Skip to main content

The role of microRNAs in the gastric cancer tumor microenvironment

Abstract

Background

Gastric cancer (GC) is one of the deadliest malignant tumors with unknown pathogenesis. Due to its treatment resistance, high recurrence rate, and lack of reliable early detection techniques, a majority of patients have a poor prognosis. Therefore, identifying new tumor biomarkers and therapeutic targets is essential. This review aims to provide fresh insights into enhancing the prognosis of patients with GC by summarizing the processes through which microRNAs (miRNAs) regulate the tumor microenvironment (TME) and highlighting their critical role in the TME.

Main text

A comprehensive literature review was conducted by focusing on the interactions among tumor cells, extracellular matrix, blood vessels, cancer-associated fibroblasts, and immune cells within the GC TME. The role of noncoding RNAs, known as miRNAs, in modulating the TME through various signaling pathways, cytokines, growth factors, and exosomes was specifically examined. Tumor formation, metastasis, and therapy in GC are significantly influenced by interactions within the TME. miRNAs regulate tumor progression by modulating these interactions through multiple signaling pathways, cytokines, growth factors, and exosomes. Dysregulation of miRNAs affects critical cellular processes such as cell proliferation, differentiation, angiogenesis, metastasis, and treatment resistance, contributing to the pathogenesis of GC.

Conclusions

miRNAs play a crucial role in the regulation of the GC TME, influencing tumor progression and patient prognosis. By understanding the mechanisms through which miRNAs control the TME, potential biomarkers and therapeutic targets can be identified to improve the prognosis of patients with GC.

Background

Gastric cancer (GC) is the fourth leading cause of cancer-related deaths worldwide and the fifth most prevalent disease globally [1]. This disease is often diagnosed at an advanced stage with metastases, as there are typically no early and accurate diagnostic methods or specific clinical symptoms. As a result, the 5-year survival rate for GC is only approximately 32% [2]. A noticeable process of connective tissue proliferation is present during solid tumor development, including that observed in GC. This process is intimately associated with immune cells, along with other types of mesenchymal stromal cells in the tumor microenvironment (TME) [3]. The TME includes fibroblasts, the extracellular matrix (ECM), blood vessels, endothelial cells, immune cells, and non-cellular elements such as cytokines and exosomes. Thus, the TME plays a significant role in cancer progression [4, 5] (Fig. 1). In this context, the external and internal environments in which the tumor cells are situated considerably affect the onset, development, and metastasis of the tumor, and they are both interdependent and competitive with each other [6]. Growth factors in the TME enhance the viability of tumor cells, reducing the uptake of chemotherapeutic agents or inactivating them. Additionally, the TME produces immunosuppressive factors, thereby promoting resistance to immunotherapy [7]. Therefore, understanding how the GC TME is regulated and applying these insights to clinical treatment is crucial to enhancing the poor prognosis of patients with GC.

Fig. 1
figure 1

TME composition in gastric cancer. TME: Tumor microenvironment

In 1993, Ambros et al. discovered the first microRNA (miRNA) in nematodes. This discovery revealed an important part of the noncoding genome that acts as a critical player in post-transcriptional gene regulation [8]. miRNA dysregulation has been identified in several human diseases, including heart disease, diabetes, cancer, and schizophrenia [9]. Dysregulation of miRNA expression levels in cancer has been associated with a range of biological features of human cancer development, including important roles in enhancing tumor cell proliferation, apoptosis, migration, epithelial-mesenchymal transition (EMT), metastasis, angiogenesis, autophagy, and interactions between malignant cells and the TME [10,11,12]. miRNA expression profiles in normal cells are very different from those in cancer tissues, and different tumor types and stages, including tumor development, progression, and metastasis, can be identified based on the expression of specific miRNAs [13, 14]. The regulation of miRNA expression has been associated with the suppression of oncogenic miRNAs and the replacement of tumor suppressor miRNAs [15]. Thus, miRNAs are a particularly important area of cancer research, with relevance to cancer prognosis, pathogenesis, diagnosis, and treatment, and are considered the perfect tool for improving cancer therapy [16].

Dysregulated miRNAs promote cancer-associated fibroblast (CAF) activation, inhibit myeloid-derived suppressor cells (MDSCs), inhibit T-cell differentiation, and facilitate angiogenesis, ultimately remodeling the TME [17]. Particularly, tumor cell-derived miRNAs are strongly associated with the production of an immunosuppressive TME and the loss of effector cells and reduced tumor immunogenicity; moreover, they are key determinants of cancer immune outcomes [18, 19]. Additionally, cancer cells secrete exosomes containing tumor suppressor miRNAs that propagate altered sets of miRNAs to different cellular compartments within the TME [20]. miRNAs may be key to immune-mediated tumor clearance, as miRNAs subtly repress genes and preferentially inhibit dose-sensitive targets [21].

Recently, miRNAs have been considered important potential biomarkers for gastric pathology, as they are frequently dysregulated in gastric tissues in preneoplastic lesions such as Helicobacter pylori infection, chronic gastritis, atrophic gastritis, and intestinal metaplasia, as well as in early-stage dysplasia and invasive cancers [22]. Meanwhile, increasing evidence indicates that miRNAs can be considered novel biomarkers; notably, many researchers have analyzed the miRNA profiles in serum and tissue samples from GC to assess their prognostic and diagnostic potential [23, 24] (Table 1). As previously described, miRNAs regulate mesenchymal interactions, immune invasion, and tumor angiogenesis, leading to malignant phenotypes of GC such as tumor growth, metastasis, angiogenesis, and drug resistance [25]. GC cells release extracellular vesicles (EVs) that are enriched in miR-1290. This miRNA enhances the inhibitory impact of GC cells on T-cell activation by targeting grainyhead-like 2 and activating the zinc finger E-box binding homeobox 1/programmed cell death ligand 1 (PD-L1) axis, facilitating GC cell immunological escape [26]. Drug resistance is one of the major challenges facing GC treatment, and manipulating miRNA expression has been shown to alleviate this therapeutic hurdle [27, 28]. Thus, miRNA-targeted GC therapies have great potential to enhance immunotherapy compared to existing therapies [29]. The investigation of microRNAs in GC have entered the clinical settings (Table 2).

Table 1 miRNAs that potentially represent GC biomarkers
Table 2 Summary of microRNAs in GC of clinical trials

A comprehensive understanding of the biological mechanisms facilitated by miRNAs in the TME of GC may, therefore, offer valuable perspectives for the identification of antitumor drugs and the advancement of targeted cancer treatments in the future. This review emphasizes the pivotal role that miRNAs play in the TME and focuses on how control of the TME by miRNAs influences GC development. Increased understanding of these processes may assist in the development of new therapies for patients with GC and the identification of new biomarkers that can improve management and follow-up strategies for patients with GC.

Main text

miRNAs and H. pylori/Epstein–Barr virus in the GC TME

H. pylori infection is one of the most significant risk factors for GC [66]. Immune monitoring of the gastric mucosa may be impeded by H. pylori-induced activation of signal transducer and activator of transcription 1 (STAT1) and PD-L1 expression, allowing malignant lesions to develop into GC [67]. The H. pylori virulence factor CagA affects multiple types of miRNAs in GC cells [68]. CagA inhibits proliferative and antitumor effects of CD8 + T cells and increases PD-L1 levels in GC cell-derived exosomes via suppressing miRNA-34a and P53 [69]. Moreover, CagA promotes miR-543 overexpression, which inhibits autophagy by targeting sirtuin 1, subsequently inducing EMT and triggering cell invasion and migration [70]. The phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signaling pathway is activated by H. pylori infection, which results in the TME secreting T regulatory cells (Tregs), suppressing tumor cell death, and enhancing the TME immunosuppressive state. This pathway activation aids in immune evasion, which in turn facilitates the development of tumors [71, 72]. On the contrary, H. pylori induces miR-223, which downregulates the expression of interleukin (IL)-6, IL-8, IL-1β, and tumor necrosis factor (TNF)-α and inhibits macrophage activation [73].

T helper 1 (Th1) and 17 (Th17) cell differentiation are influenced by miR-155 and contribute to immunity against H. pylori infection, along with infection-associated immunopathology [74]. However, Tsai et al. [75] noted that GC associated with H. pylori significantly increased miR-4286 and miR-18a-3p (5.73-fold and 6.02-fold, respectively). Moreover, invasion and miR-18a-3p, as well as lymph node metastases, tumor size, and tumor stage and miR-4286, have been shown to be significantly associated. Overexpression of miRNA-4286 and miR-18a-3p also inhibits benzodiazepine receptor-associated protein 1 expression while promoting the motility and proliferation of cancer cells. Furthermore, H. pylori infection induces IL-6, which affects STAT3 activity, inhibits miR-520d-5p expression, and activates the STAT3 and Janus kinase (JAK)/STAT pathway, leading to the proliferation of GC cells [76].

In H. pylori-infected T cells along with primary macrophages, miR-155 expression is dependent on forkhead box protein 3 (FOX3), indicating a potential functional relationship between the host immune response and miR-155 [77]. Huang et al. [78] indicated that miR-134 directly targets forkhead box protein M1 (FOXM1), and FOXM1 knockdown prevents the EMT induced by H. pylori CagA + /P + . Therefore, by targeting FOXM1, miR-134 suppresses invasion, proliferation, and EMT of SGC-7901 cells and may be protective against the GC process caused by H. pylori CagA + /P + (Fig. 2).

Fig. 2
figure 2

miRNAs and Helicobacter pylori in the gastric cancer TME. miRNAs: MicroRNAs, TME: Tumor microenvironment

Besides H. pylori, Epstein–Barr virus (EBV) is also a causative factor for GC [79]. EBV has been shown to be the first virus to encode its own miRNA. Immune escape is facilitated by EBV-encoded gene products, it-mediated epigenetic and structural variations, and miRNAs, which all assist in malignant transformation [80]. Furthermore, the EBV-miR-BART cluster, including miRBART-2, -4, -5, -18, and -22, is expressed in GC and linked to a poor prognosis [81]. Moreover, simultaneous infection with EBV also hinders the host response to H. pylori. Additionally, EBV synergism may strengthen the oncogenic potential of H. pylori CagA [82]. Notably, EMT-inducing transcription factors are induced in EBV-related GC upon downregulation of miR-200b and miR-200a [83]. In EBV-infected GC cells, miR-34a downregulation causes NADPH oxidase 2 upregulation, which promotes reactive oxygen species (ROS) generation and improves cell survival [84]. Notably, Choi et al. [85] determined that EBV-infected GC cells secrete miR-BART15-3p via exosomes that target the apoptosis inhibitor BRUCE. Subsequently, polybromo‐1 and FOXP1 separately suppress EBV-miR-BART17-3p along with EBV-miR-BART11 and increase PD-L1 transcription, thereby promoting tumor immune escape [86]. Low levels of viral antigen expression help EBV evade the host immune response. Additionally, viral miRNAs directly inhibit the release of the pro-inflammatory cytokine IL-12, thereby modulating the inflammatory response of T cells [87]. Moreover, viral miRNA-BART6-5p targets host cell Dicer and impairs host cell miRNA expression, thus helping EBV evade the host immune response and achieve chronic infection [88].

miRNAs regulate tumor angiogenesis in the GC TME

Angiogenesis plays a crucial role in cancer progression, as it is associated with immunosuppression and is essential for tumor growth, invasion, and metastasis [89, 90]. Based on the downstream targets of miRNAs, the expression of the most potent regulators of angiogenesis in different tumors has been extensively investigated, and a variety of miRNAs have been found to target angiogenic factors. Additionally to being a significant angiogenic agent, the vascular endothelial growth factor (VEGF) functions as an immunomodulator of the TME, promoting tumor-associated macrophages (TAMs) and Treg activation and preventing antigen presentation [91]. In addition to VEGF, phosphatase and tensin homolog (PTEN), mitogen-activated protein kinase (MAPK), and PI3K/AKT/mTOR are the major signaling pathways through which vascular-regulated miRNAs affect GC, and they are important mechanisms through which aberrant miRNAs regulate the development and progression of GC [92, 93].

Wu et al. [94] observed that miR-616-3p overexpression in GC triggers the downstream AKT/mTOR signaling pathway, targets PTEN, and facilitates EMT and angiogenesis [95]. Furthermore, miR-21 targets the tumor suppressor gene RECK, which is linked to tumor metastasis and angiogenesis, to cause cancer [96]. MiR-132 has been demonstrated to activate endothelial cells and targets p120RasGAP to induce pathological angiogenesis [97]. Additionally, exosomes, generated from GC cells that carried miR-23a, induced angiogenesis in a co-culture system by suppressing PTEN [98]. When GC cells overexpressed miR-574-3p and miR-210, VEGF and hypoxia-inducible factor 1-alpha (HIF-1α) were upregulated, leading to increased GC cell proliferation, migration, and invasion along with angiogenesis [99, 100]. Subsequently, reduction of invasion, migration, angiogenesis, and EMT resulting from overexpression of paired box 8 on GC cells is replicated by ectopic expression of miR-612 [101].

Meanwhile, miR-574-5p inhibits the expression of protein tyrosine phosphatase non-receptor type 3 and increases phosphorylation of p44/42 MAPKs in GC cells, which promotes angiogenesis [102]. Through its modulation of cancer stem cells (CSCs) and the EMT, the miR-29c-VEGFA/VEGFR2/extracellular signal-regulated kinases (ERK) signaling axis serves as a significant player in the course of GC metastatic disease, making it a prospective acts for GC clinical interventions [103].

With the rapid development of research on miRNAs, their function in tumor suppression through their anti-angiogenic function offers multifaceted therapeutic potential for these molecules. For example, miR-26a/b can directly act on VEGFA in GC, and its overexpression can directly suppress VEGF expression and reduce cell proliferation and angiogenesis, thereby inhibiting GC growth in mice [104]. Besides facilitating GC cell proliferation and migration, a reduction in miR-1 may also trigger pro-angiogenic signaling and encourage endothelial cell migration and proliferation [95]. Moreover, through suppression of VEGFA and fibroblast growth factor 1 expression, miR-205-5p inhibits angiogenesis in GC [105].

Furthermore, the PI3K/AKT signaling pathway mediates invasion, metastasis, angiogenesis, and lymphangiogenesis in GC after the downregulation of miR-30b-3p [106]. By suppressing ETS1 expression through a binding site in the 3′-UTR, miR-145 and miR-506 inhibit GC cell invasion, metastasis, and angiogenesis [107, 108]. Through the STAT3/VEGFA pathway, downregulation of miR-874 facilitates tumor angiogenesis in GC tissues [109]. In GC, miR-590 can concurrently modulate neuropilin 1 and VEGFR1/2. Furthermore, miR-590 overexpression can suppress GC cell migration, invasion, proliferation, and migration, as well as the release of D-MVA both in vivo and in vitro [110]. Similarly, miR-7 targets Raf-1 to suppress angiogenesis and tumorigenesis in GC cells [111].

Zhang et al. [112] confirmed the anti-angiogenic action of miR-218 in GC and demonstrated that tumor angiogenesis inhibition might be achieved therapeutically by administering miR-218. GC with low expression of miR-200C was markedly enriched for angiogenesis, hypoxia, TGF-β signaling genomes, and EMT, all of which contribute to tumor development and metastasis [113]. Interestingly, miR-29a-low GC is enriched for genes correlated with cell apoptosis, proliferation, angiogenesis, and metastasis; it is linked to less anticancer immune cell infiltration and immune-related scores [114] (Fig. 3).

