGS cellsInductionEnhancing the proliferation and migration of cancer cells[67]
DARPP-32/IGF-1R/STAT3AGS cellsInductionPromoting proliferation and invasion[289]
BMX-ARHGAP/STAT3Four human GC cell lines (SNU-5, MNK-45, AGS and SGC7901) and the normal gastric epithelial cell line (GES-1)InductionMaintaining the carcinogenesis ability of GC stem cells[279]
CXCL16/STAT3/Ror1MKN45, MKN45-Luc and KATOIII cellsInductionIncreasing progression and malignancy[290]
Complement C3/JAK2/STAT3Human SGC-7901 and MGC-803 cells, normal gastric epithelial cells (GES-1)InductionPoor prognosis and enhanced proliferation of cancer cells[291]
BTF3/JAK2/STAT3/EMTHuman gastric epithelial cell line GES-1 and human GC cell lines, including AGS, HGC-27, MKN-28, MGC-803 and SGC-7901 cellsInductionInduction of JAK2/STAT3 by BTF3
Stimulation of EMT
Enhancing the proliferation and migration of cancer cells
[292]
IGF1/IGF1R/STAT3/IFITM2GC cell linesInductionEnhancing the growth and metastasis of cancer cells[293]
IL-6/JAK2/STAT3GC cell lines SNU-1, MKN45, SGC7901 and MKN28InductionSecretion of IL-6 by CAMs
Stimulation of JAK2/STAT3 by IL-6
[294]
TNF-α/IL-6/STAT3SGC7901 cellsInductionInduction of EMT
Ensuring the metastasis of cancer cells
[295]
Succinate/STAT3/VEGFHuman gastric mucosal epithelial cell line GES-1 and human GC cell lines AGS (low-differentiated human gastric adenocarcinoma), NCI-N87 (well-differentiated human carcinoma), BGC-823 (low-differentiated human gastric adenocarcinoma) and SGC-7901 (moderate-differentiated human gastric adenocarcinoma)InductionInduction of VEGF by succinate via STAT3 overexpression
Increasing viability and invasion of cancer cells
[296]
Cyclophilin B/STAT3/miR-520d-5pGC cell linesInductionThere is feedback consisting of the downregulation of miR-520d-5p and upregulation of cyclophilin B and STAT3, leading to the enhanced growth of cancer cells[297]
CMTM3/STAT3/Twist1/EMTHuman GC cell line SGC-7901InhibitionDownregulation of STAT3 by CMTM3
Suppressing metastasis
[298]
HCCR/STAT3Human GC cell lines AGS, MKN-45, BGC823, MGC803, HGC27, SGC7901, NCI-N87InductionTriggering chemoresistance[299]
GRIM19/STAT3Immortalized normal gastric epithelial cell line GES-1, human embryonic kidney HEK-293 cells, human GC SGC-7901 and BGC-823 cell linesInhibitionSuppressing STAT3
Induction of apoptosis
[300]
ROS/IL-6/STAT3AGS cellsInductionEnhanced generation of ROS by H. pylori
Stimulation of IL-6/STAT3
Enhanced proliferation and invasion of cancer cells
[301]
IL-6/STAT3/VEGFGC cell lines including SGC-7901, MGC, MKN-28 and AGSInductionPromoting invasion and angiogenesis
Induction of STAT3 and subsequent activation of VEGF
[302]
IL-17/STAT3/VEGFHuman GC (AGS) cells and other cells SGC7901, MKN 45 and BGC823InductionEnhancing growth
Induction of angiogenesis
Activation of STAT3/VEGF
[303]

Share and Cite

MDPI and ACS Style

Ashrafizadeh, M.; Zarrabi, A.; Orouei, S.; Zarrin, V.; Rahmani Moghadam, E.; Zabolian, A.; Mohammadi, S.; Hushmandi, K.; Gharehaghajlou, Y.; Makvandi, P.; et al. STAT3 Pathway in Gastric Cancer: Signaling, Therapeutic Targeting and Future Prospects. Biology 2020, 9, 126. https://doi.org/10.3390/biology9060126

AMA Style

Ashrafizadeh M, Zarrabi A, Orouei S, Zarrin V, Rahmani Moghadam E, Zabolian A, Mohammadi S, Hushmandi K, Gharehaghajlou Y, Makvandi P, et al. STAT3 Pathway in Gastric Cancer: Signaling, Therapeutic Targeting and Future Prospects. Biology. 2020; 9(6):126. https://doi.org/10.3390/biology9060126

Chicago/Turabian Style

Ashrafizadeh, Milad, Ali Zarrabi, Sima Orouei, Vahideh Zarrin, Ebrahim Rahmani Moghadam, Amirhossein Zabolian, Shima Mohammadi, Kiavash Hushmandi, Yashar Gharehaghajlou, Pooyan Makvandi, and et al. 2020. "STAT3 Pathway in Gastric Cancer: Signaling, Therapeutic Targeting and Future Prospects" Biology 9, no. 6: 126. https://doi.org/10.3390/biology9060126

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