Skip to main content

The oncogenic mechanisms of the Janus kinase-signal transducer and activator of transcription pathway in digestive tract tumors

Abstract

Digestive tract tumors are heterogeneous and involve the dysregulation of multiple signaling pathways. The Janus kinase-signal transducer and activator of transcription (JAK–STAT) pathway plays a notable role in the oncogenesis of digestive tract tumors. Typically activated by pro-inflammatory cytokines, it regulates important biological processes, such as cell growth, differentiation, apoptosis, immune responses, and inflammation. The aberrant activation of this pathway manifests in different forms, including mutations in JAKs, overexpression of cytokine receptors, and sustained STAT activation, and contributes to promoting the malignant characteristics of cancer cells, including uncontrolled proliferation, resistance to apoptosis, enhanced invasion and metastasis, angiogenesis, acquisition of stem-like properties, and drug resistance. Numerous studies have shown that aberrant activation of the JAK-STAT pathway is closely related to the development and progression of digestive tract tumors, contributing to tumor survival, angiogenesis, changes in the tumor microenvironment, and even immune escape processes. In addition, this signaling pathway also affects the sensitivity of digestive tract tumors to chemotherapy and targeted therapy. Therefore, it is crucial to comprehensively understand the oncogenic mechanisms underlying the JAK-STAT pathway in order to develop effective therapeutic strategies against digestive tract tumors. Currently, several JAK–STAT inhibitors are undergoing clinical and preclinical trials as potential treatments for various human diseases. However, further investigation is required to determine the role of this pathway, as well as the effectiveness and safety of its inhibitors, especially in the context of digestive tract tumors. In this review, we provide an overview of the structure, classic activation, and negative regulation of the JAK-STAT pathway. Furthermore, we discuss the pathogenic mechanisms of JAK-STAT signaling in different digestive tract tumors, with the aim of identifying potential novel therapeutic targets.

Video Abstract

Introduction

The Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway is a crucial cell signaling pathway that is frequently activated by an extensive repertoire of extracellular cytokines and growth factors [1,2,3]. It plays a critical role in regulating essential biological processes, including cellular processes, inflammation, and immunological responses. As a result, it is evolutionarily conserved across different species [4,5,6,7]. Activation of the JAK-STAT pathway begins with the binding of an extracellular ligand to the cell surface receptor. This process triggers a cascade of complex steps, which includes the recruitment and subsequent phosphorylation of JAKs within the receptor complex, the phosphorylation and dimerization of STAT, the combination of STAT dimers with specific responsive element regions on the nucleus, and ultimately, the regulation of target gene transcription [8,9,10,11]. In this way, the extracellular signals and stimuli are relayed to the nucleus (Fig. 1). Under normal physiological conditions, the JAK-STAT signaling pathway regulates gene expression and cellular function by responding to extracellular signal molecules such as cytokines and growth factors. Numerous studies have demonstrated that JAK-STAT is involved in multiple biological processes, including cell proliferation, differentiation, apoptosis, immune response, hematopoietic regulation and embryonic development. Specifically, in the immune system, it participates in regulating the development, proliferation, and function of T cells and B cells; in the hematopoietic system, it controls the proliferation and differentiation of blood cells; and in embryonic development, it plays a role in organ formation and cell fate determination. Furthermore, the JAK-STAT signaling pathway can interact with other signaling pathways to form complex network regulatory systems. This network regulation helps maintain the biological balance of normal cells and ensures the normal function of tissues and organs.

Fig. 1
figure 1

The components and activation of the Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway

The dysregulation of the JAK–STAT pathway has been found to contribute to carcinogenesis by promoting several oncogenic processes, including cell proliferation, invasion, metastasis, anti-apoptosis, immune escape, and angiogenesis [1, 12, 13]. Numerous studies have reported hyperactivation and frequent mutations in JAK–STAT signaling proteins in various human disorders, such as rheumatoid arthritis, inflammatory bowel disease, inflammatory skin conditions, myeloproliferative neoplasms, and solid tumors [14,15,16,17]. Considering its near-ubiquitous role in diverse diseases, an increasing number of small-molecule inhibitors or natural products targeting JAK-STAT proteins have been synthesized or developed [18,19,20,21]. Of them, many have been approved for clinical use and more selective inhibitors are currently undergoing clinical investigation [22,23,24,25].

Digestive tract tumors encompass a heterogeneous range of cancers, typically including esophageal, gastric, colorectal, liver, and pancreatic cancers [26, 27]. Despite the widespread availability of endoscopic examinations has greatly improved the early detection rates for certain digestive tract tumors, their non-specific symptoms and the limited therapeutic interventions for advanced-stage cancers pose a formidable challenge to improving the survival rates of patients [28,29,30]. Based on the GLOBOCAN 2020 statistics, digestive tract cancers comprised 23.4% of all cancer cases and 30.9% of all cancer-related deaths worldwide, emphasizing the substantial burden they impose on global public health [31, 32]. Hence, elucidating their pathogenesis and exploring novel biomarkers and therapeutic targets is imperative [33].

Advanced molecular biology and cutting-edge sequencing technologies have consistently identified the abnormal activation of JAK-STAT signaling in several digestive tract tumors. Moreover, the dysregulation of this pathway is associated with more malignant cell behaviors and tumor development, such as increased cell migration, invasion, and metastasis [34]. By regulating the reorganization of the cell skeleton and the expression of adhesion molecules, it promotes the migration ability of tumor cells [35,36,37,38]. At the same time, the JAK-STAT signaling pathway enhances the invasive ability of tumor cells by regulating the expression and activity of matrix metalloproteinases (MMPs), which can degrade extracellular matrix and provide invasion pathways for tumor cells [39,40,41]. In addition, it interacts with key transcription factors that promote epithelial-mesenchymal transition (EMT), making tumor cells more invasive [42,43,44,45]. These findings highlight that the JAK-STAT pathway significantly drives the progression and spread of digestive tract tumors. Reviewing its role in digestive tract tumors comprehensively will help us better understand its regulatory mechanisms and provide insights for developing more precise targeted treatment strategies.

Here, we have reviewed the involvement and mechanisms of the aberrantly activated JAK-STAT pathway in digestive tract tumors. Further, we have systematically analyzed the clinical significance of this pathway as a source of both potential biomarkers for early screening and therapeutic targets. Finally, we have discussed the application and prospects of targeting this pathway to enhance the clinical management of digestive tract tumors.

JAK-STAT pathway

The JAK-STAT pathway is a crucial signaling pathway inside the cell that involves two protein families: JAK and STAT [10].