Fig. 3
figure 3

miRNAs regulate angiogenesis in the development of gastric cancer. miRNAs: MicroRNAs

Peritoneal dissemination is the main cause of patient mortality and the most frequent reason for tumor progression following GC surgery. Notably, GC development and peritoneal dissemination are significantly influenced by angiogenesis [115], and increased expression of VEGF has been found to promote the production of malignant ascites [116]. Additionally, the expression pattern of miRNAs in peritoneal exosomes serves as a valuable diagnostic tool for peritoneal metastasis treatment, reflecting the tumor load within the abdominal cavity [117]. Transitioning from these observations, research into the regulatory mechanisms of miRNAs in tumor angiogenesis has made significant strides. Despite remaining obstacles, these rapidly evolving findings will make way for the future application of miRNAs as predictive biomarkers for anti-angiogenic therapy and miRNA-based antitumor angiogenesis strategies.

miRNAs regulate CAFs in the GC TME

As the major cells in the TME of solid tumors, fibroblasts are controlled by a multitude of factors released by immune or tumor cells [118]. CAFs express a wide range of pro-inflammatory molecules, including chemokines, interleukins, and components of the ECM, which eventually stimulate the growth of tumors by regulating the inflammation associated with the tumor or directing intercellular communication [119]. In the TME, miRNAs are implicated in the whole process of CAF generation and their functional execution, promoting cancer cell proliferation, drug resistance, and immunosuppression via secreting ECM proteins, inflammatory ligands, and growth factors [120].

The high levels of miR-27a observed in GC cell exosomes stimulate the metastasis, motility, and proliferation of cancer cells both in vitro and in vivo, as well as the reprogramming of fibroblasts into CAFs [121]. Meanwhile, another study reported that transformation of CAFs in GC was linked to miR-200b downregulation. Particularly, methylation of the miR-200b promoter was detected in GC cases exhibiting elevated expression of the CAF-specific marker α-smooth muscle actin [122]. However, in contrast to normal fibroblasts, the expression of miR-224-3p was lower in CAFs from patients with squamous GC, and miR-224-3p mimics were found to attenuate CAF migration and invasion [123]. miR-214 in CAFs directly modulates fibroblast growth factor 9 expression, which facilitates cell invasion and GC migration in vitro [124]. Likewise, miR-496 upregulates IL-33, which amplifies CAFs’ tumor-promoting properties by improving GC cell proliferation, EMT, migration, and invasion [125].

It has been confirmed that CAFs elevate miR-106b levels, targeting PTEN to facilitate cell invasion and migration [126]. Zhang et al. [127] demonstrated that the heterogeneous nuclear ribonucleoprotein A1 axis and ubiquitin-specific protease 7 are activated by paclitaxel and cisplatin, which makes it easier for miR-522 to be secreted from CAFs through the de-ubiquitination pathway. Furthermore, miR-522 targets arachidonic acid lipoxygenase 15, which also prevents ROS accumulation. This suppresses ferroptosis in GC cells, causing GC cells to become resistant to chemotherapy [127]. Moreover, by targeting the STAT4/Wnt/β-catenin axis, miR-141-3p suppresses normal fibroblasts from transforming into CAFs, which in turn inhibits GC invasion and migration [128] (Fig. 4). Overall, the activation and creation of CAFs are intimately linked to miRNA dysregulation, which plays a role in both executive function and CAF generation. These results offer fresh perspectives on the relationship between GC cells and CAFs.

Fig. 4
figure 4

miRNAs regulate CAFs in the development of gastric cancer. miRNAs: MicroRNAs

miRNAs regulate immunosuppressive cells in the GC TME

The TME consists of various stromal cells such as macrophages, T cells, MDSCs, Tregs, and the ECM; furthermore, blood vessels, lymphatic vessels, cytokines, mediators, and other non-cellular components are vital in defending the human body against pathogen invasion. These cells also impact GC through the modulation of immune responses and the elimination of mutated or damaged cells [129, 130]. miRNAs are implicated in the function and maintenance of Tregs and macrophage polarization, maintaining homeostasis in vivo under physiological conditions and driving immune tolerance or immunosuppression under pathological conditions [131].

Macrophages are a crucial component of both the innate and adaptive immune systems, playing key roles in pathogen defense and the regulation of body homeostasis [132]. The polarization of macrophages is influenced by the PI3K/AKT and JAK/STAT pathways, along with critical regulators such as the STAT family, peroxisome proliferation-activated receptor-g (PPARg), and interferon modulator [133, 134]. This process can lead to the development of TAMs in response to chemokines, cytokines, and other growth factors secreted by tumor cells, as well as tumor-associated conditions. TAMs may adopt the M1 phenotype, which exhibits antitumor activity, or the M2 phenotype, which supports tumor growth [135]. Predominantly, TAMs align with the M2 phenotype and are more likely to promote tumor progression [136]. In GC tissues and ascites, TAMs are abundant and may enhance GC cell migration and invasion through the secretion of EVs [137]. Moreover, dysregulation of miRNAs in tumors facilitates the shift of macrophage polarization from M1 to M2, adversely impacting TAM phenotypes and suppressing the immune response [138]. The involvement of TAMs in GC underscores their complex role, suggesting that miRNA-based reprogramming of TAM polarization could advance tumor immunotherapy.

According to Yun et al. [139], downregulating miR-30c under hypoxic environments decreased mTOR and glycolysis activity in TAMs in GC and further suppressed M1 macrophage differentiation and antitumor effects. With PTEN and IFN-γ/STAT1, miR-21 modulates TAMs, enhancing tumor cell motility and M2 polarization while lowering the expression of PD-L1 and M1 polarization to promote cancer progression [140]. Interestingly, TAMs deficient in miR-21 had an inflammatory gene signature, and antagonism of miR-21 increased the level of granzyme B, which enhanced the cytotoxicity of CD8 + T cells in immune TME [141].

Additionally, exosomes derived from M2 macrophages may transfer miR-487a into GC cells, possibly facilitating GC progression through the downregulation of T-cell intracellular antigen [142]. Exosomes derived from M2 macrophages produce miR-588, which targets cylindromatosis and enhances resistance to cisplatin in GC cells [143]. Moreover, exosome miR-21 translocates directly from TAMs to GC cells and modulates GC resistance to cisplatin by targeting PTEN, suppressing apoptosis, and activating the PI3K/AKT signaling pathway [144]. These studies highlight the importance of miRNA regulation of macrophages through key signaling pathways.

Compared with other intra-abdominal tumors, GC is more prone to peritoneal metastases, and the peritoneal immune microenvironment is critical for GC progression [145, 146]. Notably, TAM in malignant ascites of GC showed a significant M2-like phenotype, which promotes peritoneal metastasis of GC [147, 148]. Microarray analysis revealed a significant connection in GC tissue between the expression of miR-210 and CD204 + M2-like TAM infiltration. TNF-α, released by CD204 + M2-like TAMs, upregulates miR-210 through NF-κB/HIF-1α signaling to facilitate GC progression [149]. In summary, these candidate preclinical and clinical miRNAs underscore their roles as TME immune modulators and their therapeutic potential. A deeper understanding of how different miRNAs influence the M1/M2 balance could aid in developing targeted therapies to re-educate macrophages toward the M1 phenotype.

After antigenic stimulation, naive CD4 + T cells differentiate into multiple effector Th subpopulations with distinct phenotypes, such as Th1, Th2, Treg, and IL-17-producing Th17 [150]. Certain miRNAs have been shown to regulate T-cell differentiation. For example, the differentiation of Treg/Th17 and Th1 cells is inhibited by miR-23 and -27, whereas miR-24 facilitates their differentiation, creating an immunosuppressive microenvironment conducive to GC progression and metastasis [151]. Furthermore, the miR-192-5p/Rb1/NF-κBp65 signaling axis stimulates Treg differentiation by modulating IL-10 production in GC while also facilitating EMT in tumor cells [152]. Importantly, exosomes promote the differentiation of primary neoplastic Treg cells at the expense of antitumor Th1/Th17 differentiation, suggesting that tumor miRNAs can orchestrate immune evasion through multiple simultaneous mechanisms [153, 154]. Furthermore, the secretion of exosomal miR-451, which escalates under low-glycemic conditions and is subsequently transferred to T cells, supports the differentiation of T cells into Th17 cells by diminishing AMP-activated protein kinase and enhancing mTOR activity, marking a potential indicator of poor prognosis [155]. Additionally, a hypoxic TME reduces miR-34a expression, resulting in elevated lactate levels in GC tumor-infiltrating lymphocytes and a reduction in Th1 cells and cytotoxic T lymphocytes (CTLs), thereby compromising the immune efficacy of GCs [156].

Moreover, MDSCs, a heterogeneous group of myeloid-derived cells, facilitate tumor invasion and metastasis through diverse mechanisms, with tumor miRNAs directly governing the recruitment and functionality of MDSCs [157]. Notably, MDSCs characterized by the expression of the myeloid differentiation factor schlafen4 + , a regulator of myeloid differentiation, have been identified in GC, particularly in preneoplastic lesions infected with H. pylori [158]. miR-130b is increased in Schlafen4 + GC cells and promotes gastric epithelial cell proliferation, which is essential for MDSCs to suppress T-cell functions [159]. Exosomes secreted by GC deliver miR-107 to host MDSCs and induce their amplification and activation by targeting DICER1 and PTEN genes, thus providing new cancer therapeutic targets for GC [160]. Furthermore, miR-200C reduces PTEN and friend of Gata 2 expression, induces the PI3K/Akt cascade, promotes MDSCs amplification, and suppresses immune response in TME [161] (Fig. 5).

Fig. 5
figure 5

miRNAs regulate immunosuppressive cells in the development of gastric cancer. miRNAs: MicroRNAs, CAFs: Cancer-associated fibroblasts

The miRNAs can also directly affect immunosuppressive signaling, thereby altering the TME. Meanwhile, miR-4510 inhibits GC cell metastasis by altering immunosuppressive signals in the TME through the downregulation of glypican-3 [162]. miR-148b-5p deficiency results in immunological tolerance and GC development via the CSF1 and miR-148b-5p/ATPIF1/TNFa + IL6 axis [163].

These studies suggest that many miRNAs play essential roles in regulating TME-mediated immunosuppressive mechanisms. However, this area of research still needs to be further explored.

miRNAs modulate immunoreactive cells in the TME of GC

T cells are vital to maintaining health and preventing disease and are divided into two main subpopulations: CD4 + and CD8 + T-cell subpopulations [164]. Longer survival from cancer is linked to infiltration of CD8 + T cells; however, low immunogenicity of tumor cells in the TME inhibits T lymphocyte immunological activity, which reduces their antitumor capacity [165]. Post-transcriptional gene regulation via miRNAs has emerged as a major control mechanism for a variety of biological processes, including T-cell development and function [166]. Given that T cells can perform both pro-inflammatory and pro-absorptive tasks, identification and characterization of miRNAs associated with T-cell function will reveal miRNA-mediated mechanisms as therapeutic targets for immunotherapy against a wide range of diseases with inflammatory and immunosuppressive environments [167].

miRNAs regulate the expression of immune checkpoint ligands and protect tumors from T-cell-mediated lysis [168]. For instance, miR-105-5p, serving as a key player in the post-transcriptional suppression of PD-L1 in GC, prevents immunological escape resulting from upregulation of PD-L1 in cancer cells [169]. Furthermore, miR-424 has been identified as a potential inhibitor of the PD-L1/PD-1 pathway, and restoration of miR-424 expression reverses chemotherapy resistance [170].

miR-138 mainly modulates the immune system by interacting with CTLA-4 and PD-1 to repress tumor-infiltrating Tregs, thereby mitigating damage to immune-disordered cells in the TME [171]. Notably, H. pylori-positive GC has considerably higher PD-L1 expression levels, and miR-140 overexpression suppresses the proliferation and tumor growth of GC cells by blocking PD-L1 and mTOR activity [172]. Additionally, by repressing the expression of miR-513, reducing the translational repression of PD-L1, activating the pathway of JAK2/STAT1/IFR-1, and augmenting PD-L1 expression, INF-γ induces GC immune escape [173]. Notably, in vitro silencing of PD-1 enhances miR-21 expression, increases the proportion of Th17 cells, and decreases that of Treg cells [174]. miRNAs play an essential role in regulating the immune response, and miRNAs can interact with immune checkpoint inhibitors.

Additionally, elevated miR-152 levels improve immune responses by facilitating effector cytokine production and T-cell proliferation through the suppression of the B7-H1/PD-1 pathway. MiR-152 may be a potential therapeutic approach for GC [175]. Notably, manipulation of immune checkpoint protein expression by miRNA-based therapies combined with anti-immune checkpoint drugs may be an improved approach to GC treatment.

Dendritic cells (DCs) are the most potent antigen-presenting cells, capable of efficiently cross-presenting antigens. DCs contribute significantly to antitumor immunity by modulating the TME and attracting and activating anticancer T cells [176]. Thus, by impairing DC activation, antigen presentation, maturation, recruitment, and differentiation, TME and GC cells evade immune control [177]. Many miRNAs are implicated in the development and differentiation of DCs and in the regulation of inflammatory responses in DCs. Tumor miRNAs can directly or indirectly control DCs maturation and induce a tolerant state [178]. miR-17-5p decreased the secretion of TNF-α and IL-12 while increasing the production of IL-10. This shift inhibits the stimulation of T cells by DCs and promotes the expansion of Tregs. Furthermore, it can be utilized as a biomarker for GC originating from GC cells [179]. Additionally, in gastric TME, H. pylori can suppress miRNA-375 expression. This triggers the JAK2-STAT3 pathway, consequently promoting the release of VEGF, IL-10, and IL-6. These released factors promote DCs to differentiate immaturely and contribute to the induction of GC [180] (Fig. 6). These studies have demonstrated that miRNAs regulate the development, differentiation, and function of DCs, establishing them as pivotal regulators of the immune response. Another critical cellular component of innate immunity is the natural killer (NK) cells, which are essential in the immune response against cancer by killing tumor cells and secreting immunostimulatory cytokines [181]. Variations in miRNA expression influence the progression of NK and invariant NKT cells differently. For example, invariant NKT cells in the peripheral and thymus lymphoid organs are negatively regulated by miR-150 [182]. Conversely, miR-155 enhances NK cell function by increasing NKG2D, IFN-γ, and granzyme B production [183]. Furthermore, lncRNA-GAS5 enhances IFN-γ secretion by targeting miR-18a, thus promoting NK cell responses against GC cells [184]. In addition to modulating receptor signaling, miRNAs directly affect the production of effector molecules that determine NK cell activity.

Fig. 6
figure 6

miRNAs modulate immunoreactive cells in gastric cancer. miRNAs: MicroRNAs

Exosome-derived miRNAs regulate the GC TME

The discovery of exosomes and their multiple functions in cancer biology is undoubtedly one of the most exciting discoveries in recent years. Exosomes are nanoscale (30–150 nm in diameter) EVs that can transport a broad range of substances, including metabolites, proteins, lipids, and nucleic acids [185]. miRNAs in cancer-derived exosomes promote intercellular communication, targeting themselves and contributing to the regulation of multiple components of the immune system, ultimately modulating the TME to regulate GC development, metastasis, invasion, drug resistance, and angiogenesis [186, 187]. They are very valuable for the prognosis and early GC diagnosis and, to some extent, reflect the malignant characteristics of the tumor [188, 189]. Meanwhile, miRNAs play a role in the communication between tumor cells and TME through exosomal secretion and transport [190].

GC cells release exosomes containing miR-582-3p, which targets VEGF to stimulate cell invasion and proliferation [191]. Exosomes produced from GC cells carrying miR-135b have been found by Bai et al. [192] to lower FOXO1 protein levels and stimulate angiogenesis. GC cells can give rise to exosomes enriched in miR-301a-3p in hypoxic TMEs, which contribute to EMT, GC proliferation, invasion, and migration, along with HIF-1α accumulation [193]. Additionally, individuals with GC hepatic metastases demonstrate serum exosomes exhibiting considerably higher miR-519a-3p levels compared to individuals without liver metastases. Moreover, by targeting DUSP2, exosomal miR-519a-3p promotes the MAPK/ERK pathway, leading to M2-like polarization of macrophages, resulting in angiogenesis, facilitating the development of pre-metastatic niches in the liver, and accelerating the process of liver metastasis [194].