The JAK family comprises four non-receptor tyrosine kinases, namely JAK1, JAK2, JAK3, and tyrosine kinase 2, which have a shared domain structure but distinct functions within the cell [34, 46, 47]. By analyzing, we have been able to understand the complete domain structure of JAKs with apparent molecular masses of 120–140 kDa [48]. JAKs are composed of seven Jak homology (JH) regions spanning four functional domains: a C-terminal tyrosine kinase domain formed by JH1, a pseudokinase domain constituted by JH2, a Src-homology 2 (SH2) domain comprising the JH3-JH4 regions, and an N-terminal FERM domain (band 4.1, ezrin, radixin, and moesin) containing the JH5-JH7 regions [49,50,51]. Thoroughly dissecting the functions and interactions of each domain within the JAK family will enhance our understanding of its role in signal transduction. The kinase domain, which is the most pivotal domain of JAKs, exhibits tyrosine kinase activity to phosphorylate target substrates like STATs, thus activating downstream signaling pathways and cellular responses [52]. Inhibiting the activity of the kinase domain usually disrupts an aberrantly activated JAK-STAT pathway, making this domain the major target for the development of JAK inhibitors [53,54,55]. The characteristic pseudokinase domain, which is beside the kinase domain, executes crucial regulatory functions rather than catalytic functions. It modulates JAK activation and substrate specificity by interacting with other proteins, but also prevents the excessive activation of JAK-STAT signaling by providing negative feedback [56]. Moreover, mutations in the pseudokinase domain have been proven to affect the basal activity of the kinase domain [57]. The SH2 domain functions as scaffolding to both facilitate the localization of JAK to activated receptors and phosphorylate STAT. As a result, it affects the nuclear translocation of STAT and downstream gene expression regulation [58,59,60]. The FERM domain is involved in interacting with transmembrane receptors and maintaining kinase activity [61, 62]. Several studies have reported that variations in the FERM domain contribute to aberrant JAK-STAT signaling in a wide range of human diseases [63, 64].

Signal transduction through the JAK-STAT pathway is mediated by four cytosolic JAKs situated near the cell membrane [65, 66]. Each JAK can bind to multiple types of cytokine receptors, resulting in different downstream effects [67]. JAK1, JAK2, and tyrosine kinase 2 are ubiquitously expressed in mammals, whereas the expression of JAK3 is predominantly restricted to hematopoietic, endothelial, and vascular smooth muscle cells [58, 68,69,70,71]. These four JAKs have been widely recognized as potential drug targets in diverse diseases, such as leukemia, polycythemia vera, myelofibrosis, essential thrombocythemia, cutaneous T-cell lymphoma, and inflammatory bowel disease [72,73,74,75]. However, the therapeutic effectiveness and safety of targeting the JAK family needs further clinical verification [76, 77].

The STAT family, first discovered while studying the activation of the interferon system in 1994, is a family of seven latent cytoplasmic transcription factors (STAT1-STAT4, STAT5A, STAT5B, and STAT6) in humans with a conserved separate window ranging from 750 to 850 amino acids [78,79,80]. Most STAT proteins possess similar structures: an N-terminal domain, a coiled-coil domain, a DNA-binding domain, a linker domain, an SH2 domain, and a C-terminal transactivation domain (STAT2 and STAT6 are exceptions because they lack the PMSP motif) [81,82,83]. The C-terminal transactivation domain is the main site for the phosphorylation of serine residues. The SH2 domain mediates STAT phosphorylation as well as the interaction between STATs and JAKs leading to STAT dimerization. The DNA-binding domain contains precise amino acid sequences to recognize and bind to specific DNA sequences, thus dictating DNA-binding specificity [84,85,86,87,88]. Multiple studies have discovered that different molecules activate specific STATs (especially STAT3 and STAT5) to initiate distinct regulatory mechanisms and functions that contribute to normal physiology as well as disease development [82, 89, 90]. Moreover, STAT3 and STAT5 are considered the most significant of all STATs because they are involved in malignant transformation [91,92,93]. Inhibiting constitutively activated STATs has also been demonstrated to suppress tumor growth, justifying the development of small-molecule STAT inhibitors to treat human cancers [94,95,96].

Excessive or prolonged activation of the JAK-STAT pathway is prevented by multiple molecules that form a negative feedback loop to regulate the duration and intensity of the pathway. Activated STATs stimulate suppressor of cytokine signaling (SOCS) proteins, which inhibit the further activation of STAT signaling by competing with STATs for binding, ubiquitinating and degrading SOCS substrates, and directly repressing JAK activity [97,98,99]. Protein inhibitors of activated STATs (PIAS) also negatively regulate JAK-STAT signaling by blocking the STAT-DNA interaction, inducing protein SUMOylation, and recruiting transcriptional co-repressors to STAT target genes [100, 101]. In addition, protein tyrosine phosphatases dephosphorylate activated STATs, leading to the inactivation and termination of STAT signaling [102, 103].

Abnormal JAK-STAT pathway in digestive tract tumors

The JAK pathway is aberrantly activated in multiple digestive tract tumors due to mutations in JAKs or STATs, gene fusions with JAKs or STATs, and the restrained expression of negative regulators, ultimately engendering tumor cell malignant behaviors, such as proliferation, invasion, drug resistance, immune escape, and metastasis [3, 104,105,106,107]. A coherent understanding of the aberrant activity of this pathway can help researchers devise therapeutic strategies to decelerate tumor progression [108]. The JAK-STAT signaling pathway has emerged as a potential therapeutic target in gastrointestinal tumor treatment [109,110,111]. One intriguing aspect is the involvement of JAK-STAT signaling in chemotherapy resistance in digestive tract tumors [112, 113]. Emerging evidence underscores the pivotal role of aberrant JAK-STAT pathway activation in conferring resistance to commonly used chemotherapy agents in the clinical management of digestive tract tumors [114,115,116,117]. This aberrant activation has been linked to the upregulation of anti-apoptotic proteins in tumor cells, thereby imparting resistance to chemotherapy-induced apoptosis in digestive tract tumors [118, 119]. Additionally, within the realm of treatment resistance, cancer stem cells (CSCs) have garnered significant attention due to their involvement in tumorigenesis, metastasis, and therapy resistance [120,121,122]. Studies have elucidated that JAK-STAT signaling fosters the stemness properties of CSCs in digestive tract tumors, ultimately contributing to therapy resistance and tumor recurrence [45, 123, 124]. The compelling body of research pointing to the involvement of JAK-STAT signaling in chemotherapy resistance underscores the potential of targeting this pathway as a promising strategy for surmounting treatment obstacles in digestive tract tumors. A comprehensive understanding of the intricate interplay between JAK-STAT signaling, tumor cell apoptosis, and the stemness properties of CSCs will be instrumental in shaping effective therapeutic interventions to combat chemotherapy resistance and improve patient outcomes in the clinical management of digestive tract tumors. Currently, several JAK-STAT inhibitors are available for clinical use in diverse diseases, such as rheumatoid arthritis, myeloproliferative neoplasms, and inflammatory bowel disease [9, 19, 23]. The efficacy of some inhibitors, particularly of digestive tract tumors, is currently being optimized, and combination therapies are being explored to achieve better clinical outcomes [13, 18, 125, 126]. The heterogeneity of different malignant digestive tract tumors and individual variability warrant further in-depth research to determine the efficacy and safety of a particular treatment strategy [127]. Here, we have summarized the carcinogenic mechanisms of dysregulated JAK-STAT signaling in digestive tract tumors, including esophageal, gastric, colorectal, liver, and pancreatic cancer (Fig. 2). We have also presented the efficacy and mechanism of some JAK-STAT inhibitors used for managing digestive tract tumors.

Fig. 2
figure 2

The involvement of the JAK-STAT pathway in digestive tract tumors

Liver cancer

Numerous studies have established that the ubiquitous activation and mutations to the JAK-STAT pathway are essential determinants of tumor development and drug sensitivity in liver cancer (Fig. 3) [128, 129]. Recently, tumor margins have received considerable attention. These regions are known to significantly influence the infiltration and invasion of tumor cells [130,131,132,133]. A detailed assessment of the characteristics and biological properties of tumor margins offers better insights into the development of anti-angiogenesis therapies, tumor invasiveness, and the risk of recurrence [134, 135]. A spatial transcriptomic analysis of liver cancer revealed that JAK-STAT3 signaling, abnormally activated by C-X-C motif chemokine ligand 6, induced the damaged hepatocytes in tumor margins to highly express serum amyloids A1 and A2. This led to macrophage accumulation and M2 polarization, facilitating local immunosuppression and liver cancer progression [136, 137].

Fig. 3
figure 3

The regulatory mechanisms of the JAK-STAT pathway in the progression of liver cancer

Growth hormone (GH) crucially regulates human longitudinal growth, metabolism, and tissue repair by directly or indirectly acting on the liver [138, 139]. Tumor-derived GH has been comprehensively linked with the pathogenesis and progression of various cancers, such as liver, breast, and prostate cancers [140,141,142,143]. Sustained exposure to high levels of GH can cause liver cancer to occur frequently and develop aggressively [144,145,146,147,148]. The loss of STAT5 in liver cells reverses the pathological changes associated with chronic inflammation caused by the overactivation of GH signaling; however, it leads to the earlier occurrence of liver cancer with a more aggressive phenotype. The loss of STAT5 is compensated by the activation of the STAT3 and c-JUN pathways to facilitate the malignant transformation of hepatocytes. This may be attributed to the synthetic actions of lipodystrophy, the deletion of hepatic protective mediators, the activation of the STAT3-c-JUN pathways, and DNA damage [149].

Leptin is a peptide hormone that plays an important role in broad biological processes, including energy metabolism, appetite regulation, and insulin sensitivity [150, 151]. Emerging studies have suggested that abnormalities in leptin levels are correlated with the carcinogenic processes of diverse cancers [152,153,154,155]. Leptin was found to enhance the malignant properties, such as cell invasion and migration potential, of both HepG2 and Huh7 cells by stimulating the JAK-STAT-phosphoinositide 3-kinase-AKT-extracellular signal-regulated kinase (ERK) axis. In the absence of the STAT3 inhibitor AG490, leptin-induced malignant behaviors were notably restrained, further confirming the powerful carcinogenic effect of leptin in liver cancer [156]. Transforming growth factor β1 markedly induced the migration and invasion of liver cancer cells by promoting epithelial-to-mesenchymal transition. It activated JAK-STAT3 signaling and further upregulated Twist in HepG2 cells, whose enhanced migratory and invasive abilities were reversed after AG490 treatment [157].

Hepatitis B virus (HBV) was also found to contribute to cell migration in liver cancer. In vitro experiments showed that HBV rescued the inhibition of cell migration by downregulating miR-340-5p and elevating STAT3 levels [158]. Interferon-alpha (IFN-α) is a well-known treatment option for HBV-induced hepatitis [159,160,161]. Recent studies have reported that sodium butyrate, a differentiation inducer, arrested cell proliferation and strengthened the anti-tumor efficacy of IFN-α in liver cancer by specifically activating STAT1 and enhancing IFN-α-mediated STAT1 expression [162].

The glycine N-methyltransferase (GNMT) gene functions as a tumor susceptibility gene for liver cancer and exhibits a unique tissue expression pattern [163,164,165]. GNMT, which is generally expressed in normal liver tissue, is found to be undetectable in liver cancer and shows attenuated expression in the livers of patients at risk of developing hepatocellular carcinoma [166, 167]. Knocking out GNMT in mice liver activated JAK-STAT pathways, which promoted the malignant transformation of normal liver cells, accompanied by the downregulation of SOCS1, SOCS2, SOCS3, and cytokine-inducible SH2-containing protein and the upregulation of JAK1/2, STAT1, and STAT3 [168].

Eyes absent homolog 2 (EYA2) is considered a tumor suppressor gene in liver cancer, usually exhibiting a pattern of somatic mutations (p.Ala510Glu). Downregulated EYA2 was found to transcriptionally upregulate SOCS3 with the help of dachshund homolog 1. SOCS3 further blocked the JAK2-STAT3 pathway to check the progression of liver cancer [169]. In addition, SH2 domain-containing phosphatase 1, a tumor suppressor of liver cancer, was detected to be markedly downregulated in human liver cancer tissues and associated with poor overall survival. Knocking it down enhanced the activity of the JAK-STAT3 pathway to aggravate hepatocarcinogenesis and exacerbate the malignant phenotype of liver cancer [170]. Akt/β-catenin-driven tumors possess a subtype of side population/CD44 + tumorigenic cells with stem/progenitor-like properties that develop resistance to chemotherapeutic drugs. Targeting the JAK-STAT3 pathway has shown great promise in patients with Akt/β-catenin-driven liver cancer [171].

Gastric cancer

Numerous studies have reported the constitutive activation of STAT3 in gastric cancers as well as its tight association with the prognosis and clinicopathological characteristics of gastric cancer patients [172,173,174]. STAT3 is known to exert its oncogenic effects and regulate various malignant cell behaviors in gastric cancer by interacting with diverse downstream targets [175, 176]. A previous study demonstrated that STAT3 directly upregulated Toll-like receptor 2, an inflammatory mediator, to inhibit epithelial proliferation and anti-apoptosis, thereby enhancing tumorigenesis instead of inflammation in gastric cancer [177]. The integrity of the gastric mucosa is protected by trefoil factor 1 (TFF1), a small cysteine-rich acidic secreted protein that exerts both anti-inflammatory and pro-apoptotic effects [178,179,180]. Recently, a study showed that the loss of TFF1 is responsible for the activation of STAT3. TFF1 impeded the combination of interleukin-6 (IL-6) with IL-6 Rα and further disrupted the activation of STAT3 in the gastric cancer cell lines AGS and STKM2 [181].