Exosomal miR-106a and miR-21-5p activates the TGF-β pathway by targeting TIMP2 and SMAD7, disrupts the mesothelial barrier, and promotes the peritoneal spread of GC by integrating into peritoneal mesothelial cells [195, 196]. Moreover, serum exosomes from patients with GC were enriched in miR-423-5p, and a significant correlation existed between lymph node metastasis and extracellular miR-423-5p levels, which facilitated cancer growth and metastasis [197].

Macrophages produce exosomes containing miR-16-5p that translocate to GC cells and target PD-L1 to activate T cells, thereby suppressing GC development [198]. Exosomes containing miR-21 are produced in tumors when the EMT transcription factor Snail activates miR-21. These exosomes are taken up by CD14 + human monocytes, which then cause a rise in M2 marker expression and ultimately accelerate tumor progression [199]. Furthermore, exosomal miR-15b-3p suppresses apoptosis in vivo and in vitro by inhibiting the expression of DYNLT1, cleaved caspase-3, and caspase-9. This promotes the proliferation, invasion, and migration of GC cells [200]. Additionally, GC cells secrete exosomes capable of delivering miR-107 to MDSCs, which causes the activation and amplification in MDSCs by targeting PTEN and DICER1 [160].

Notably, exosomal miR-122-5p inhibits both tumor development in vivo and GC cell migration and proliferation in vitro [201]. Furthermore, exosomal miR-139 produced by CAFs suppresses GC cell metastasis and tumor growth by decreasing the expression of matrix metalloproteinase 11 both in vitro and in vivo [202]. Moreover, exosomal miR-29b-1-5p generated from CAFs inhibits GC cell survival, invasion, and migration, as well as vascular mimicry development; however, it also stimulates apoptosis [203]. Additionally, CAF-derived EVs containing miR-199a-5p downregulate FKBP5, resulting in elevated AKT1 phosphorylation and mammalian target of rapamycin complex 1 activation, thereby promoting GC [204].

Chemotherapy is the cornerstone of cancer treatment; however, some individuals develop resistance to the drugs administered. GC has the highest rate of drug-resistant recurrence among all cancer types; this phenomenon considerably restricts the long-term prospects of patients with cancer, with 5-year survival rates dropping as low as 30% [205]. An increasing number of miRNAs have been found to be aberrantly expressed in drug-resistant GC tissues and are involved in the process of chemoresistance. These miRNAs function through complex mechanisms, including inactivation of apoptotic signaling pathways, loss of cell cycle checkpoint control, accelerated cell proliferation and autophagy flux, enhanced DNA damage repair, and drug transport and regulation. Furthermore, they activate CSCs and EMT [206,207,208] (Fig. 7). These correlations suggest that miRNA analysis will be a valuable tool for accurately assessing cellular sensitivity to chemotherapy and can be used to develop novel therapeutic approaches capable of overcoming resistance to GC chemotherapy [209] (Table 3).

Fig. 7
figure 7

miRNAs modulate GC chemoresistance through several mechanisms. miRNAs: MicroRNAs, GC: Gastric cancer

Table 3 miRNAs that play roles in GC chemoresistance

This phenomenon of chemoresistance is also linked to miRNAs in exosomes. For example, patients with GC who have elevated miR-500a-3p levels in their plasma exosomes are more likely to be resistant to cisplatin, which lowers their progression-free survival rate [269]. Additionally, exosomes allow miR-21 to be transported from macrophages to GC cells, which significantly lowers the sensitivity of GC cells to cisplatin treatment both in vitro and in vivo, partly through modulation of the PTEN/PI3K/AKT signaling pathway [144].

Clinically, GC tissues display markedly elevated levels of miR-223 expression. Moreover, a strong correlation between high expression levels of plasma exosomal miR-223 and doxorubicin resistance is observed in patients with GC [257]. For example, biologically active miR-769-5p spreads cisplatin resistance by integrating into exosomes and infiltrating sensitive cells. Furthermore, by targeting CASP9, miR-769-5p enables the ubiquitin–proteasome pathway to degrade p53, an apoptosis-associated protein, while suppressing the downstream caspase pathway [283]. Furthermore, by modulating the high mobility group A2/mTOR/P-GP axis, exosome-secreted miR-107 dramatically increases the sensitivity of drug-resistant GC cells to chemotherapeutic drugs [284]. Finally, in paclitaxel-resistant GC cells, exosomal administration of miR-155-5p promotes chemoresistant phenotypes and EMT, which may be mediated by suppression of TP53INP1 and GATA3 [244] (Fig. 8). Overall, these findings imply that exosomal-derived miRNAs are essential for the development of medication resistance.

Fig. 8
figure 8

Exosome-derived miRNAs regulate TME and participate in the development of gastric cancer. miRNAs: MicroRNAs, TME: Tumor microenvironment

Conclusions

Despite treatment efforts, GC remains one of the deadliest tumors. Over the past years, growing research has indicated the significant role of the TME in the development, advancement, invasion, and metastasis of GC. Recent studies have shown a strong correlation between GC and miRNA dysregulation, which has a significant impact on TME-related activities and provides new insights into the relationship between immune cells, mesenchymal stromal cells, malignant cells, and non-cellular components of the TME, promoting tumor proliferation, angiogenesis, and metastasis.

Particularly, malignant and drug-resistant tumor cells secrete exosomes containing specific miRNAs. Therefore, exosomes are crucial for material exchange, energy flow, and signaling between the different cellular components of the TME. An in-depth study of the effect of miRNAs on TME is of great significance in furthering our understanding of the biology of GC. Based on the role of miRNAs in TME, the development of miRNAs as synergistic tumor immunotherapeutics is of great significance to improve the efficacy of monotherapy and reduce tumor survival.

Notably, several challenges remain to be addressed before these studies can be translated into clinical applications. Firstly, due to the complexity of the TME, the exact mechanisms of different miRNAs in different cell types in the TME remain largely unknown [285]. To select the optimal targets, a deeper understanding of the role of each specific miRNA in all immune cell subpopulations and their complete regulatory networks is essential. Additionally, given that naked miRNAs have a short half-life in vivo and are easily degraded, there is an urgent need to identify a safe, effective, and targeted vector to protect the miRNAs and ensure their delivery to the intended sites [286].

In conclusion, this review describes the communication mechanisms of miRNAs between the TME and GC tumor cells. Dysregulated miRNAs are found in both non-tumor and tumor cells within the TME, emphasizing the key role played by the TME and miRNAs in the development and metastasis of cancer. While their exact mechanism of action is still being investigated, several miRNAs have emerged as potential therapeutic targets and GC biomarkers. Exploring and studying the regulatory effects of naturally derived drugs on the TME at the miRNA level holds promise, especially considering the polygenic targeting of miRNAs and the anticancer effects of natural drugs on various types of mesenchymal stromal cells within the TME.

Availability of data and materials

Not applicable.

Data availability

No datasets were generated or analysed during the current study.

Abbreviations

GC:

Gastric cancer

TME:

Tumor microenvironment

CAFs:

Cancer-associated fibroblasts

miRNAs:

MicroRNAs

ECM:

Extracellular matrix

DC:

Dendritic cells

Tregs:

Regulatory T cells

MDSCs:

Myeloid-derived suppressor cells

EVs:

Extracellular vesicles

VEGF:

Vascular endothelial growth factor

IL:

Interleukin

JAK2-STAT3:

Janus kinase 2

MAPK/ERK:

Mitogen-activated protein kinase/extracellular signal-regulated kinases

EMT:

Epithelial-mesenchymal transition

PI3K/AKT/mTOR:

Phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin

FOXM1:

Forkhead box M1

PD-L1:

Programmed death ligand 1

HIF-1α:

Hypoxia-inducible factor 1-alpha

TAMs:

Tumor-associated macrophages

PTEN:

Phosphatase and tensin homolog

Th1:

T helper 1 cells

Th17:

T helper 17 cells

CTLs:

Cytotoxic T lymphocytes

NKs:

Natural killer cells

ROS:

Reactive oxygen species

EBV:

Epstein–Barr virus

ZEB1:

Zinc finger E-box-binding homeobox 1

TNF:

Tumor necrosis factor

CSCs:

Cancer stem cells

References

  1. Guan WL, He Y, Xu RH. Gastric cancer treatment: recent progress and future perspectives. J Hematol Oncol. 2023;16:57.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. Ferlay J, Colombet M, Soerjomataram I, Mathers C, Parkin DM, Piñeros M, Znaor A, Bray F. Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int J Cancer. 2019;144:1941–53.

    Article  PubMed  CAS  Google Scholar 

  3. Wei J, Yang Y, Wang G, Liu M. Current landscape and future directions of bispecific antibodies in cancer immunotherapy. Front Immunol. 2022;13:1035276.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Li C, Teixeira AF, Zhu HJ, Ten Dijke P. Cancer associated-fibroblast-derived exosomes in cancer progression. Mol Cancer. 2021;20:154.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Yang E, Wang X, Gong Z, Yu M, Wu H, Zhang D. Exosome-mediated metabolic reprogramming: the emerging role in tumor microenvironment remodeling and its influence on cancer progression. Signal Transduct Target Ther. 2020;5:242.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Zhang C, Wei S, Dai S, Li X, Wang H, Zhang H, Sun G, Shan B, Zhao L. The NR_109/FUBP1/c-Myc axis regulates TAM polarization and remodels the tumor microenvironment to promote cancer development. J Immunother Cancer. 2023;11:e006230.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Boyd LNC, Andini KD, Peters GJ, Kazemier G, Giovannetti E. Heterogeneity and plasticity of cancer-associated fibroblasts in the pancreatic tumor microenvironment. Semin Cancer Biol. 2022;82:184–96.

    Article  PubMed  CAS  Google Scholar 

  8. Rawat M, Kadian K, Gupta Y, Kumar A, Chain PSG, Kovbasnjuk O, Kumar S, Parasher G. MicroRNA in Pancreatic Cancer: From Biology to Therapeutic Potential. Genes (Basel). 2019;10:752.

    Article  PubMed  CAS  Google Scholar 

  9. Berindan-Neagoe I, Monroig Pdel C, Pasculli B, Calin GA. MicroRNAome genome: a treasure for cancer diagnosis and therapy. CA Cancer J Clin. 2014;64:311–36.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Holjencin C, Jakymiw A. MicroRNAs and their big therapeutic impacts: delivery strategies for cancer intervention. Cells. 2022;11(15):2332.

  11. Xu X, Tao Y, Shan L, Chen R, Jiang H, Qian Z, Cai F, Ma L, Yu Y. The Role of MicroRNAs in Hepatocellular Carcinoma. J Cancer. 2018;9:3557–69.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Hussen BM, Abdullah SR, Rasul MF, Jawhar ZH, Faraj GSH, Kiani A, Taheri M. MiRNA-93: a novel signature in human disorders and drug resistance. Cell Commun Signal. 2023;21:79.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Chakrabortty A, Patton DJ, Smith BF, Agarwal P. miRNAs: Potential as biomarkers and therapeutic targets for cancer. Genes (Basel). 2023;14(7):1375.

  14. Xuan J, Liu Y, Zeng X, Wang H. Sequence requirements for miR-424–5p regulating and function in cancers. Int J Mol Sci. 2022;23(7):4037.

  15. Rhim J, Baek W, Seo Y, Kim JH. From molecular mechanisms to therapeutics: understanding MicroRNA-21 in cancer. Cells. 2022;11(18):2791.

  16. Zhang C, Sun C, Zhao Y, Wang Q, Guo J, Ye B, Yu G. Overview of MicroRNAs as diagnostic and prognostic biomarkers for high-incidence cancers in 2021. Int J Mol Sci. 2022;23(19):11389.

  17. Fanini F, Fabbri M. Cancer-derived exosomic microRNAs shape the immune system within the tumor microenvironment: State of the art. Semin Cell Dev Biol. 2017;67:23–8.

    Article  PubMed  CAS  Google Scholar 

  18. Marzagalli M, Ebelt ND, Manuel ER. Unraveling the crosstalk between melanoma and immune cells in the tumor microenvironment. Semin Cancer Biol. 2019;59:236–50.

    Article  PubMed  CAS  Google Scholar 

  19. Yi M, Xu L, Jiao Y, Luo S, Li A, Wu K. The role of cancer-derived microRNAs in cancer immune escape. J Hematol Oncol. 2020;13:25.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Jia Z, Jia J, Yao L, Li Z. Crosstalk of Exosomal Non-Coding RNAs in The Tumor Microenvironment: Novel Frontiers. Front Immunol. 2022;13: 900155.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Lim LP, Lau NC, Garrett-Engele P, Grimson A, Schelter JM, Castle J, Bartel DP, Linsley PS, Johnson JM. Microarray analysis shows that some microRNAs downregulate large numbers of target mRNAs. Nature. 2005;433:769–73.

    Article  PubMed  CAS  Google Scholar 

  22. Link A, Kupcinskas J. MicroRNAs as non-invasive diagnostic biomarkers for gastric cancer: Current insights and future perspectives. World J Gastroenterol. 2018;24:3313–29.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Chadda KR, Blakey EE, Coleman N, Murray MJ. The clinical utility of dysregulated microRNA expression in paediatric solid tumours. Eur J Cancer. 2022;176:133–54.

    Article  PubMed  CAS  Google Scholar 

  24. Azari H, Nazari E, Mohit R, Asadnia A, Maftooh M, Nassiri M, Hassanian SM, Ghayour-Mobarhan M, Shahidsales S, Khazaei M, et al. Machine learning algorithms reveal potential miRNAs biomarkers in gastric cancer. Sci Rep. 2023;13:6147.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Zhang C, Zhang CD, Liang Y, Wu KZ, Pei JP, Dai DQ. The comprehensive upstream transcription and downstream targeting regulation network of miRNAs reveal potential diagnostic roles in gastric cancer. Life Sci. 2020;253: 117741.

    Article  PubMed  CAS  Google Scholar 

  26. Liang Y, Liu Y, Zhang Q, Zhang H, Du J. Tumor-derived extracellular vesicles containing microRNA-1290 promote immune escape of cancer cells through the Grhl2/ZEB1/PD-L1 axis in gastric cancer. Transl Res. 2021;231:102–12.

    Article  PubMed  CAS  Google Scholar 

  27. Zhu F, Wu Q, Ni Z, Lei C, Li T, Shi Y. miR-19a/b and MeCP2 repress reciprocally to regulate multidrug resistance in gastric cancer cells. Int J Mol Med. 2018;42:228–36.

    PubMed  PubMed Central  CAS  Google Scholar 

  28. Mirzaei S, Zarrabi A, Hashemi F, Zabolian A, Saleki H, Ranjbar A, Seyed Saleh SH, Bagherian M, Sharifzadeh SO, Hushmandi K, et al. Regulation of Nuclear Factor-KappaB (NF-κB) signaling pathway by non-coding RNAs in cancer: Inhibiting or promoting carcinogenesis? Cancer Lett. 2021;509:63–80.

    Article  PubMed  CAS  Google Scholar 

  29. He S, Song W, Cui S, Li J, Jiang Y, Chen X, Peng L. Modulation of miR-146b by N6-methyladenosine modification remodels tumor-associated macrophages and enhances anti-PD-1 therapy in colorectal cancer. Cell Oncol (Dordr). 2023;46:1731–46.