Tumor-associated macrophages constitute a large proportion of the infiltrating inflammatory cells in the tumor microenvironment (TME) and display remarkable versatility and plasticity [182,183,184]. Recently, macrophages were identified to secrete CXCL8 under hypoxic conditions, which hyperactivated the JAK-STAT1 pathway in gastric cancer by interacting with C-X-C motif chemokine receptor 1/2 (CXCR1/2) on the cell membrane. Subsequently, IL-10 was overexpressed and M2-type macrophages became polarized, establishing a positive feedback loop between macrophages and gastric cancer progression [185]. Tumor-associated macrophages have also been shown to be tightly associated with stimulator of interferon genes (STING), which is indispensable for regulating the innate and adaptive immune systems. Knocking-down this regulator induced macrophages to differentiate into pro-inflammatory subtypes via the IL-6R-JAK-STAT-IL-24 pathway, thus achieving pro-apoptotic effects in gastric cancer [186].

Dendritic cells function as antigen-presenting cells to dynamically balance the immune response [187,188,189]. Recent flow cytometry results suggest that YTHDF1 knockout recruited dendritic cells and consequently, enhanced the infiltration of T helper cells and cytotoxic T cells in the TME of gastric cancer, promoting the reactivation of adaptive antitumor immunity. YTHDF1 knockout was found to upregulate type I IFN-γ and trigger the JAK-STAT1 pathway to maintain a sustainable systemic antitumor immunity [190].

Additionally, emerging studies have indicated that tumor-infiltrating neutrophils account for the most influential components in the gastric cancer TME and are correlated with poor patient survival [191,192,193]. A novel FasL+PD‐L2+ neutrophil phenotype was discovered in advanced gastric cancer, and these cells exerted immunosuppressive effects in tumor development. Mechanistically, T helper 17 cells secrete IL‐17 A, which subsequently triggers the ERK-nuclear factor κB (NF‐κB) pathway and contributes to the expression of FasL on neutrophils. Tumor‐derived granulocyte colony-stimulating factor markedly activated the JAK-STAT3 pathway and further upregulated programmed cell death-ligand 2 (PD-L2) in neutrophils [194]. Tumor-activated neutrophils also highly expressed PD-L1 and strongly retarded the immunity of normal T cells. Granulocyte macrophage colony-stimulating factor in the TME activated the JAK-STAT3 pathway to upregulate PD-L1 in neutrophils, thus promoting tumor-related immunosuppression and progression [195]. In addition, NF-κB1 polymorphisms were found associated with pro-tumorigenic activity in diverse human cancers, especially digestive tract tumors [196,197,198]. The loss of NF-κB1 in the gastric epithelial and hematopoietic compartments resulted in abnormal gastric inflammation and invasive tumor progression [199]. It also contributed to the overexpression of tumor necrosis factor and STAT1 to further increase the expression of inflammatory effectors and inhibitory immune checkpoint regulators, thereby exacerbating inflammation-associated tumor development [200, 201].

Colorectal cancer

Colorectal cancer is a multifarious disease that involves the dysregulation of multiple signaling pathways, including the JAK-STAT pathway, which regulates the tumor growth, proliferation, migration, and self-renewal characteristics [202,203,204]. Notably, a conspicuous local inflammatory reaction is correlated with improved survival of colorectal cancer patients, whereas an elevated systemic inflammatory response is correlated with worse clinical outcomes [205,206,207,208]. STAT3 levels were found to be especially elevated in stage I-III colorectal cancer patients undergoing surgery, leading to abnormal local and systemic inflammatory responses and poorer prognoses [209]. Numerous studies have shown that the constitutive activation of STAT3 in colorectal cancer drives cell proliferation and tumor growth, thus providing novel insights into treating this disease [210].

Using a transgenic mouse model (∆133p53 isoform) prone to tumors, researchers showed that IL-6 drove the oncogenic activity of the ∆133p53 isoform by upregulating the JAK-STAT3 pathway. Moreover, overexpression of ∆133TP53 mRNA in human colorectal cancers signified a more aggressive tumor phenotype and poorer patient prognosis [211]. The protein tyrosine kinases BMX and HCK were shown to significantly activate the JAK-STAT3 pathway, which promoted the hyperproliferative characteristics of normal epithelial NCM460 cells and initiated adenoma formation in human intestinal organoids. These results contribute to our understanding of adenoma-carcinoma transformation during colorectal carcinogenesis [212].

The circular RNA circSPARC was found upregulated in colorectal cancer, where it served as a competing endogenous RNA to combine with miR-485-3p, thus elevating JAK levels, STAT3 phosphorylation, and STAT3 nuclear translocation. These changes ultimately accelerated tumor growth and metastasis of colorectal cancer [36]. In addition, the long non-coding RNA FEZF1-AS1 was discovered to be overexpressed in colorectal cancer tissues and was associated with poor patient survival. Functional analysis revealed that FEZF1-AS1 upregulated pyruvate kinase 2 to promote aerobic glycolysis and further activate STAT3 signaling. These FEZF1-AS1-induced changes accelerated cell proliferation and metastasis in colorectal cancer [213].

Studies have also explored the regulatory relationship between STAT3 and microRNAs, which ultimately influences tumor oncogenesis [214,215,216]. Elevated miR-572 expression and downregulation of modulator of apoptosis-1 were observed in colorectal cancer with high expression of STAT3. Mechanistically, STAT3 increased miR-572 levels to inhibit the expression of modulator of apoptosis-1, leading to enhanced cell growth, migration, and invasion in colorectal cancer [217]. In addition, PIAS3, a negative regulator of STAT signaling, was found to decrease the expression of miR-18a to restrain the activity of NF-κB and STAT3 in an azoxymethane-dextran sulfate sodium-induced mouse model. The PIAS3-mediated feedback loops exhibited the powerful ability to control cell proliferation in the progression of colitis-associated colorectal cancer, thus offering promising therapeutic targets [218].

Pancreatic cancer

Aberrant stimulation of JAK-STAT signaling also contributes to the oncogenesis of pancreatic cancer [219, 220]. Patients with high STAT3 expression exhibited advanced tumor clinicopathological parameters and worse survival [219]. IL-6 activated STAT3 and increased its phosphorylation, thus upregulating matrix metalloproteinase 2 and vascular endothelial growth factor in the pancreatic cancer line Capan-2. The STAT3-mediated enhanced invasion of Capan-2 cells was counteracted by AG490 [221].

Stellate cells in the TME of pancreatic cancer also secrete IL-6 and drive the activation of the JAK2-STAT3 pathway, which leads to the accumulation of myeloid-derived suppressor cells and the maintenance of an immunosuppressive microenvironment [222]. TEA domain transcription factor 2 was found to upregulate CD109 in the basal-like subtype cells of pancreatic cancer, subsequently hyperactivating the JAK-STAT3 pathway and leading to enhanced metastasis [223]. Pancreatitis was demonstrated to mediate acinar-to-ductal metaplasia and gradually evolve into pancreatic cancer [224].

Numerous studies have validated that the KRAS oncogene is commonly mutated in the early stages of pancreatic cancer [225, 226]. KRAS mutations were found to upregulate the transcriptional regulators yes-associated protein 1 and transcriptional coactivator with PDZ-binding motif to further activate the JAK-STAT3 pathway, thus reprogramming acinar cells and initiating tumorigenesis [227]. Moreover, elevated IL-22 levels during pancreatic tumor development affected the plasticity of acinar cells and induced ductal formation, epithelial-to-mesenchymal transition, and tumor metastasis, all of which were reversed by inhibiting the JAK-STAT3 pathway [228].

IFN-α induced the survival response of human epidermoid cancer cells by hyperactivating the RAS-RAF1-MEK1-ERK1/2 pathway in an epidermal growth factor (EGF)-dependent manner [229, 230]. The activation of peroxisome proliferator-activated receptor γ also enhanced pancreatic cancer cell invasion and migration through diverse mechanisms involving crosstalk with STAT3 [231,232,233,234]. Given these findings, researchers explored the synergistic effect of IFN-β and troglitazone, an agonist of peroxisome proliferator-activated receptor γ, on the growth and autophagy of the pancreatic cancer cell line BxPC-3. IFN-β and troglitazone together exerted a stronger inhibitory influence on STAT3-dependent escape pathways involving the activation of STAT3, mitogen-activated protein kinase, and AKT [235].

Pancreatic cancer patients displayed increased levels of prolactin (PRL). PRL induced the phosphorylation of the JAK2-STA3-ERK-AKT pathway to facilitate the formation of pancospheres and enhance the migratory capacity of cells. These pro-cancer effects of PRL were counteracted by some antipsychotic drugs like penfluridol in pancreatic cancer mouse models [236].

Esophageal cancer

STAT3 plays pivotal roles in esophageal cancer as well. Activated STAT3 acts as an oncogene in esophageal cancer by promoting cell viability, tumor angiogenesis, and metastasis [110, 237,238,239]. Polo-like kinase 1 (PLK1) is preclinically considered a functional regulator in multiple critical cell events during tumor progression [240,241,242]. PLK1 was found overexpressed in esophageal cancer and showed promising prognostic efficacy [243]. Constitutively activated STAT3 and positively regulated PLK1 collectively enhanced proliferation and apoptosis resistance in the esophageal cancer cell line KYSE510 [244].

Similarly, EGF receptor was also shown to augment cell migration in esophageal cancer. The EGF receptor-mediated phosphorylation of STAT1 at Tyr701 led to the formation of the STAT1-STAT3 complex and its translocation into the nucleus. JAK-STAT signaling also upregulated matrix metalloproteinase-1, thereby increasing keratinocyte migration in esophageal cancer [245].

Furthermore, elevated levels of ring finger protein 168 were reported to contribute to malignant cell proliferation and invasion in esophageal cancer. Ring finger protein 168 repressed STAT1 polyubiquitination and degradation to upregulate JAK-STAT1 signaling and the downstream functional genes, contributing to the growth and invasion of esophageal cancer [246]. Small nucleolar RNA host gene 6 was discovered to be markedly upregulated in KYSE150 and KYSE450 cells, and was positively associated with colony formation, migration, tumor malignancy, and 5-fluorouracil resistance in esophageal cancer. It increased the levels of enhancer of zeste homolog 2 to promote STAT3 phosphorylation and H3K27me3 expression, thereby enhancing 5-fluorouracil resistance [247].

Additionally, JAK-STAT signaling pathway plays a crucial role not only through its direct influence on tumor cell survival, proliferation, and therapy resistance, but also through the crosstalk with other signaling pathways [248,249,250]. The interaction between JAK-STAT signaling and other pathways is essential for the regulation of tumor development and progression in digestive tract tumors [251,252,253,254]. One vital cross-interaction in digestive tract tumors involves the interplay between JAK-STAT and PI3K-AKT signaling pathways [255, 256]. The crosstalk between JAK-STAT and PI3K-AKT pathways creates a positive feedback loop that amplifies tumor-promoting signals and contributes to tumor growth and therapeutic resistance in digestive tract tumors [257, 258]. In addition to the PI3K-AKT pathway, JAK-STAT signaling also interacts with other major signaling pathways, such as the MAPK-ERK pathway, contributing to the aggressive phenotype and therapy resistance observed in digestive tract tumors [259]. The cross-talk between JAK-STAT signaling and other pathways highlights the importance of a comprehensive understanding of the intricate network of molecular interactions in cancer progression and therapy response of digestive tract tumors. Targeting multiple signaling pathways simultaneously may be a promising approach to overcome therapy resistance and improve patient outcomes in digestive tract tumors.

Conclusion and prospects

In conclusion, the JAK-STAT pathway has emerged as a crucial factor in the pathogenesis of digestive tract tumors. Its aberrant activation, triggered by pro-inflammatory cytokines, disrupts various biological processes such as cell growth, apoptosis, and migration. The current understanding of the classic activation and regulation of the JAK-STAT pathway has provided a foundation for identifying potential therapeutic targets for digestive tract tumors. However, recent research has shown that the use of JAK inhibitors raises safety concerns due to their lack of specificity, as they inhibit multiple signal transduction pathways. Therefore, careful monitoring and management of infection complications is imperative when administering JAK inhibitors to treat digestive tract tumors.

To fully understand the mechanisms underlying the JAK-STAT pathway in digestive tract tumors, future research should focus on gaining a better understanding of the interplay between this pathway and other signaling pathways. Additionally, it is important to identify specific molecular targets within the JAK-STAT pathway that can be selectively modulated to achieve maximal therapeutic benefit. Recent preclinical and clinical trials have shown promising results with drugs targeting this pathway; however, striking a balance between efficacy and safety remains a challenge.

In summary, a better understanding of the JAK-STAT pathway in digestive tract tumors will pave the way for the development of targeted therapies that are both safe and effective. Further research is needed to fully elucidate the mechanisms involved in the dysregulation of this pathway and to optimize therapeutic strategies for the treatment of digestive tract tumors Tables 1 and 2.

Table 1 Expression and outcomes of the JAK–STAT pathway in digestive tract tumors
Table 2 The roles and mechanisms of the JAK–STAT pathway in digestive tract tumors

Availability of data and materials

Not applicable.

References

  1. Furqan M, Mukhi N, Lee B, Liu D. Dysregulation of JAK-STAT pathway in hematological malignancies and JAK inhibitors for clinical application. Biomark Res. 2013;1:5.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Hou SX, Zheng Z, Chen X, Perrimon N. The Jak/STAT pathway in model organisms: emerging roles in cell movement. Dev Cell. 2002;3:765–78.