    Article  PubMed  CAS  Google Scholar 

  30. Wu J, Li G, Wang Z, Yao Y, Chen R, Pu X, Wang J. Circulating MicroRNA-21 Is a Potential Diagnostic Biomarker in Gastric Cancer. Dis Markers. 2015;2015: 435656.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Emami SS, Nekouian R, Akbari A, Faraji A, Abbasi V, Agah S. Evaluation of circulating miR-21 and miR-222 as diagnostic biomarkers for gastric cancer. J Cancer Res Ther. 2019;15:115–9.

    Article  PubMed  CAS  Google Scholar 

  32. Tang Y, Liu X, Su B, Zhang Z, Zeng X, Lei Y, Shan J, Wu Y, Tang H, Su Q. microRNA-22 acts as a metastasis suppressor by targeting metadherin in gastric cancer. Mol Med Rep. 2015;11:454–60.

    Article  PubMed  CAS  Google Scholar 

  33. Chen TH, Chiu CT, Lee C, Chu YY, Cheng HT, Hsu JT, Wu RC, Yeh TS, Lin KH. Circulating microRNA-22-3p Predicts the Malignant Progression of Precancerous Gastric Lesions from Intestinal Metaplasia to Early Adenocarcinoma. Dig Dis Sci. 2018;63:2301–8.

    Article  PubMed  CAS  Google Scholar 

  34. Valladares-Ayerbes M, Reboredo M, Medina-Villaamil V, Iglesias-Díaz P, Lorenzo-Patiño MJ, Haz M, Santamarina I, Blanco M, Fernández-Tajes J, Quindós M, et al. Circulating miR-200c as a diagnostic and prognostic biomarker for gastric cancer. J Transl Med. 2012;10:186.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Xiao F, Cheng Z, Wang P, Gong B, Huang H, Xing Y, Liu F. MicroRNA-28-5p inhibits the migration and invasion of gastric cancer cells by suppressing AKT phosphorylation. Oncol Lett. 2018;15:9777–85.

    PubMed  PubMed Central  Google Scholar 

  36. Han TS, Hur K, Xu G, Choi B, Okugawa Y, Toiyama Y, Oshima H, Oshima M, Lee HJ, Kim VN, et al. MicroRNA-29c mediates initiation of gastric carcinogenesis by directly targeting ITGB1. Gut. 2015;64:203–14.

    Article  PubMed  CAS  Google Scholar 

  37. Wang N, Wang L, Yang Y, Gong L, Xiao B, Liu X. A serum exosomal microRNA panel as a potential biomarker test for gastric cancer. Biochem Biophys Res Commun. 2017;493:1322–8.

    Article  PubMed  CAS  Google Scholar 

  38. Cui L, Zhang X, Ye G, Zheng T, Song H, Deng H, Xiao B, Xia T, Yu X, Le Y, Guo J. Gastric juice MicroRNAs as potential biomarkers for the screening of gastric cancer. Cancer. 2013;119:1618–26.

    Article  PubMed  CAS  Google Scholar 

  39. Dong X, Liu Y. Expression and significance of miR-24 and miR-101 in patients with advanced gastric cancer. Oncol Lett. 2018;16:5769–74.

    PubMed  PubMed Central  CAS  Google Scholar 

  40. Liu F, Hu H, Zhao J, Zhang Z, Ai X, Tang L, Xie L. miR-124-3p acts as a potential marker and suppresses tumor growth in gastric cancer. Biomed Rep. 2018;9:147–55.

    PubMed  PubMed Central  CAS  Google Scholar 

  41. Yu X, Luo L, Wu Y, Yu X, Liu Y, Yu X, Zhao X, Zhang X, Cui L, Ye G, et al. Gastric juice miR-129 as a potential biomarker for screening gastric cancer. Med Oncol. 2013;30:365.

    Article  PubMed  Google Scholar 

  42. Shao J, Fang PH, He B, Guo LL, Shi MY, Zhu Y, Bo P, Zhen-Wen ZW. Downregulated MicroRNA-133a in Gastric Juice as a Clinicopathological Biomarker for Gastric Cancer Screening. Asian Pac J Cancer Prev. 2016;17:2719–22.

    PubMed  Google Scholar 

  43. Cha Y, He Y, Ouyang K, Xiong H, Li J, Yuan X. MicroRNA-140-5p suppresses cell proliferation and invasion in gastric cancer by targeting WNT1 in the WNT/β-catenin signaling pathway. Oncol Lett. 2018;16:6369–76.

    PubMed  PubMed Central  CAS  Google Scholar 

  44. Li Z, Guo Q, Lu Y, Tian T. Increased expression of miR-181d is associated with poor prognosis and tumor progression of gastric cancer. Cancer Biomark. 2019;26:353–60.

    Article  PubMed  Google Scholar 

  45. Chen W, Cui Y, Wang J, Yuan Y, Sun X, Zhang L, Shen S, Cheng J. Effects of downregulated expression of microRNA-187 in gastric cancer. Exp Ther Med. 2018;16:1061–70.

    PubMed  PubMed Central  Google Scholar 

  46. Tsai MM, Wang CS, Tsai CY, Huang CG, Lee KF, Huang HW, Lin YH, Chi HC, Kuo LM, Lu PH, Lin KH. Circulating microRNA-196a/b are novel biomarkers associated with metastatic gastric cancer. Eur J Cancer. 2016;64:137–48.

    Article  PubMed  CAS  Google Scholar 

  47. Chen TH, Lee C, Chiu CT, Chu YY, Cheng HT, Hsu JT, Tsou YK, Wu RC, Chen TC, Chang NC, et al. Circulating microRNA-196a is an early gastric cancer biomarker. Oncotarget. 2018;9:10317–23.

    Article  PubMed  Google Scholar 

  48. Imaoka H, Toiyama Y, Okigami M, Yasuda H, Saigusa S, Ohi M, Tanaka K, Inoue Y, Mohri Y, Kusunoki M. Circulating microRNA-203 predicts metastases, early recurrence, and poor prognosis in human gastric cancer. Gastric Cancer. 2016;19:744–53.

    Article  PubMed  CAS  Google Scholar 

  49. Shao JP, Su F, Zhang SP, Chen HK, Li ZJ, Xing GQ, Liu HJ, Li YY. miR-212 as potential biomarker suppresses the proliferation of gastric cancer via targeting SOX4. J Clin Lab Anal. 2020;34: e23511.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Tang L, Hu H, He Y, McLeod HL, Xiao D, Chen P, Shen L, Zeng S, Yin X, Ge J, et al. The relationship between miR-302b and EphA2 and their clinical significance in gastric cancer. J Cancer. 2018;9:3109–16.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Zhang J, Wang C, Yan S, Yang Y, Zhang X, Guo W. miR-345 inhibits migration and stem-like cell phenotype in gastric cancer via inactivation of Rac1 by targeting EPS8. Acta Biochim Biophys Sin (Shanghai). 2020;52:259–67.

    Article  PubMed  CAS  Google Scholar 

  52. Xu M, Qin S, Cao F, Ding S, Li M. MicroRNA-379 inhibits metastasis and epithelial-mesenchymal transition via targeting FAK/AKT signaling in gastric cancer. Int J Oncol. 2017;51:867–76.

    Article  PubMed  CAS  Google Scholar 

  53. Ge X, Liu X, Lin F, Li P, Liu K, Geng R, Dai C, Lin Y, Tang W, Wu Z, et al. MicroRNA-421 regulated by HIF-1α promotes metastasis, inhibits apoptosis, and induces cisplatin resistance by targeting E-cadherin and caspase-3 in gastric cancer. Oncotarget. 2016;7:24466–82.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Chen J, Wu L, Sun Y, Yin Q, Chen X, Liang S, Meng Q, Long H, Li F, Luo C, Xiao X. Mir-421 in plasma as a potential diagnostic biomarker for precancerous gastric lesions and early gastric cancer. PeerJ. 2019;7: e7002.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Zhang X, Cui L, Ye G, Zheng T, Song H, Xia T, Yu X, Xiao B, Le Y, Guo J. Gastric juice microRNA-421 is a new biomarker for screening gastric cancer. Tumour Biol. 2012;33:2349–55.

    Article  PubMed  CAS  Google Scholar 

  56. Li Y, Liu Y, Yao J, Li R, Fan X. Downregulation of miR-484 is associated with poor prognosis and tumor progression of gastric cancer. Diagn Pathol. 2020;15:25.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Su H, Ren F, Jiang H, Chen Y, Fan X. Upregulation of microRNA-520a-3p inhibits the proliferation, migration and invasion via spindle and kinetochore associated 2 in gastric cancer. Oncol Lett. 2019;18:3323–30.

    PubMed  PubMed Central  CAS  Google Scholar 

  58. Cui HB, Ge HE, Wang YS, Bai XY. MiR-208a enhances cell proliferation and invasion of gastric cancer by targeting SFRP1 and negatively regulating MEG3. Int J Biochem Cell Biol. 2018;102:31–9.

    Article  PubMed  CAS  Google Scholar 

  59. Feng X, Zhu M, Liao B, Tian T, Li M, Wang Z, Chen G. Upregulation of miR-552 Predicts Unfavorable Prognosis of Gastric Cancer and Promotes the Proliferation, Migration, and Invasion of Gastric Cancer Cells. Oncol Res Treat. 2020;43:103–11.

    Article  PubMed  CAS  Google Scholar 

  60. Hu L, Wu H, Wan X, Liu L, He Y, Zhu L, Liu S, Yao H, Zhu Z. MicroRNA-585 suppresses tumor proliferation and migration in gastric cancer by directly targeting MAPK1. Biochem Biophys Res Commun. 2018;499:52–8.

    Article  PubMed  CAS  Google Scholar 

  61. Min C, Zhang A, Qin J. Increased expression of miR-601 is associated with poor prognosis and tumor progression of gastric cancer. Diagn Pathol. 2019;14:107.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Zheng H, Zhang F, Lin X, Huang C, Zhang Y, Li Y, Lin J, Chen W, Lin X. MicroRNA-1225-5p inhibits proliferation and metastasis of gastric carcinoma through repressing insulin receptor substrate-1 and activation of β-catenin signaling. Oncotarget. 2016;7:4647–63.

    Article  PubMed  Google Scholar 

  63. An JX, Ma ZS, Ma MH, Shao S, Cao FL, Dai DQ. MiR-1236-3p serves as a new diagnostic and prognostic biomarker for gastric cancer. Cancer Biomark. 2019;25:127–32.

    Article  PubMed  CAS  Google Scholar 

  64. Liu S, Tian Y, Zhu C, Yang X, Sun Q. High miR-718 Suppresses Phosphatase and Tensin Homolog (PTEN) Expression and Correlates to Unfavorable Prognosis in Gastric Cancer. Med Sci Monit. 2018;24:5840–50.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  65. Abe S, Matsuzaki J, Sudo K, Oda I, Katai H, Kato K, Takizawa S, Sakamoto H, Takeshita F, Niida S, et al. A novel combination of serum microRNAs for the detection of early gastric cancer. Gastric Cancer. 2021;24:835–43.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  66. Choi JM, Kim SG, Yang HJ, Lim JH, Cho NY, Kim WH, Kim JS, Jung HC. Helicobacter pylori Eradication Can Reverse the Methylation-Associated Regulation of miR-200a/b in Gastric Carcinogenesis. Gut Liver. 2020;14:571–80.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Li X, Pan K, Vieth M, Gerhard M, Li W, Mejías-Luque R. JAK-STAT1 signaling pathway is an early response to helicobacter pylori infection and contributes to immune escape and gastric carcinogenesis. Int J Mol Sci. 2022;23(8):4147.

  68. Yang F, Xu Y, Liu C, Ma C, Zou S, Xu X, Jia J, Liu Z. NF-κB/miR-223-3p/ARID1A axis is involved in Helicobacter pylori CagA-induced gastric carcinogenesis and progression. Cell Death Dis. 2018;9:12.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Wang J, Deng R, Chen S, Deng S, Hu Q, Xu B, Li J, He Z, Peng M, Lei S, et al. Helicobacter pylori CagA promotes immune evasion of gastric cancer by upregulating PD-L1 level in exosomes. iScience. 2023;26:108414.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  70. Shi Y, Yang Z, Zhang T, Shen L, Li Y, Ding S. SIRT1-targeted miR-543 autophagy inhibition and epithelial-mesenchymal transition promotion in Helicobacter pylori CagA-associated gastric cancer. Cell Death Dis. 2019;10:625.

    Article  PubMed  PubMed Central  Google Scholar 

  71. Koh V, Chakrabarti J, Torvund M, Steele N, Hawkins JA, Ito Y, Wang J, Helmrath MA, Merchant JL, Ahmed SA, et al. Hedgehog transcriptional effector GLI mediates mTOR-Induced PD-L1 expression in gastric cancer organoids. Cancer Lett. 2021;518:59–71.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  72. Shao W, Yang Z, Fu Y, Zheng L, Liu F, Chai L, Jia J. The Pyroptosis-Related Signature Predicts Prognosis and Indicates Immune Microenvironment Infiltration in Gastric Cancer. Front Cell Dev Biol. 2021;9: 676485.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Matsushima K, Isomoto H, Inoue N, Nakayama T, Hayashi T, Nakayama M, Nakao K, Hirayama T, Kohno S. MicroRNA signatures in Helicobacter pylori-infected gastric mucosa. Int J Cancer. 2011;128:361–70.

    Article  PubMed  CAS  Google Scholar 

  74. Oertli M, Engler DB, Kohler E, Koch M, Meyer TF, Müller A. MicroRNA-155 is essential for the T cell-mediated control of Helicobacter pylori infection and for the induction of chronic Gastritis and Colitis. J Immunol. 2011;187:3578–86.

    Article  PubMed  CAS  Google Scholar 

  75. Tsai CC, Chen TY, Tsai KJ, Lin MW, Hsu CY, Wu DC, Tsai EM, Hsieh TH. NF-κB/miR-18a-3p and miR-4286/BZRAP1 axis may mediate carcinogenesis in Helicobacter pylori-Associated gastric cancer. Biomed Pharmacother. 2020;132: 110869.

    Article  PubMed  CAS  Google Scholar 

  76. Li T, Guo H, Zhao X, Jin J, Zhang L, Li H, Lu Y, Nie Y, Wu K, Shi Y, Fan D. Gastric Cancer Cell Proliferation and Survival Is Enabled by a Cyclophilin B/STAT3/miR-520d-5p Signaling Feedback Loop. Cancer Res. 2017;77:1227–40.

    Article  PubMed  CAS  Google Scholar 

  77. Fassi Fehri L, Koch M, Belogolova E, Khalil H, Bolz C, Kalali B, Mollenkopf HJ, Beigier-Bompadre M, Karlas A, Schneider T, et al. Helicobacter pylori induces miR-155 in T cells in a cAMP-Foxp3-dependent manner. PLoS ONE. 2010;5: e9500.

    Article  PubMed  PubMed Central  Google Scholar 

  78. Huang L, Wang ZY, Pan DD. Penicillin-binding protein 1A mutation-positive Helicobacter pylori promotes epithelial-mesenchymal transition in gastric cancer via the suppression of microRNA-134. Int J Oncol. 2019;54:916–28.

    PubMed  CAS  Google Scholar 

  79. Bai Y, Xie T, Wang Z, Tong S, Zhao X, Zhao F, Cai J, Wei X, Peng Z, Shen L. Efficacy and predictive biomarkers of immunotherapy in Epstein-Barr virus-associated gastric cancer. J Immunother Cancer. 2022;10(3):e004080.

  80. Bauer M, Jasinski-Bergner S, Mandelboim O, Wickenhauser C, Seliger B. Epstein-Barr virus-associated malignancies and immune escape: the role of the tumor microenvironment and tumor cell evasion strategies. Cancers (Basel). 2021;13(20):5189.