    Article  CAS  PubMed  Google Scholar 

  3. Baldini C, Moriconi FR, Galimberti S, Libby P, De Caterina R. The JAK-STAT pathway: an emerging target for Cardiovascular Disease in rheumatoid arthritis and myeloproliferative Neoplasms. Eur Heart J. 2021;42:4389–400.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Jere SW, Abrahamse H, Houreld NN. The JAK/STAT signaling pathway and photobiomodulation in chronic wound healing. Cytokine Growth Factor Rev. 2017;38:73–9.

    Article  CAS  PubMed  Google Scholar 

  5. Lai SY, Johnson FM. Defining the role of the JAK-STAT pathway in head and neck and thoracic malignancies: implications for future therapeutic approaches. Drug Resist Updat. 2010;13:67–78.

    Article  CAS  PubMed  Google Scholar 

  6. Waldmann TA, Chen J. Disorders of the JAK/STAT pathway in T cell Lymphoma pathogenesis: implications for Immunotherapy. Annu Rev Immunol. 2017;35:533–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Owen KL, Brockwell NK, Parker BS. JAK-STAT signaling: a double-edged Sword of Immune Regulation and Cancer Progression. Cancers (Basel). 2019;11:2002.

  8. Darnell JE Jr., Kerr IM, Stark GR. Jak-STAT pathways and transcriptional activation in response to IFNs and other extracellular signaling proteins. Science. 1994;264:1415–21.

    Article  CAS  PubMed  Google Scholar 

  9. Coskun M, Salem M, Pedersen J, Nielsen OH. Involvement of JAK/STAT signaling in the pathogenesis of inflammatory bowel Disease. Pharmacol Res. 2013;76:1–8.

    Article  CAS  PubMed  Google Scholar 

  10. Xue C, Yao Q, Gu X, Shi Q, Yuan X, Chu Q, Bao Z, Lu J, Li L. Evolving cognition of the JAK-STAT signaling pathway: autoimmune disorders and cancer. Signal Transduct Target Ther. 2023;8:204.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Beckman JD, DaSilva A, Aronovich E, Nguyen A, Nguyen J, Hargis G, Reynolds D, Vercellotti GM, Betts B, Wood DK. JAK-STAT inhibition reduces endothelial prothrombotic activation and leukocyte-endothelial proadhesive interactions. J Thromb Haemost. 2023;21:1366–80.

    Article  PubMed  Google Scholar 

  12. Souza-Neto JA, Sim S, Dimopoulos G. An evolutionary conserved function of the JAK-STAT pathway in anti-dengue defense. Proc Natl Acad Sci U S A. 2009;106:17841–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Cai Z, Zhang S, Wu P, Ren Q, Wei P, Hong M, Feng Y, Wong CK, Tang H, Zeng H. A novel potential target of IL-35-regulated JAK/STAT signaling pathway in lupus Nephritis. Clin Transl Med. 2021;11:e309.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Iznardo H, Roé E, Serra-Baldrich E, Puig L. Efficacy and safety of JAK1 inhibitor abrocitinib in atopic dermatitis. Pharmaceutics. 2023;15.

  15. Banerjee S, Biehl A, Gadina M, Hasni S, Schwartz DM. JAK-STAT signaling as a target for inflammatory and autoimmune Diseases: current and future prospects. Drugs. 2017;77:521–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Barry SP, Townsend PA, Latchman DS, Stephanou A. Role of the JAK-STAT pathway in myocardial injury. Trends Mol Med. 2007;13:82–9.

    Article  CAS  PubMed  Google Scholar 

  17. Qi F, Liu F, Gao L. Janus Kinase Inhibitors in the treatment of Vitiligo: a review. Front Immunol. 2021;12:790125.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Goker Bagca B, Biray Avci C. The potential of JAK/STAT pathway inhibition by ruxolitinib in the treatment of COVID-19. Cytokine Growth Factor Rev. 2020;54:51–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Favoino E, Prete M, Catacchio G, Ruscitti P, Navarini L, Giacomelli R, Perosa F. Working and safety profiles of JAK/STAT signaling inhibitors. Are these small molecules also smart? Autoimmun Rev. 2021;20:102750.

    Article  CAS  PubMed  Google Scholar 

  20. Thomas S, Fisher K, Snowden J, Danson S, Brown S, Zeidler M. Effect of methotrexate on JAK/STAT pathway activation in myeloproliferative Neoplasms. Lancet. 2015;385(Suppl 1):98.

    Article  Google Scholar 

  21. Solimani F, Meier K, Ghoreschi K. Emerging topical and systemic JAK inhibitors in Dermatology. Front Immunol. 2019;10:2847.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Damsky W, King BA. JAK inhibitors in dermatology: the promise of a new drug class. J Am Acad Dermatol. 2017;76:736–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Springuel L, Renauld JC, Knoops L. JAK kinase targeting in hematologic malignancies: a sinuous pathway from identification of genetic alterations towards clinical indications. Haematologica. 2015;100:1240–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Patel NM, Collotta D, Aimaretti E, Ferreira Alves G, Kröller S, Coldewey SM, Collino M, Thiemermann C. Inhibition of the JAK/STAT pathway with Baricitinib reduces the multiple organ dysfunction caused by hemorrhagic shock in rats. Ann Surg. 2023;278:e137–46.

    Article  PubMed  Google Scholar 

  25. Liang X, Liu H, Zhang Y. Novel-targeted therapy for hematological malignancies with JAK and HDAC dual inhibitors. Future Med Chem. 2019;11:1849–52.

    Article  CAS  PubMed  Google Scholar 

  26. Nagtegaal ID, Odze RD, Klimstra D, Paradis V, Rugge M, Schirmacher P, Washington KM, Carneiro F, Cree IA. The 2019 WHO classification of tumours of the digestive system. Histopathology. 2020;76:182–8.

    Article  PubMed  Google Scholar 

  27. Gonzalez RS, Raza A, Propst R, Adeyi O, Bateman J, Sopha SC, Shaw J, Auerbach A. Recent advances in Digestive Tract tumors: updates from the 5th Edition of the World Health Organization Blue Book. Arch Pathol Lab Med. 2021;145:607–26.

    Article  CAS  PubMed  Google Scholar 

  28. Tullio V, Gasperi V, Catani MV, Savini I. The impact of whole grain intake on gastrointestinal tumors: a focus on colorectal, gastric, and esophageal cancers. Nutrients. 2020;13.

  29. Arnold M, Sierra MS, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global patterns and trends in Colorectal cancer incidence and mortality. Gut. 2017;66:683–91.

    Article  PubMed  Google Scholar 

  30. Wagner A, Zandanell S, Kiesslich T, Neureiter D, Klieser E, Holzinger J, Berr F. Systematic review on Optical diagnosis of early gastrointestinal neoplasia. J Clin Med. 2021;10.

  31. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global Cancer statistics 2020: GLOBOCAN estimates of incidence and Mortality Worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71:209–49.