  81. Pandya D, Mariani M, He S, Andreoli M, Spennato M, Dowell-Martino C, Fiedler P, Ferlini C. Epstein-Barr Virus MicroRNA Expression Increases Aggressiveness of Solid Malignancies. PLoS ONE. 2015;10: e0136058.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Saju P, Murata-Kamiya N, Hayashi T, Senda Y, Nagase L, Noda S, Matsusaka K, Funata S, Kunita A, Urabe M, et al. Host SHP1 phosphatase antagonizes Helicobacter pylori CagA and can be downregulated by Epstein-Barr virus. Nat Microbiol. 2016;1:16026.

    Article  PubMed  CAS  Google Scholar 

  83. Shinozaki A, Sakatani T, Ushiku T, Hino R, Isogai M, Ishikawa S, Uozaki H, Takada K, Fukayama M. Downregulation of microRNA-200 in EBV-associated gastric carcinoma. Cancer Res. 2010;70:4719–27.

    Article  PubMed  CAS  Google Scholar 

  84. Kim SM, Hur DY, Hong SW, Kim JH. EBV-encoded EBNA1 regulates cell viability by modulating miR34a-NOX2-ROS signaling in gastric cancer cells. Biochem Biophys Res Commun. 2017;494:550–5.

    Article  PubMed  CAS  Google Scholar 

  85. Choi H, Lee H, Kim SR, Gho YS, Lee SK. Epstein-Barr virus-encoded microRNA BART15-3p promotes cell apoptosis partially by targeting BRUCE. J Virol. 2013;87:8135–44.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  86. Wang J, Ge J, Wang Y, Xiong F, Guo J, Jiang X, Zhang L, Deng X, Gong Z, Zhang S, et al. EBV miRNAs BART11 and BART17-3p promote immune escape through the enhancer-mediated transcription of PD-L1. Nat Commun. 2022;13:866.

    Article  PubMed  PubMed Central  Google Scholar 

  87. Albanese M, Tagawa T, Bouvet M, Maliqi L, Lutter D, Hoser J, Hastreiter M, Hayes M, Sugden B, Martin L, et al. Epstein-Barr virus microRNAs reduce immune surveillance by virus-specific CD8+ T cells. Proc Natl Acad Sci U S A. 2016;113:E6467-e6475.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  88. Iizasa H, Wulff BE, Alla NR, Maragkakis M, Megraw M, Hatzigeorgiou A, Iwakiri D, Takada K, Wiedmer A, Showe L, et al. Editing of Epstein-Barr virus-encoded BART6 microRNAs controls their dicer targeting and consequently affects viral latency. J Biol Chem. 2010;285:33358–70.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  89. Shafqat A, Omer MH, Ahmed EN, Mushtaq A, Ijaz E, Ahmed Z, Alkattan K, Yaqinuddin A. Reprogramming the immunosuppressive tumor microenvironment: exploiting angiogenesis and thrombosis to enhance immunotherapy. Front Immunol. 2023;14:1200941.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  90. Liu ZL, Chen HH, Zheng LL, Sun LP, Shi L. Angiogenic signaling pathways and anti-angiogenic therapy for cancer. Signal Transduct Target Ther. 2023;8:198.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  91. Rahma OE, Hodi FS. The intersection between tumor angiogenesis and immune suppression. Clin Cancer Res. 2019;25:5449–57.

  92. Kim S, Bae WJ, Ahn JM, Heo JH, Kim KM, Choi KW, Sung CO, Lee D. MicroRNA signatures associated with lymph node metastasis in intramucosal gastric cancer. Mod Pathol. 2021;34:672–83.

    Article  PubMed  CAS  Google Scholar 

  93. Zhang H, Qu Y, Duan J, Deng T, Liu R, Zhang L, Bai M, Li J, Zhou L, Ning T, et al. Integrated analysis of the miRNA, gene and pathway regulatory network in gastric cancer. Oncol Rep. 2016;35:1135–46.

    Article  PubMed  CAS  Google Scholar 

  94. Wu ZH, Lin C, Liu CC, Jiang WW, Huang MZ, Liu X, Guo WJ. MiR-616-3p promotes angiogenesis and EMT in gastric cancer via the PTEN/AKT/mTOR pathway. Biochem Biophys Res Commun. 2018;501:1068–73.

    Article  PubMed  CAS  Google Scholar 

  95. Xie M, Dart DA, Guo T, Xing XF, Cheng XJ, Du H, Jiang WG, Wen XZ, Ji JF. MicroRNA-1 acts as a tumor suppressor microRNA by inhibiting angiogenesis-related growth factors in human gastric cancer. Gastric Cancer. 2018;21:41–54.

    Article  PubMed  Google Scholar 

  96. Gutiérrez J, Droppelmann CA, Salsoso R, Westermeier F, Toledo F, Salomon C, Sanhueza C, Pardo F, Leiva A, Sobrevia L. A Hypothesis for the Role of RECK in Angiogenesis. Curr Vasc Pharmacol. 2016;14:106–15.

    Article  PubMed  Google Scholar 

  97. Anand S, Majeti BK, Acevedo LM, Murphy EA, Mukthavaram R, Scheppke L, Huang M, Shields DJ, Lindquist JN, Lapinski PE, et al. MicroRNA-132-mediated loss of p120RasGAP activates the endothelium to facilitate pathological angiogenesis. Nat Med. 2010;16:909–14.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  98. Du J, Liang Y, Li J, Zhao JM, Lin XY. Gastric Cancer Cell-Derived Exosomal microRNA-23a Promotes Angiogenesis by Targeting PTEN. Front Oncol. 2020;10:326.

    Article  PubMed  PubMed Central  Google Scholar 

  99. Zhang R, Miao Z, Liu Y, Zhang X, Yang Q. A positive feedback loop between miR-574-3p and HIF-1α in promoting angiogenesis under hypoxia. Microvasc Res. 2023;150: 104589.

    Article  PubMed  CAS  Google Scholar 

  100. Bavelloni A, Ramazzotti G, Poli A, Piazzi M, Focaccia E, Blalock W, Faenza I. MiRNA-210: A Current Overview. Anticancer Res. 2017;37:6511–21.

    PubMed  CAS  Google Scholar 

  101. Wang L, Bo X, Zheng Q, Ge W, Liu Y, Li B. Paired box 8 suppresses tumor angiogenesis and metastasis in gastric cancer through repression of FOXM1 via induction of microRNA-612. J Exp Clin Cancer Res. 2018;37:159.

    Article  PubMed  PubMed Central  Google Scholar 

  102. Zhang S, Zhang R, Xu R, Shang J, He H, Yang Q. MicroRNA-574-5p in gastric cancer cells promotes angiogenesis by targeting protein tyrosine phosphatase non-receptor type 3 (PTPN3). Gene. 2020;733: 144383.

    Article  PubMed  CAS  Google Scholar 

  103. Yu B, Zhu N, Fan Z, Li J, Kang Y, Liu B. miR-29c inhibits metastasis of gastric cancer cells by targeting VEGFA. J Cancer. 2022;13:3566–74.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  104. Si Y, Zhang H, Ning T, Bai M, Wang Y, Yang H, Wang X, Li J, Ying G, Ba Y. miR-26a/b Inhibit Tumor Growth and Angiogenesis by Targeting the HGF-VEGF Axis in Gastric Carcinoma. Cell Physiol Biochem. 2017;42:1670–83.

    Article  PubMed  CAS  Google Scholar 

  105. Zhang J, Zhang J, Pang X, Chen Z, Zhang Z, Lei L, Xu H, Wen L, Zhu J, Jiang Y, et al. MiR-205-5p suppresses angiogenesis in gastric cancer by downregulating the expression of VEGFA and FGF1. Exp Cell Res. 2021;404: 112579.

    Article  PubMed  CAS  Google Scholar 

  106. Liu HT, Ma RR, Lv BB, Zhang H, Shi DB, Guo XY, Zhang GH, 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;122:1825–36.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  107. Zheng L, Pu J, Qi T, Qi M, Li D, Xiang X, Huang K, Tong Q. miRNA-145 targets v-ets erythroblastosis virus E26 oncogene homolog 1 to suppress the invasion, metastasis, and angiogenesis of gastric cancer cells. Mol Cancer Res. 2013;11:182–93.

    Article  PubMed  CAS  Google Scholar 

  108. Li Z, Liu Z, Dong S, Zhang J, Tan J, Wang Y, Ge C, Li R, Xue Y, Li M, et al. miR-506 Inhibits Epithelial-to-Mesenchymal Transition and Angiogenesis in Gastric Cancer. Am J Pathol. 2015;185:2412–20.

    Article  PubMed  CAS  Google Scholar 

  109. 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;6:1605–17.

    Article  PubMed  PubMed Central  Google Scholar 

  110. Mei B, Chen J, Yang N, Peng Y. The regulatory mechanism and biological significance of the Snail-miR590-VEGFR-NRP1 axis in the angiogenesis, growth and metastasis of gastric cancer. Cell Death Dis. 2020;11:241.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  111. Lin J, Liu Z, Liao S, Li E, Wu X, Zeng W. Elevated microRNA-7 inhibits proliferation and tumor angiogenesis and promotes apoptosis of gastric cancer cells via repression of Raf-1. Cell Cycle. 2020;19:2496–508.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  112. Zhang X, Dong J, He Y, Zhao M, Liu Z, Wang N, Jiang M, Zhang Z, Liu G, Liu H, et al. miR-218 inhibited tumor angiogenesis by targeting ROBO1 in gastric cancer. Gene. 2017;615:42–9.

    Article  PubMed  CAS  Google Scholar 

  113. Shichiri K, Oshi M, Ziazadeh D, Endo I, Takabe K. High miR-200c expression is associated with suppressed epithelial-mesenchymal transition, TGF-β signaling and better survival despite enhanced cell proliferation in gastric cancer patients. Am J Cancer Res. 2023;13:3027–40.

    PubMed  PubMed Central  CAS  Google Scholar 

  114. Tokumaru Y, Oshi M, Huyser MR, Yan L, Fukada M, Matsuhashi N, Futamura M, Akao Y, Yoshida K, Takabe K. Low expression of miR-29a is associated with aggressive biology and worse survival in gastric cancer. Sci Rep. 2021;11:14134.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  115. Cheng W, Liao Y, Xie Y, Wang Q, Li L, Chen Y, Zhao Y, Zhou J. Helicobacter pylori-induced fibroblast-derived Serpin E1 promotes gastric cancer growth and peritoneal dissemination through p38 MAPK/VEGFA-mediated angiogenesis. Cancer Cell Int. 2023;23:326.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  116. Al-Marzouki L, Stavrakos VS, Pal S, Giannias B, Bourdeau F, Rayes R, Bertos N, Najmeh S, Spicer JD, Cools-Lartigue J, et al. Soluble factors in malignant ascites promote the metastatic adhesion of gastric adenocarcinoma cells. Gastric Cancer. 2023;26:55–68.

    Article  PubMed  CAS  Google Scholar 

  117. Ohzawa H, Kumagai Y, Yamaguchi H, Miyato H, Sakuma Y, Horie H, Hosoya Y, Kawarai Lefor A, Sata N, Kitayama J. Exosomal microRNA in peritoneal fluid as a biomarker of peritoneal metastases from gastric cancer. Ann Gastroenterol Surg. 2020;4:84–93.

    Article  PubMed  Google Scholar 

  118. Yang F, Ning Z, Ma L, Liu W, Shao C, Shu Y, Shen H. Exosomal miRNAs and miRNA dysregulation in cancer-associated fibroblasts. Mol Cancer. 2017;16:148.

    Article  PubMed  PubMed Central  Google Scholar 

  119. Kennel KB, Bozlar M, De Valk AF, Greten FR. Cancer-Associated Fibroblasts in Inflammation and Antitumor Immunity. Clin Cancer Res. 2023;29:1009–16.

    Article  PubMed  CAS  Google Scholar 

  120. Neophytou CM, Panagi M, Stylianopoulos T, Papageorgis P. The role of tumor microenvironment in cancer metastasis: molecular mechanisms and therapeutic opportunities. Cancers (Basel). 2021;13(9):2053.

  121. Wang J, Guan X, Zhang Y, Ge S, Zhang L, Li H, Wang X, Liu R, Ning T, Deng T, et al. Exosomal miR-27a Derived from Gastric Cancer Cells Regulates the Transformation of Fibroblasts into Cancer-Associated Fibroblasts. Cell Physiol Biochem. 2018;49:869–83.

    Article  PubMed  CAS  Google Scholar 

  122. Kurashige J, Mima K, Sawada G, Takahashi Y, Eguchi H, Sugimachi K, Mori M, Yanagihara K, Yashiro M, Hirakawa K, et al. Epigenetic modulation and repression of miR-200b by cancer-associated fibroblasts contribute to cancer invasion and peritoneal dissemination in gastric cancer. Carcinogenesis. 2015;36:133–41.

    Article  PubMed  CAS  Google Scholar 

  123. Takahashi S, Takagane K, Itoh G, Kuriyama S, Umakoshi M, Goto A, Yanagihara K, Yashiro M, Iijima K, Tanaka M. CCDC85A is regulated by miR-224-3p and augments cancer cell resistance to endoplasmic reticulum stress. Front Oncol. 2023;13:1196546.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  124. Wang R, Sun Y, Yu W, Yan Y, Qiao M, Jiang R, Guan W, Wang L. Downregulation of miRNA-214 in cancer-associated fibroblasts contributes to migration and invasion of gastric cancer cells through targeting FGF9 and inducing EMT. J Exp Clin Cancer Res. 2019;38:20.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  125. Huang C, Liu J, He L, Wang F, Xiong B, Li Y, Yang X. The long noncoding RNA noncoding RNA activated by DNA damage (NORAD)-microRNA-496-Interleukin-33 axis affects carcinoma-associated fibroblasts-mediated gastric cancer development. Bioengineered. 2021;12:11738–55.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  126. Yang TS, Yang XH, Chen X, Wang XD, Hua J, Zhou DL, Zhou B, Song ZS. MicroRNA-106b in cancer-associated fibroblasts from gastric cancer promotes cell migration and invasion by targeting PTEN. FEBS Lett. 2014;588:2162–9.

    Article  PubMed  CAS  Google Scholar 

  127. Zhang H, Deng T, Liu R, Ning T, Yang H, Liu D, Zhang Q, Lin D, Ge S, Bai M, et al. CAF secreted miR-522 suppresses ferroptosis and promotes acquired chemo-resistance in gastric cancer. Mol Cancer. 2020;19:43.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  128. Zhou Y, Zhong JH, Gong FS, Xiao J. MiR-141-3p suppresses gastric cancer induced transition of normal fibroblast and BMSC to cancer-associated fibroblasts via targeting STAT4. Exp Mol Pathol. 2019;107:85–94.

    Article  PubMed  CAS  Google Scholar 

  129. Sammarco G, Varricchi G, Ferraro V, Ammendola M, De Fazio M, Altomare DF, Luposella M, Maltese L, Currò G, Marone G, et al. Mast cells, angiogenesis and lymphangiogenesis in human gastric cancer. Int J Mol Sci. 2019;20(9):2106.

  130. Oya Y, Hayakawa Y, Koike K. Tumor microenvironment in gastric cancers. Cancer Sci. 2020;111:2696–707.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  131. Bahmani L, Baghi M, Peymani M, Javeri A, Ghaedi K. MiR-141-3p and miR-200a-3p are involved in Th17 cell differentiation by negatively regulating RARB expression. Hum Cell. 2021;34:1375–87.

    Article  PubMed  CAS  Google Scholar 

  132. Li M, Yang Y, Xiong L, Jiang P, Wang J, Li C. Metabolism, metabolites, and macrophages in cancer. J Hematol Oncol. 2023;16:80.

    Article  PubMed  PubMed Central  Google Scholar 

  133. Daniel B, Nagy G, Horvath A, Czimmerer Z, Cuaranta-Monroy I, Poliska S, Hays TT, Sauer S, Francois-Deleuze J, Nagy L. The IL-4/STAT6/PPARγ signaling axis is driving the expansion of the RXR heterodimer cistrome, providing complex ligand responsiveness in macrophages. Nucleic Acids Res. 2018;46:4425–39.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  134. Sun X, Gao J, Meng X, Lu X, Zhang L, Chen R. Polarized Macrophages in Periodontitis: Characteristics, Function, and Molecular Signaling. Front Immunol. 2021;12: 763334.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  135. Christofides A, Strauss L, Yeo A, Cao C, Charest A, Boussiotis VA. The complex role of tumor-infiltrating macrophages. Nat Immunol. 2022;23:1148–56.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  136. Nascimento C, Ferreira F. Tumor microenvironment of human breast cancer, and feline mammary carcinoma as a potential study model. Biochim Biophys Acta Rev Cancer. 2021;1876: 188587.

    Article  PubMed  CAS  Google Scholar 

  137. Zheng P, Luo Q, Wang W, Li J, Wang T, Wang P, Chen L, Zhang P, Chen H, Liu Y, et al. Tumor-associated macrophages-derived exosomes promote the migration of gastric cancer cells by transfer of functional Apolipoprotein E. Cell Death Dis. 2018;9:434.

    Article  PubMed  PubMed Central  Google Scholar 

  138. Chen C, Liu JM, Luo YP. MicroRNAs in tumor immunity: functional regulation in tumor-associated macrophages. J Zhejiang Univ Sci B. 2020;21:12–28.

    Article  PubMed  PubMed Central  Google Scholar 

  139. Zhihua Y, Yulin T, Yibo W, Wei D, Yin C, Jiahao X, Runqiu J, Xuezhong X. Hypoxia decreases macrophage glycolysis and M1 percentage by targeting microRNA-30c and mTOR in human gastric cancer. Cancer Sci. 2019;110:2368–77.

    Article  PubMed  PubMed Central  Google Scholar 

  140. Xi J, Huang Q, Wang L, Ma X, Deng Q, Kumar M, Zhou Z, Li L, Zeng Z, Young KH, et al. miR-21 depletion in macrophages promotes tumoricidal polarization and enhances PD-1 immunotherapy. Oncogene. 2018;37:3151–65.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  141. Sahraei M, Chaube B, Liu Y, Sun J, Kaplan A, Price NL, Ding W, Oyaghire S, García-Milian R, Mehta S, et al. Suppressing miR-21 activity in tumor-associated macrophages promotes an antitumor immune response. J Clin Invest. 2019;129:5518–36.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  142. Yang X, Cai S, Shu Y, Deng X, Zhang Y, He N, Wan L, Chen X, Qu Y, Yu S. Exosomal miR-487a derived from m2 macrophage promotes the progression of gastric cancer. Cell Cycle. 2021;20:434–44.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  143. Cui HY, Rong JS, Chen J, Guo J, Zhu JQ, Ruan M, Zuo RR, Zhang SS, Qi JM, Zhang BH. Exosomal microRNA-588 from M2 polarized macrophages contributes to cisplatin resistance of gastric cancer cells. World J Gastroenterol. 2021;27:6079–92.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  144. Zheng P, Chen L, Yuan X, Luo Q, Liu Y, Xie G, Ma Y, Shen L. Exosomal transfer of tumor-associated macrophage-derived miR-21 confers cisplatin resistance in gastric cancer cells. J Exp Clin Cancer Res. 2017;36:53.

    Article  PubMed  PubMed Central  Google Scholar 

  145. Gwee YX, Chia DKA, So J, Ceelen W, Yong WP, Tan P, Ong CJ, Sundar R. Integration of Genomic Biology Into Therapeutic Strategies of Gastric Cancer Peritoneal Metastasis. J Clin Oncol. 2022;40:2830.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  146. Takahashi K, Kurashina K, Yamaguchi H, Kanamaru R, Ohzawa H, Miyato H, Saito S, Hosoya Y, Lefor AK, Sata N, Kitayama J. Altered intraperitoneal immune microenvironment in patients with peritoneal metastases from gastric cancer. Front Immunol. 2022;13: 969468.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  147. Yamaguchi T, Fushida S, Yamamoto Y, Tsukada T, Kinoshita J, Oyama K, Miyashita T, Tajima H, Ninomiya I, Munesue S, et al. Tumor-associated macrophages of the M2 phenotype contribute to progression in gastric cancer with peritoneal dissemination. Gastric Cancer. 2016;19:1052–65.

    Article  PubMed  CAS  Google Scholar 

  148. Eum HH, Kwon M, Ryu D, Jo A, Chung W, Kim N, Hong Y, Son DS, Kim ST, Lee J, et al. Tumor-promoting macrophages prevail in malignant ascites of advanced gastric cancer. Exp Mol Med. 2020;52:1976–88.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  149. Chen CW, Wang HC, Tsai IM, Chen IS, Chen CJ, Hou YC, Shan YS. CD204-positive M2-like tumor-associated macrophages increase migration of gastric cancer cells by upregulating miR-210 to reduce NTN4 expression. Cancer Immunol Immunother. 2024;73:1.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  150. Ruterbusch M, Pruner KB, Shehata L, Pepper M. In Vivo CD4(+) T Cell Differentiation and Function: Revisiting the Th1/Th2 Paradigm. Annu Rev Immunol. 2020;38:705–25.

    Article  PubMed  CAS  Google Scholar 

  151. Cho S, Wu CJ, Yasuda T, Cruz LO, Khan AA, Lin LL, Nguyen DT, Miller M, Lee HM, Kuo ML, et al. miR-232724 clusters control effector T cell differentiation and function. J Exp Med. 2016;213:235–49.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  152. Song J, Lin Z, Liu Q, Huang S, Han L, Fang Y, Zhong P, Dou R, Xiang Z, Zheng J, et al. MiR-192-5p/RB1/NF-κBp65 signaling axis promotes IL-10 secretion during gastric cancer EMT to induce Treg cell differentiation in the tumour microenvironment. Clin Transl Med. 2022;12: e992.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  153. Yin Y, Cai X, Chen X, Liang H, Zhang Y, Li J, Wang Z, Chen X, Zhang W, Yokoyama S, et al. Tumor-secreted miR-214 induces regulatory T cells: a major link between immune evasion and tumor growth. Cell Res. 2014;24:1164–80.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  154. Zhuansun Y, Du Y, Huang F, Lin L, Chen R, Jiang S, Li J. MSCs exosomal miR-1470 promotes the differentiation of CD4(+)CD25(+)FOXP3(+) Tregs in asthmatic patients by inducing the expression of P27KIP1. Int Immunopharmacol. 2019;77: 105981.

    Article  PubMed  CAS  Google Scholar 

  155. Liu F, Bu Z, Zhao F, Xiao D. Increased T-helper 17 cell differentiation mediated by exosome-mediated microRNA-451 redistribution in gastric cancer infiltrated T cells. Cancer Sci. 2018;109:65–73.

    Article  PubMed  CAS  Google Scholar 

  156. Ping W, Senyan H, Li G, Yan C, Long L. Increased Lactate in Gastric Cancer Tumor-Infiltrating Lymphocytes Is Related to Impaired T Cell Function Due to miR-34a Deregulated Lactate Dehydrogenase A. Cell Physiol Biochem. 2018;49:828–36.

    Article  PubMed  CAS  Google Scholar 

  157. Safarzadeh E, Orangi M, Mohammadi H, Babaie F, Baradaran B. Myeloid-derived suppressor cells: Important contributors to tumor progression and metastasis. J Cell Physiol. 2018;233:3024–36.

    Article  PubMed  CAS  Google Scholar 

  158. Xiang X, Wu Y, Li H, Li C, Yan L, Li Q. Plasmacytoid Dendritic Cell-Derived Type I Interferon Is Involved in Helicobacter pylori Infection-Induced Differentiation of Schlafen 4-Expressing Myeloid-Derived Suppressor Cells. Infect Immun. 2021;89: e0040721.

    Article  PubMed  Google Scholar 

  159. Ding L, Li Q, Chakrabarti J, Munoz A, Faure-Kumar E, Ocadiz-Ruiz R, Razumilava N, Zhang G, Hayes MH, Sontz RA, et al. MiR130b from Schlafen4(+) MDSCs stimulates epithelial proliferation and correlates with preneoplastic changes prior to gastric cancer. Gut. 2020;69:1750–61.

    Article  PubMed  CAS  Google Scholar 

  160. Ren W, Zhang X, Li W, Feng Q, Feng H, Tong Y, Rong H, Wang W, Zhang D, Zhang Z, et al. Exosomal miRNA-107 induces myeloid-derived suppressor cell expansion in gastric cancer. Cancer Manag Res. 2019;11:4023–40.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  161. Mei S, Xin J, Liu Y, Zhang Y, Liang X, Su X, Yan H, Huang Y, Yang R. MicroRNA-200c Promotes Suppressive Potential of Myeloid-Derived Suppressor Cells by Modulating PTEN and FOG2 Expression. PLoS ONE. 2015;10: e0135867.

    Article  PubMed  PubMed Central  Google Scholar 

  162. Ma HF, Shu P, Shi XH, Wang M, Jiang MF. Identification of miR-4510 as a metastasis suppressor of gastric cancer through regulation of tumor microenvironment via targeting GPC3. Clin Exp Metastasis. 2022;39:363–74.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  163. Zhang Y, Huo W, Sun L, Wu J, Zhang C, Wang H, Wang B, Wei J, Qu C, Cao H, Jiang X. Targeting miR-148b-5p Inhibits Immunity Microenvironment and Gastric Cancer Progression. Front Immunol. 2021;12: 590447.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  164. Sun L, Su Y, Jiao A, Wang X, Zhang B. T cells in health and disease. Signal Transduct Target Ther. 2023;8:235.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  165. Park J, Hsueh PC, Li Z, Ho PC. Microenvironment-driven metabolic adaptations guiding CD8(+) T cell anti-tumor immunity. Immunity. 2023;56:32–42.

    Article  PubMed  CAS  Google Scholar 

  166. Giri PS, Dwivedi M, Begum R. Decreased suppression of CD8(+) and CD4(+) T cells by peripheral regulatory T cells in generalized vitiligo due to reduced NFATC1 and FOXP3 proteins. Exp Dermatol. 2020;29:759–75.

    Article  PubMed  CAS  Google Scholar 

  167. Naqvi RA, Gupta M, George A, Naqvi AR. MicroRNAs in shaping the resolution phase of inflammation. Semin Cell Dev Biol. 2022;124:48–62.

    Article  PubMed  CAS  Google Scholar 

  168. Zabeti Touchaei A, Vahidi S. MicroRNAs as regulators of immune checkpoints in cancer immunotherapy: targeting PD-1/PD-L1 and CTLA-4 pathways. Cancer Cell Int. 2024;24:102.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  169. Miliotis C, Slack FJ. miR-105-5p regulates PD-L1 expression and tumor immunogenicity in gastric cancer. Cancer Lett. 2021;518:115–26.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  170. Xu S, Tao Z, Hai B, Liang H, Shi Y, Wang T, Song W, Chen Y, OuYang J, Chen J, et al. miR-424(322) reverses chemoresistance via T-cell immune response activation by blocking the PD-L1 immune checkpoint. Nat Commun. 2016;7:11406.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  171. Wei J, Nduom EK, Kong LY, Hashimoto Y, Xu S, Gabrusiewicz K, Ling X, Huang N, Qiao W, Zhou S, et al. MiR-138 exerts anti-glioma efficacy by targeting immune checkpoints. Neuro Oncol. 2016;18:639–48.

    Article  PubMed  CAS  Google Scholar 

  172. Zhao M, Liu Q, Liu W, Zhou H, Zang X, Lu J. MicroRNA-140 suppresses Helicobacter pylori-positive gastric cancer growth by enhancing the antitumor immune response. Mol Med Rep. 2019;20:2484–92.

    PubMed  CAS  Google Scholar 

  173. Moon JW, Kong SK, Kim BS, Kim HJ, Lim H, Noh K, Kim Y, Choi JW, Lee JH, Kim YS. IFNγ induces PD-L1 overexpression by JAK2/STAT1/IRF-1 signaling in EBV-positive gastric carcinoma. Sci Rep. 2017;7:17810.

    Article  PubMed  PubMed Central  Google Scholar 

  174. 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;26:884–93.

    Article  PubMed  Google Scholar 

  175. Wang Y, Wang D, Xie G, Yin Y, Zhao E, Tao K, Li R. MicroRNA-152 regulates immune response via targeting B7–H1 in gastric carcinoma. Oncotarget. 2017;8:28125–34.

    Article  PubMed  PubMed Central  Google Scholar 

  176. Friedrich M, Hahn M, Michel J, Sankowski R, Kilian M, Kehl N, Günter M, Bunse T, Pusch S, von Deimling A, et al. Dysfunctional dendritic cells limit antigen-specific T cell response in glioma. Neuro Oncol. 2023;25:263–76.

    Article  PubMed  CAS  Google Scholar 

  177. Sathe A, Ayala C, Bai X, Grimes SM, Lee B, Kin C, Shelton A, Poultsides G, Ji HP. GITR and TIGIT immunotherapy provokes divergent multicellular responses in the tumor microenvironment of gastrointestinal cancers. Genome Med. 2023;15:100.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  178. Scalavino V, Liso M, Serino G. Role of microRNAs in the regulation of dendritic cell generation and function. Int J Mol Sci. 2020;21(4):1319.

  179. Cui ZJ, Xie XL, Qi W, Yang YC, Bai Y, Han J, Ding Q, Jiang HQ. Cell-free miR-17-5p as a diagnostic biomarker for gastric cancer inhibits dendritic cell maturation. Onco Targets Ther. 2019;12:2661–75.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  180. Zhang Z, Chen S, Fan M, Ruan G, Xi T, Zheng L, Guo L, Ye F, Xing Y. Helicobacter pylori induces gastric cancer via down-regulating miR-375 to inhibit dendritic cell maturation. Helicobacter. 2021;26: e12813.

    Article  PubMed  CAS  Google Scholar 

  181. Saultz JN, Freud AG, Mundy-Bosse BL. MicroRNA regulation of natural killer cell development and function in leukemia. Mol Immunol. 2019;115:12–20.

    Article  PubMed  CAS  Google Scholar 

  182. Bezman NA, Chakraborty T, Bender T, Lanier LL. miR-150 regulates the development of NK and iNKT cells. J Exp Med. 2011;208:2717–31.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  183. Trotta R, Chen L, Ciarlariello D, Josyula S, Mao C, Costinean S, Yu L, Butchar JP, Tridandapani S, Croce CM, Caligiuri MA. miR-155 regulates IFN-γ production in natural killer cells. Blood. 2012;119:3478–85.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  184. Zhou C, Li X, Zhang X, Liu X, Tan Z, Yang C, Zhang J. microRNA-372 maintains oncogene characteristics by targeting TNFAIP1 and affects NFκB signaling in human gastric carcinoma cells. Int J Oncol. 2013;42:635–42.

    Article  PubMed  CAS  Google Scholar 

  185. Yang D, Zhang W, Zhang H, Zhang F, Chen L, Ma L, Larcher LM, Chen S, Liu N, Zhao Q, et al. Progress, opportunity, and perspective on exosome isolation - efforts for efficient exosome-based theranostics. Theranostics. 2020;10:3684–707.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  186. Eichmüller SB, Osen W, Mandelboim O, Seliger B. Immune modulatory microRNAs involved in tumor attack and tumor immune escape. J Natl Cancer Inst. 2017;109(10).