    Article  PubMed  Google Scholar 

  32. Arnold M, Abnet CC, Neale RE, Vignat J, Giovannucci EL, McGlynn KA, Bray F. Global burden of 5 major types of gastrointestinal Cancer. Gastroenterology. 2020;159:335–349e315.

    Article  PubMed  Google Scholar 

  33. Choi EL, Taheri N, Chandra A, Hayashi Y. Cellular Senescence, inflammation, and Cancer in the gastrointestinal tract. Int J Mol Sci. 2023;24.

  34. Zhang Y, Yang C, Cheng H, Fan Z, Huang Q, Lu Y, Fan K, Luo G, Jin K, Wang Z, et al. Novel agents for pancreatic ductal adenocarcinoma: emerging therapeutics and future directions. J Hematol Oncol. 2018;11:14.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Woo J, Lim JW, Kim H. Astaxanthin inhibits integrin α5 expression by suppressing activation of JAK1/STAT3 in Helicobacter pylori–stimulated gastric epithelial cells. Mol Med Rep. 2023;27.

  36. Wang J, Zhang Y, Song H, Yin H, Jiang T, Xu Y, Liu L, Wang H, Gao H, Wang R, Song J. The circular RNA circSPARC enhances the migration and proliferation of Colorectal cancer by regulating the JAK/STAT pathway. Mol Cancer. 2021;20:81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Trivedi S, Starz-Gaiano M. Drosophila Jak/STAT signaling: regulation and relevance in Human Cancer and Metastasis. Int J Mol Sci. 2018;19.

  38. Valle-Mendiola A, Gutiérrez-Hoya A, Soto-Cruz I. JAK/STAT signaling and Cervical Cancer: from the cell surface to the Nucleus. Genes (Basel). 2023;14.

  39. Cho HJ, Park JH, Nam JH, Chang YC, Park B, Hoe HS. Ascochlorin suppresses MMP-2-Mediated Migration and Invasion by targeting FAK and JAK-STAT signaling cascades. J Cell Biochem. 2018;119:300–13.

    Article  CAS  PubMed  Google Scholar 

  40. Rajakumar T, Pugalendhi P. Allyl Isothiocyanate inhibits invasion and angiogenesis in Breast cancer via EGFR-mediated JAK-1/STAT-3 signaling pathway. Amino Acids. 2023;55:981–92.

    Article  CAS  PubMed  Google Scholar 

  41. Xu CH, Liu Y, Xiao LM, Chen LK, Zheng SY, Zeng EM, Li DH, Li YP. Silencing microRNA-221/222 cluster suppresses glioblastoma angiogenesis by suppressor of cytokine signaling-3-dependent JAK/STAT pathway. J Cell Physiol. 2019;234:22272–84.

    Article  CAS  PubMed  Google Scholar 

  42. Yin J, Li Z, Ye L, Birkin E, Li L, Xu R, Chen G, Ji J, Zhang Z, Jiang WG, Cui Y. EphB2 represents an Independent prognostic marker in patients with gastric cancer and promotes tumour cell aggressiveness. J Cancer. 2020;11:2778–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Krzysiek-Maczka G, Targosz A, Szczyrk U, Strzalka M, Brzozowski T, Ptak-Belowska A. Involvement of epithelial-mesenchymal transition-inducing transcription factors in the mechanism of Helicobacter pylori-induced fibroblasts activation. J Physiol Pharmacol. 2019;70.

  44. Shen M, Xu Z, Xu W, Jiang K, Zhang F, Ding Q, Xu Z, Chen Y. Inhibition of ATM reverses EMT and decreases metastatic potential of cisplatin-resistant Lung cancer cells through JAK/STAT3/PD-L1 pathway. J Exp Clin Cancer Res. 2019;38:149.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Jin W. Role of JAK/STAT3 signaling in the regulation of Metastasis, the transition of Cancer Stem cells, and Chemoresistance of Cancer by epithelial-mesenchymal transition. Cells. 2020;9.

  46. Davies SC, Hussein IM, Nguyen TM, Parker CE, Khanna R, Jairath V. Oral Janus kinase inhibitors for maintenance of remission in ulcerative Colitis. Cochrane Database Syst Rev. 2020;1:Cd012381.

    PubMed  Google Scholar 

  47. Kim BH, Jee JG, Yin CH, Sandoval C, Jayabose S, Kitamura D, Bach EA, Baeg GH. NSC114792, a novel small molecule identified through structure-based computational database screening, selectively inhibits JAK3. Mol Cancer. 2010;9:36.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Yamaoka K, Saharinen P, Pesu M, Holt VE 3rd, Silvennoinen O, O’Shea JJ. The Janus kinases (Jaks). Genome Biol. 2004;5:253.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Leonard WJ, O’Shea JJ. Jaks and STATs: biological implications. Annu Rev Immunol. 1998;16:293–322.

    Article  CAS  PubMed  Google Scholar 

  50. Sayyah J, Gnanasambandan K, Kamarajugadda S, Tsuda S, Caldwell-Busby J, Sayeski PP. Phosphorylation of Y372 is critical for Jak2 tyrosine kinase activation. Cell Signal. 2011;23:1806–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Shawky AM, Almalki FA, Abdalla AN, Abdelazeem AH, Gouda AM. A comprehensive overview of globally approved JAK inhibitors. Pharmaceutics. 2022;14.

  52. Manning G, Whyte DB, Martinez R, Hunter T, Sudarsanam S. The protein kinase complement of the human genome. Science. 2002;298:1912–34.

    Article  CAS  PubMed  Google Scholar 

  53. Pérez-Jeldres T, Tyler CJ, Boyer JD, Karuppuchamy T, Yarur A, Giles DA, Yeasmin S, Lundborg L, Sandborn WJ, Patel DR, Rivera-Nieves J. Targeting Cytokine Signaling and Lymphocyte Traffic via Small molecules in Inflammatory Bowel Disease: JAK inhibitors and S1PR agonists. Front Pharmacol. 2019;10:212.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Liau NPD, Laktyushin A, Lucet IS, Murphy JM, Yao S, Whitlock E, Callaghan K, Nicola NA, Kershaw NJ, Babon JJ. The molecular basis of JAK/STAT inhibition by SOCS1. Nat Commun. 2018;9:1558.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Maude SL, Dolai S, Delgado-Martin C, Vincent T, Robbins A, Selvanathan A, Ryan T, Hall J, Wood AC, Tasian SK, et al. Efficacy of JAK/STAT pathway inhibition in murine xenograft models of early T-cell precursor (ETP) acute lymphoblastic Leukemia. Blood. 2015;125:1759–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Saharinen P, Takaluoma K, Silvennoinen O. Regulation of the Jak2 tyrosine kinase by its pseudokinase domain. Mol Cell Biol. 2000;20:3387–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Saharinen P, Silvennoinen O. The pseudokinase domain is required for suppression of basal activity of Jak2 and Jak3 tyrosine kinases and for cytokine-inducible activation of signal transduction. J Biol Chem. 2002;277:47954–63.