  187. Duan SL, Fu WJ, Jiang YK, Peng LS, Ousmane D, Zhang ZJ, Wang JP. Emerging role of exosome-derived non-coding RNAs in tumor-associated angiogenesis of tumor microenvironment. Front Mol Biosci. 2023;10:1220193.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  188. Wu S, Tang C, Zhang QW, Zhuang Q, Ye X, Xia J, Shi Y, Ning M, Dong ZX, Wan XJ. Overexpression of RAB31 in gastric cancer is associated with released exosomes and increased tumor cell invasion and metastasis. Cancer Med. 2023;12:13497–510.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  189. Lu X, Lu J, Wang S, Zhang Y, Ding Y, Shen X, Jing R, Ju S, Chen H, Cong H. Circulating serum exosomal miR-92a-3p as a novel biomarker for early diagnosis of gastric cancer. Future Oncol. 2021;17:907–19.

    Article  PubMed  CAS  Google Scholar 

  190. Nail HM, Chiu CC, Leung CH, Ahmed MMM, Wang HD. Exosomal miRNA-mediated intercellular communications and immunomodulatory effects in tumor microenvironments. J Biomed Sci. 2023;30(1):69.

  191. Xie M, Yu T, Jing X, Ma L, Fan Y, Yang F, Ma P, Jiang H, Wu X, Shu Y, Xu T. Exosomal circSHKBP1 promotes gastric cancer progression via regulating the miR-582-3p/HUR/VEGF axis and suppressing HSP90 degradation. Mol Cancer. 2020;19:112.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  192. Bai M, Li J, Yang H, Zhang H, Zhou Z, Deng T, Zhu K, Ning T, Fan Q, Ying G, Ba Y. Retraction Notice to: miR-135b Delivered by Gastric Tumor Exosomes Inhibits FOXO1 Expression in Endothelial Cells and Promotes Angiogenesis. Mol Ther. 2022;30:2636.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  193. Xia X, Wang S, Ni B, Xing S, Cao H, Zhang Z, Yu F, Zhao E, Zhao G. Hypoxic gastric cancer-derived exosomes promote progression and metastasis via MiR-301a-3p/PHD3/HIF-1α positive feedback loop. Oncogene. 2020;39:6231–44.

    Article  PubMed  CAS  Google Scholar 

  194. Qiu S, Xie L, Lu C, Gu C, Xia Y, Lv J, Xuan Z, Fang L, Yang J, Zhang L, et al. Gastric cancer-derived exosomal miR-519a-3p promotes liver metastasis by inducing intrahepatic M2-like macrophage-mediated angiogenesis. J Exp Clin Cancer Res. 2022;41:296.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  195. Zhu M, Zhang N, Ma J, He S. Integration of exosomal miR-106a and mesothelial cells facilitates gastric cancer peritoneal dissemination. Cell Signal. 2022;91: 110230.

    Article  PubMed  CAS  Google Scholar 

  196. Li Q, Li B, Li Q, Wei S, He Z, Huang X, Wang L, Xia Y, Xu Z, Li Z, et al. Exosomal miR-21-5p derived from gastric cancer promotes peritoneal metastasis via mesothelial-to-mesenchymal transition. Cell Death Dis. 2018;9:854.

    Article  PubMed  PubMed Central  Google Scholar 

  197. Yang H, Fu H, Wang B, Zhang X, Mao J, Li X, Wang M, Sun Z, Qian H, Xu W. Exosomal miR-423-5p targets SUFU to promote cancer growth and metastasis and serves as a novel marker for gastric cancer. Mol Carcinog. 2018;57:1223–36.

    Article  PubMed  CAS  Google Scholar 

  198. Li Z, Suo B, Long G, Gao Y, Song J, Zhang M, Feng B, Shang C, Wang D. Exosomal miRNA-16-5p Derived From M1 Macrophages Enhances T Cell-Dependent Immune Response by Regulating PD-L1 in Gastric Cancer. Front Cell Dev Biol. 2020;8: 572689.

    Article  PubMed  PubMed Central  Google Scholar 

  199. Hsieh CH, Tai SK, Yang MH. Snail-overexpressing Cancer Cells Promote M2-Like Polarization of Tumor-Associated Macrophages by Delivering MiR-21-Abundant Exosomes. Neoplasia. 2018;20:775–88.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  200. Wei S, Peng L, Yang J, Sang H, Jin D, Li X, Chen M, Zhang W, Dang Y, Zhang G. Exosomal transfer of miR-15b-3p enhances tumorigenesis and malignant transformation through the DYNLT1/Caspase-3/Caspase-9 signaling pathway in gastric cancer. J Exp Clin Cancer Res. 2020;39:32.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  201. Jiao Y, Zhang L, Li J, He Y, Zhang X, Li J. Exosomal miR-122-5p inhibits tumorigenicity of gastric cancer by downregulating GIT1. Int J Biol Markers. 2021;36:36–46.

    Article  PubMed  CAS  Google Scholar 

  202. Xu G, Zhang B, Ye J, Cao S, Shi J, Zhao Y, Wang Y, Sang J, Yao Y, Guan W, et al. Exosomal miRNA-139 in cancer-associated fibroblasts inhibits gastric cancer progression by repressing MMP11 expression. Int J Biol Sci. 2019;15:2320–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  203. Wu C, Li D, Cheng X, Gu H, Qian Y, Feng L. Downregulation of cancer-associated fibroblast exosome-derived miR-29b-1-5p restrains vasculogenic mimicry and apoptosis while accelerating migration and invasion of gastric cancer cells via immunoglobulin domain-containing 1/zonula occluden-1 axis. Cell Cycle. 2023;22:1807–26.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  204. Wang Y, Li T, Yang L, Zhang X, Wang X, Su X, Ji C, Wang Z. Cancer-associated fibroblast-released extracellular vesicles carrying miR-199a-5p induces the progression of​ gastric cancer through regulation of FKBP5-mediated AKT1/mTORC1 signaling pathway. Cell Cycle. 2022;21:2590–601.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  205. Thrift AP, El-Serag HB. Burden of Gastric Cancer. Clin Gastroenterol Hepatol. 2020;18:534–42.

    Article  PubMed  Google Scholar 

  206. Zhang L, Lu Z, Zhao X. Targeting Bcl-2 for cancer therapy. Biochim Biophys Acta Rev Cancer. 2021;1876: 188569.

    Article  PubMed  CAS  Google Scholar 

  207. Osterlund P, Kinos S, Pfeiffer P, Salminen T, Kwakman JJM, Frödin JE, Shah CH, Sorbye H, Ristamäki R, Halonen P, et al. Continuation of fluoropyrimidine treatment with S-1 after cardiotoxicity on capecitabine- or 5-fluorouracil-based therapy in patients with solid tumours: a multicentre retrospective observational cohort study. ESMO Open. 2022;7: 100427.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  208. Nakamura Y, Kawazoe A, Lordick F, Janjigian YY, Shitara K. Biomarker-targeted therapies for advanced-stage gastric and gastro-oesophageal junction cancers: an emerging paradigm. Nat Rev Clin Oncol. 2021;18:473–87.

    Article  PubMed  Google Scholar 

  209. Li Y, Tian Z, Tan Y, Lian G, Chen S, Chen S, Li J, Li X, Huang K, Chen Y. Bmi-1-induced miR-27a and miR-155 promote tumor metastasis and chemoresistance by targeting RKIP in gastric cancer. Mol Cancer. 2020;19:109.

    Article  PubMed  PubMed Central  Google Scholar 

  210. Deng LM, Tan T, Zhang TY, Xiao XF, Gu H. miR-1 reverses multidrug resistance in gastric cancer cells via downregulation of sorcin through promoting the accumulation of intracellular drugs and apoptosis of cells. Int J Oncol. 2019;55:451–61.

    PubMed  PubMed Central  CAS  Google Scholar 

  211. Jin HF, Wang JF, Shao M, Zhou K, Ma X, Lv XP. Down-Regulation of miR-7 in Gastric Cancer Is Associated With Elevated LDH-A Expression and Chemoresistance to Cisplatin. Front Cell Dev Biol. 2020;8: 555937.

    Article  PubMed  PubMed Central  Google Scholar 

  212. Zhao D, Zhang Y, Song L. MiR-16-1 Targeted Silences Far Upstream Element Binding Protein 1 to Advance the Chemosensitivity to Adriamycin in Gastric Cancer. Pathol Oncol Res. 2018;24:483–8.

    Article  PubMed  CAS  Google Scholar 

  213. Wu DM, Hong XW, Wang LL, Cui XF, Lu J, Chen GQ, Zheng YL. MicroRNA-17 inhibition overcomes chemoresistance and suppresses epithelial-mesenchymal transition through a DEDD-dependent mechanism in gastric cancer. Int J Biochem Cell Biol. 2018;102:59–70.

    Article  PubMed  CAS  Google Scholar 

  214. Wang Z, Ji F. Downregulation of microRNA-17-5p inhibits drug resistance of gastric cancer cells partially through targeting p21. Oncol Lett. 2018;15:4585–91.

    PubMed  PubMed Central  Google Scholar 

  215. Xia L, Zhang D, Du R, Pan Y, Zhao L, Sun S, Hong L, Liu J, Fan D. miR-15b and miR-16 modulate multidrug resistance by targeting BCL2 in human gastric cancer cells. Int J Cancer. 2008;123:372–9.

    Article  PubMed  CAS  Google Scholar 

  216. Wang F, Li T, Zhang B, Li H, Wu Q, Yang L, Nie Y, Wu K, Shi Y, Fan D. MicroRNA-19a/b regulates multidrug resistance in human gastric cancer cells by targeting PTEN. Biochem Biophys Res Commun. 2013;434:688–94.

    Article  PubMed  CAS  Google Scholar 

  217. Zhu M, Zhou X, Du Y, Huang Z, Zhu J, Xu J, Cheng G, Shu Y, Liu P, Zhu W, Wang T. miR-20a induces cisplatin resistance of a human gastric cancer cell line via targeting CYLD. Mol Med Rep. 2016;14:1742–50.

    Article  PubMed  CAS  Google Scholar 

  218. Zhou L, Li X, Zhou F, Jin Z, Chen D, Wang P, Zhang S, Zhuge Y, Shang Y, Zou X. Downregulation of leucine-rich repeats and immunoglobulin-like domains 1 by microRNA-20a modulates gastric cancer multidrug resistance. Cancer Sci. 2018;109:1044–54.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  219. Yang SM, Huang C, Li XF, Yu MZ, He Y, Li J. miR-21 confers cisplatin resistance in gastric cancer cells by regulating PTEN. Toxicology. 2013;306:162–8.

    Article  PubMed  CAS  Google Scholar 

  220. An Y, Zhang Z, Shang Y, Jiang X, Dong J, Yu P, Nie Y, Zhao Q. miR-23b-3p regulates the chemoresistance of gastric cancer cells by targeting ATG12 and HMGB2. Cell Death Dis. 2015;6: e1766.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  221. He J, Qi H, Chen F, Cao C. MicroRNA-25 contributes to cisplatin resistance in gastric cancer cells by inhibiting forkhead box O3a. Oncol Lett. 2017;14:6097–102.

    PubMed  PubMed Central  Google Scholar 

  222. Abbasi A, Hosseinpourfeizi M, Safaralizadeh R. All-trans retinoic acid-mediated miR-30a up-regulation suppresses autophagy and sensitizes gastric cancer cells to cisplatin. Life Sci. 2022;307: 120884.

    Article  PubMed  CAS  Google Scholar 

  223. Korourian A, Roudi R, Shariftabrizi A, Madjd Z. MicroRNA-31 inhibits RhoA-mediated tumor invasion and chemotherapy resistance in MKN-45 gastric adenocarcinoma cells. Exp Biol Med (Maywood). 2017;242:1842–7.

    Article  PubMed  CAS  Google Scholar 

  224. Ji Q, Hao X, Meng Y, Zhang M, Desano J, Fan D, Xu L. Restoration of tumor suppressor miR-34 inhibits human p53-mutant gastric cancer tumorspheres. BMC Cancer. 2008;8:266.

    Article  PubMed  PubMed Central  Google Scholar 

  225. Zhang Z, Kong Y, Yang W, Ma F, Zhang Y, Ji S, Ma EM, Liu H, Chen Y, Hua Y. Upregulation of microRNA-34a enhances the DDP sensitivity of gastric cancer cells by modulating proliferation and apoptosis via targeting MET. Oncol Rep. 2016;36:2391–7.

    Article  PubMed  CAS  Google Scholar 

  226. Zheng H, Wang JJ, Yang XR, Yu YL. Upregulation of miR-34c after silencing E2F transcription factor 1 inhibits paclitaxel combined with cisplatin resistance in gastric cancer cells. World J Gastroenterol. 2020;26:499–513.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  227. Wu H, Huang M, Lu M, Zhu W, Shu Y, Cao P, Liu P. Regulation of microtubule-associated protein tau (MAPT) by miR-34c-5p determines the chemosensitivity of gastric cancer to paclitaxel. Cancer Chemother Pharmacol. 2013;71:1159–71.

    Article  PubMed  CAS  Google Scholar 

  228. Lang C, Xu M, Zhao Z, Chen J, Zhang L. MicroRNA-96 expression induced by low-dose cisplatin or doxorubicin regulates chemosensitivity, cell death and proliferation in gastric cancer SGC7901 cells by targeting FOXO1. Oncol Lett. 2018;16:4020–6.

    PubMed  PubMed Central  Google Scholar 

  229. Zhang Y, Xu W, Ni P, Li A, Zhou J, Xu S. MiR-99a and MiR-491 Regulate Cisplatin Resistance in Human Gastric Cancer Cells by Targeting CAPNS1. Int J Biol Sci. 2016;12:1437–47.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  230. Bao J, Xu Y, Wang Q, Zhang J, Li Z, Li D, Li J. miR-101 alleviates chemoresistance of gastric cancer cells by targeting ANXA2. Biomed Pharmacother. 2017;92:1030–7.

    Article  PubMed  CAS  Google Scholar 

  231. Zhang Y, Lu Q, Cai X. MicroRNA-106a induces multidrug resistance in gastric cancer by targeting RUNX3. FEBS Lett. 2013;587:3069–75.

    Article  PubMed  CAS  Google Scholar 

  232. Fang Y, Shen H, Li H, Cao Y, Qin R, Long L, Zhu X, Xie C, Xu W. miR-106a confers cisplatin resistance by regulating PTEN/Akt pathway in gastric cancer cells. Acta Biochim Biophys Sin (Shanghai). 2013;45:963–72.

    Article  PubMed  CAS  Google Scholar 

  233. Wang P, Li Z, Liu H, Zhou D, Fu A, Zhang E. MicroRNA-126 increases chemosensitivity in drug-resistant gastric cancer cells by targeting EZH2. Biochem Biophys Res Commun. 2016;479:91–6.

    Article  PubMed  CAS  Google Scholar 

  234. Liang L, Kang H, Jia J. HCP5 contributes to cisplatin resistance in gastric cancer through miR-128/HMGA2 axis. Cell Cycle. 2021;20:1080–90.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  235. Lu C, Shan Z, Li C, Yang L. MiR-129 regulates cisplatin-resistance in human gastric cancer cells by targeting P-gp. Biomed Pharmacother. 2017;86:450–6.

    Article  PubMed  CAS  Google Scholar 

  236. Chu H, Han N, Xu J. CMPK1 Regulated by miR-130b Attenuates Response to 5-FU Treatment in Gastric Cancer. Front Oncol. 2021;11: 637470.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  237. Zhang L, Guo X, Zhang D, Fan Y, Qin L, Dong S, Zhang L. Upregulated miR-132 in Lgr5(+) gastric cancer stem cell-like cells contributes to cisplatin-resistance via SIRT1/CREB/ABCG2 signaling pathway. Mol Carcinog. 2017;56:2022–34.

    Article  PubMed  CAS  Google Scholar 

  238. Pan Y, Ren F, Zhang W, Liu G, Yang D, Hu J, Feng K, Feng Y. Regulation of BGC-823 cell sensitivity to adriamycin via miRNA-135a-5p. Oncol Rep. 2014;32:2549–56.

    Article  PubMed  CAS  Google Scholar 

  239. Zhou J, Chen Q. Poor expression of microRNA-135b results in the inhibition of cisplatin resistance and proliferation and induces the apoptosis of gastric cancer cells through MST1-mediated MAPK signaling pathway. Faseb j. 2019;33:3420–36.

    Article  PubMed  CAS  Google Scholar 

  240. Zeng JF, Ma XQ, Wang LP, Wang W. MicroRNA-145 exerts tumor-suppressive and chemo-resistance lowering effects by targeting CD44 in gastric cancer. World J Gastroenterol. 2017;23:2337–45.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  241. 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;410:212–27.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  242. Ning J, Jiao Y, Xie X, Deng X, Zhang Y, Yang Y, Zhao C, Wang H, Gu K. miR-138-5p modulates the expression of excision repair cross-complementing proteins ERCC1 and ERCC4, and regulates the sensitivity of gastric cancer cells to cisplatin. Oncol Rep. 2019;41:1131–9.

    PubMed  CAS  Google Scholar 

  243. Lin H, Ni R, Li D, Zhao M, Li Y, Li K, Zhang Q, Huang C, Huang S. LncRNA MIR155HG Overexpression Promotes Proliferation, Migration, and Chemoresistance in Gastric Cancer Cells. Int J Med Sci. 2023;20:933–42.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  244. Wang M, Qiu R, Yu S, Xu X, Li G, Gu R, Tan C, Zhu W, Shen B. Paclitaxel-resistant gastric cancer MGC-803 cells promote epithelial-to-mesenchymal transition and chemoresistance in paclitaxel-sensitive cells via exosomal delivery of miR-155-5p. Int J Oncol. 2019;54:326–38.

    PubMed  CAS  Google Scholar 

  245. Lin Y, Zhao J, Wang H, Cao J, Nie Y. miR-181a modulates proliferation, migration and autophagy in AGS gastric cancer cells and downregulates MTMR3. Mol Med Rep. 2017;15:2451–6.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  246. Jin L, Ma X, Zhang N, Zhang Q, Chen X, Zhang Z, Ding G, Yu H. Targeting Oncogenic miR-181a-2-3p Inhibits Growth and Suppresses Cisplatin Resistance of Gastric Cancer. Cancer Manag Res. 2021;13:8599–609.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  247. Zhu W, Shan X, Wang T, Shu Y, Liu P. miR-181b modulates multidrug resistance by targeting BCL2 in human cancer cell lines. Int J Cancer. 2010;127:2520–9.

    Article  PubMed  CAS  Google Scholar 

  248. Li Q, Wang JX, He YQ, Feng C, Zhang XJ, Sheng JQ, Li PF. MicroRNA-185 regulates chemotherapeutic sensitivity in gastric cancer by targeting apoptosis repressor with caspase recruitment domain. Cell Death Dis. 2014;5: e1197.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  249. Lee SD, Yu D, Lee DY, Shin HS, Jo JH, Lee YC. Upregulated microRNA-193a-3p is responsible for cisplatin resistance in CD44(+) gastric cancer cells. Cancer Sci. 2019;110:662–73.

    Article  PubMed  CAS  Google Scholar 

  250. Nie H, Mu J, Wang J, Li Y. miR-195-5p regulates multi-drug resistance of gastric cancer cells via targeting ZNF139. Oncol Rep. 2018;40:1370–8.

    PubMed  PubMed Central  CAS  Google Scholar 

  251. Zhu W, Xu H, Zhu D, Zhi H, Wang T, Wang J, Jiang B, Shu Y, Liu P. miR-200bc/429 cluster modulates multidrug resistance of human cancer cell lines by targeting BCL2 and XIAP. Cancer Chemother Pharmacol. 2012;69:723–31.

    Article  PubMed  CAS  Google Scholar 

  252. Jiang T, Dong P, Li L, Ma X, Xu P, Zhu H, Wang Y, Yang B, Liu K, Liu J, et al. MicroRNA-200c regulates cisplatin resistance by targeting ZEB2 in human gastric cancer cells. Oncol Rep. 2017;38:151–8.

    Article  PubMed  CAS  Google Scholar 

  253. Sacconi A, Biagioni F, Canu V, Mori F, Di Benedetto A, Lorenzon L, Ercolani C, Di Agostino S, Cambria AM, Germoni S, et al. miR-204 targets Bcl-2 expression and enhances responsiveness of gastric cancer. Cell Death Dis. 2012;3: e423.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  254. Li LQ, Pan D, Chen Q, Zhang SW, Xie DY, Zheng XL, Chen H. Sensitization of Gastric Cancer Cells to 5-FU by MicroRNA-204 Through Targeting the TGFBR2-Mediated Epithelial to Mesenchymal Transition. Cell Physiol Biochem. 2018;47:1533–45.

    Article  PubMed  CAS  Google Scholar 

  255. Zhang XL, Shi HJ, Wang JP, Tang HS, Wu YB, Fang ZY, Cui SZ, Wang LT. MicroRNA-218 is upregulated in gastric cancer after cytoreductive surgery and hyperthermic intraperitoneal chemotherapy and increases chemosensitivity to cisplatin. World J Gastroenterol. 2014;20:11347–55.

    Article  PubMed  PubMed Central  Google Scholar 

  256. Zhou X, Jin W, Jia H, Yan J, Zhang G. MiR-223 promotes the cisplatin resistance of human gastric cancer cells via regulating cell cycle by targeting FBXW7. J Exp Clin Cancer Res. 2015;34:28.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  257. Gao H, Ma J, Cheng Y, Zheng P. Exosomal Transfer of Macrophage-Derived miR-223 Confers Doxorubicin Resistance in Gastric Cancer. Onco Targets Ther. 2020;13:12169–79.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  258. Jian B, Li Z, Xiao D, He G, Bai L, Yang Q. Downregulation of microRNA-193-3p inhibits tumor proliferation migration and chemoresistance in human gastric cancer by regulating PTEN gene. Tumour Biol. 2016;37:8941–9.

    Article  PubMed  CAS  Google Scholar 

  259. Zhu T, Hu Z, Wang Z, Ding H, Li R, Wang J, Wang G. microRNA-301b-3p from mesenchymal stem cells-derived extracellular vesicles inhibits TXNIP to promote multidrug resistance of gastric cancer cells. Cell Biol Toxicol. 2023;39:1923–37.

    Article  PubMed  CAS  Google Scholar 

  260. Tian L, Zhao Z, Xie L, Zhu J. MiR-361-5p suppresses chemoresistance of gastric cancer cells by targeting FOXM1 via the PI3K/Akt/mTOR pathway. Oncotarget. 2018;9:4886–96.

    Article  PubMed  Google Scholar 

  261. Zhang PF, Sheng LL, Wang G, Tian M, Zhu LY, Zhang R, Zhang J, Zhu JS. miR-363 promotes proliferation and chemo-resistance of human gastric cancer via targeting of FBW7 ubiquitin ligase expression. Oncotarget. 2016;7:35284–92.

    Article  PubMed  PubMed Central  Google Scholar 

  262. Zhou N, Qu Y, Xu C, Tang Y. Upregulation of microRNA-375 increases the cisplatin-sensitivity of human gastric cancer cells by regulating ERBB2. Exp Ther Med. 2016;11:625–30.

    Article  PubMed  CAS  Google Scholar 

  263. Li Y, Liu H, Cui Y, Chen H, Cui X, Shao J, Su F, He X. miR-424-3p Contributes to the Malignant Progression and Chemoresistance of Gastric Cancer. Onco Targets Ther. 2020;13:12201–11.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  264. Wang X, He R, Geng L, Yuan J, Fan H. Ginsenoside Rg3 Alleviates Cisplatin Resistance of Gastric Cancer Cells Through Inhibiting SOX2 and the PI3K/Akt/mTOR Signaling Axis by Up-Regulating miR-429. Front Genet. 2022;13: 823182.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  265. Wu S, Xie J, Shi H, Wang ZW. miR-492 promotes chemoresistance to CDDP and metastasis by targeting inhibiting DNMT3B and induces stemness in gastric cancer. Biosci Rep. 2020;40(3):BSR20194342.

  266. Makinoya M, Miyatani K, Matsumi Y, Sakano Y, Shimizu S, Shishido Y, Hanaki T, Kihara K, Matsunaga T, Yamamoto M, et al. Exosomal miR-493 suppresses MAD2L1 and induces chemoresistance to intraperitoneal paclitaxel therapy in gastric cancer patients with peritoneal metastasis. Sci Rep. 2024;14:10075.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  267. Chen S, Wu J, Jiao K, Wu Q, Ma J, Chen D, Kang J, Zhao G, Shi Y, Fan D, Zhao G. MicroRNA-495-3p inhibits multidrug resistance by modulating autophagy through GRP78/mTOR axis in gastric cancer. Cell Death Dis. 2018;9:1070.

    Article  PubMed  PubMed Central  Google Scholar 

  268. Zhu W, Zhu D, Lu S, Wang T, Wang J, Jiang B, Shu Y, Liu P. miR-497 modulates multidrug resistance of human cancer cell lines by targeting BCL2. Med Oncol. 2012;29:384–91.

    Article  PubMed  CAS  Google Scholar 

  269. Lin H, Zhang L, Zhang C, Liu P. Exosomal MiR-500a-3p promotes cisplatin resistance and stemness via negatively regulating FBXW7 in gastric cancer. J Cell Mol Med. 2020;24:8930–41.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  270. Wang T, Ge G, Ding Y, Zhou X, Huang Z, Zhu W, Shu Y, Liu P. MiR-503 regulates cisplatin resistance of human gastric cancer cell lines by targeting IGF1R and BCL2. Chin Med J (Engl). 2014;127:2357–62.

    Article  PubMed  CAS  Google Scholar 

  271. Shang Y, Zhang Z, Liu Z, Feng B, Ren G, Li K, Zhou L, Sun Y, Li M, Zhou J, et al. miR-508-5p regulates multidrug resistance of gastric cancer by targeting ABCB1 and ZNRD1. Oncogene. 2014;33:3267–76.

    Article  PubMed  CAS  Google Scholar 

  272. Wang J, Xue X, Hong H, Qin M, Zhou J, Sun Q, Liang H, Gao L. Upregulation of microRNA-524-5p enhances the cisplatin sensitivity of gastric cancer cells by modulating proliferation and metastasis via targeting SOX9. Oncotarget. 2017;8:574–82.

    Article  PubMed  Google Scholar 

  273. Shen B, Yu S, Zhang Y, Yuan Y, Li X, Zhong J, Feng J. miR-590-5p regulates gastric cancer cell growth and chemosensitivity through RECK and the AKT/ERK pathway. Onco Targets Ther. 2016;9:6009–19.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  274. Jiang L, Yang W, Bian W, Yang H, Wu X, Li Y, Feng W, Liu X. MicroRNA-623 Targets Cyclin D1 to Inhibit Cell Proliferation and Enhance the Chemosensitivity of Cells to 5-Fluorouracil in Gastric Cancer. Oncol Res. 2018;27:19–27.

    Article  PubMed  PubMed Central  Google Scholar 

  275. Cao W, Wei W, Zhan Z, Xie D, Xie Y, Xiao Q. Regulation of drug resistance and metastasis of gastric cancer cells via the microRNA647-ANK2 axis. Int J Mol Med. 2018;41:1958–66.

    PubMed  PubMed Central  CAS  Google Scholar 

  276. Aliabadi P, Sadri M, Siri G, Ebrahimzadeh F, Yazdani Y, Gusarov AM, Kharkouei SA, Asadi F, Adili A, Mardi A, Mohammadi H. Restoration of miR-648 overcomes 5-FU-resistance through targeting ET-1 in gastric cancer cells in-vitro. Pathol Res Pract. 2022;239: 154139.

    Article  PubMed  CAS  Google Scholar 

  277. Shang J, Wang Q, Wang J, Xu B, Liu S. miR-708-3p promotes gastric cancer progression through downregulating ETNK1. Heliyon. 2023;9: e19544.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  278. Chen Q, Lin L, Xiong B, Yang W, Huang J, Shi H, Wang Z. MiR-873-5p targets THUMPD1 to inhibit gastric cancer cell behavior and chemoresistance. J Gastrointest Oncol. 2021;12:2061–72.

    Article  PubMed  PubMed Central  Google Scholar 

  279. Huang H, Tang J, Zhang L, Bu Y, Zhang X. miR-874 regulates multiple-drug resistance in gastric cancer by targeting ATG16L1. Int J Oncol. 2018;53:2769–79.

    PubMed  CAS  Google Scholar 

  280. Nishibeppu K, Komatsu S, Imamura T, Kiuchi J, Kishimoto T, Arita T, Kosuga T, Konishi H, Kubota T, Shiozaki A, et al. Plasma microRNA profiles: identification of miR-1229-3p as a novel chemoresistant and prognostic biomarker in gastric cancer. Sci Rep. 2020;10:3161.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  281. Cao W, Wei W, Zhan Z, Xie Y, Xiao Q. MiR-1284 modulates multidrug resistance of gastric cancer cells by targeting EIF4A1. Oncol Rep. 2016;35:2583–91.

    Article  PubMed  CAS  Google Scholar 

  282. Yan R, Li K, Yuan DW, Wang HN, Zhang Y, Dang CX, Zhu K. Downregulation of microRNA-4295 enhances cisplatin-induced gastric cancer cell apoptosis through the EGFR/PI3K/Akt signaling pathway by targeting LRIG1. Int J Oncol. 2018;53:2566–78.

    PubMed  PubMed Central  CAS  Google Scholar 

  283. Jing X, Xie M, Ding K, Xu T, Fang Y, Ma P, Shu Y. Exosome-transmitted miR-769-5p confers cisplatin resistance and progression in gastric cancer by targeting CASP9 and promoting the ubiquitination degradation of p53. Clin Transl Med. 2022;12: e780.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  284. Jiang L, Zhang Y, Guo L, Liu C, Wang P, Ren W. Exosomal microRNA-107 reverses chemotherapeutic drug resistance of gastric cancer cells through HMGA2/mTOR/P-gp pathway. BMC Cancer. 2021;21:1290.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  285. Hao NB, He YF, Li XQ, Wang K, Wang RL. The role of miRNA and lncRNA in gastric cancer. Oncotarget. 2017;8:81572–82.

    Article  PubMed  PubMed Central  Google Scholar 

  286. Cui M, Wang H, Yao X, Zhang D, Xie Y, Cui R, Zhang X. Circulating MicroRNAs in Cancer: Potential and Challenge. Front Genet. 2019;10:626.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This study was supported by the National Natural Science Foundation of China (No. 82202989); the China Postdoctoral Science Foundation (No. 2022M722279, China); the Sichuan Science and Technology Program (No. 2023YFS0163, China); Postdoctoral Research Project of West China Hospital, Sichuan University, Chengdu, China (No. 2021HXBH045); Fundamental Research Funds for the Central Universities (No. 2022SCU1206); and Sichuan University Postdoctoral Interdisciplinary Innovation Fund (awarded to LZ, China).

Author information

Authors and Affiliations

Authors

Contributions

XZ Y and Y Z conceptualized the manuscript. XZ Y wrote the first draft. QH Z, FM L and LL Z contributed substantially by revising the manuscript. All authors approved the submitted version and are fully accountable for every aspect of the work.

Corresponding authors

Correspondence to Fengming Luo, Qinghua Zhou or Lingling Zhu.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yu, X., Zhang, Y., Luo, F. et al. The role of microRNAs in the gastric cancer tumor microenvironment. Mol Cancer 23, 170 (2024). https://doi.org/10.1186/s12943-024-02084-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12943-024-02084-x

Keywords