    Article  CAS  PubMed  Google Scholar 

  58. Wallweber HJ, Tam C, Franke Y, Starovasnik MA, Lupardus PJ. Structural basis of recognition of interferon-α receptor by tyrosine kinase 2. Nat Struct Mol Biol. 2014;21:443–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Radtke S, Haan S, Jörissen A, Hermanns HM, Diefenbach S, Smyczek T, Schmitz-Vandeleur H, Heinrich PC, Behrmann I, Haan C. The Jak1 SH2 domain does not fulfill a classical SH2 function in Jak/STAT signaling but plays a structural role for receptor interaction and up-regulation of receptor surface expression. J Biol Chem. 2005;280:25760–8.

    Article  CAS  PubMed  Google Scholar 

  60. Padmasuta K, Sangarlangkarn S, Bunyaratvej A. Evaluation of halothane effect on blood coagulation and bleeding time: a study of 129 cases with elective operations. J Med Assoc Thai. 1987;70:261–4.

    CAS  PubMed  Google Scholar 

  61. Gordon GM, Lambert QT, Daniel KG, Reuther GW. Transforming JAK1 mutations exhibit differential signalling, FERM domain requirements and growth responses to interferon-γ. Biochem J. 2010;432:255–65.

    Article  CAS  PubMed  Google Scholar 

  62. Ferrao R, Lupardus PJ. The Janus kinase (JAK) FERM and SH2 domains: bringing specificity to JAK-Receptor interactions. Front Endocrinol (Lausanne). 2017;8:71.

    Article  PubMed  Google Scholar 

  63. Zhou YJ, Chen M, Cusack NA, Kimmel LH, Magnuson KS, Boyd JG, Lin W, Roberts JL, Lengi A, Buckley RH, et al. Unexpected effects of FERM domain mutations on catalytic activity of Jak3: structural implication for Janus kinases. Mol Cell. 2001;8:959–69.

    Article  CAS  PubMed  Google Scholar 

  64. Haan S, Margue C, Engrand A, Rolvering C, Schmitz-Van de Leur H, Heinrich PC, Behrmann I, Haan C. Dual role of the Jak1 FERM and kinase domains in cytokine receptor binding and in stimulation-dependent jak activation. J Immunol. 2008;180:998–1007.

    Article  CAS  PubMed  Google Scholar 

  65. Garrido-Trigo A, Salas A. Molecular structure and function of Janus Kinases: implications for the development of inhibitors. J Crohns Colitis. 2020;14:713–s724.

    Article  Google Scholar 

  66. Leonard WJ, Lin JX. Strategies to therapeutically modulate cytokine action. Nat Rev Drug Discov. 2023;22:827–54.

    Article  CAS  PubMed  Google Scholar 

  67. Ott N, Faletti L, Heeg M, Andreani V, Grimbacher B. JAKs and STATs from a clinical perspective: loss-of-function mutations, Gain-of-function mutations, and their multidimensional consequences. J Clin Immunol. 2023;43:1326–59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Strobl B, Stoiber D, Sexl V, Mueller M. Tyrosine kinase 2 (TYK2) in cytokine signalling and host immunity. Front Biosci (Landmark Ed). 2011;16:3214–32.

    Article  PubMed  Google Scholar 

  69. Min X, Ungureanu D, Maxwell S, Hammarén H, Thibault S, Hillert EK, Ayres M, Greenfield B, Eksterowicz J, Gabel C, et al. Structural and functional characterization of the JH2 pseudokinase domain of JAK family tyrosine kinase 2 (TYK2). J Biol Chem. 2015;290:27261–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Russell SM, Tayebi N, Nakajima H, Riedy MC, Roberts JL, Aman MJ, Migone TS, Noguchi M, Markert ML, Buckley RH, et al. Mutation of Jak3 in a patient with SCID: essential role of Jak3 in lymphoid development. Science. 1995;270:797–800.

    Article  CAS  PubMed  Google Scholar 

  71. Villa A, Sironi M, Macchi P, Matteucci C, Notarangelo LD, Vezzoni P, Mantovani A. Monocyte function in a severe combined immunodeficient patient with a donor splice site mutation in the Jak3 gene. Blood. 1996;88:817–23.

    Article  CAS  PubMed  Google Scholar 

  72. Hernandez-Rocha C, Vande Casteele N. JAK inhibitors: current position in treatment strategies for use in inflammatory bowel Disease. Curr Opin Pharmacol. 2020;55:99–109.

    Article  CAS  PubMed  Google Scholar 

  73. Jayavelu AK, Schnöder TM, Perner F, Herzog C, Meiler A, Krishnamoorthy G, Huber N, Mohr J, Edelmann-Stephan B, Austin R, et al. Splicing factor YBX1 mediates persistence of JAK2-mutated Neoplasms. Nature. 2020;588:157–63.

    Article  PubMed  Google Scholar 

  74. Ashino S, Takeda K, Li H, Taylor V, Joetham A, Pine PR, Gelfand EW. Janus kinase 1/3 signaling pathways are key initiators of TH2 differentiation and lung allergic responses. J Allergy Clin Immunol. 2014;133:1162–74.

    Article  CAS  PubMed  Google Scholar 

  75. Wehde BL, Rädler PD, Shrestha H, Johnson SJ, Triplett AA, Wagner KU. Janus Kinase 1 plays a critical role in Mammary Cancer Progression. Cell Rep. 2018;25:2192–2207e2195.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Harigai M, Honda S. Selectivity of Janus Kinase Inhibitors in rheumatoid arthritis and other Immune-mediated inflammatory Diseases: is expectation the Root of all Headache? Drugs. 2020;80:1183–201.

  77. O’Shea JJ, Schwartz DM, Villarino AV, Gadina M, McInnes IB, Laurence A. The JAK-STAT pathway: impact on human Disease and therapeutic intervention. Annu Rev Med. 2015;66:311–28.

    Article  PubMed  PubMed Central  Google Scholar 

  78. Akira S, Nishio Y, Inoue M, Wang XJ, Wei S, Matsusaka T, Yoshida K, Sudo T, Naruto M, Kishimoto T. Molecular cloning of APRF, a novel IFN-stimulated gene factor 3 p91-related transcription factor involved in the gp130-mediated signaling pathway. Cell. 1994;77:63–71.

    Article  CAS  PubMed  Google Scholar 

  79. Darnell JE. Jr.: STATs and gene regulation. Science. 1997;277:1630–5.

    Article  CAS  PubMed  Google Scholar 

  80. Arnould C, Philippe C, Bourdon V, Gr goire MJ, Berger R, Jonveaux P. The signal transducer and activator of transcription STAT5b gene is a new partner of retinoic acid receptor alpha in acute promyelocytic-like Leukaemia. Hum Mol Genet. 1999;8:1741–9.

    Article  CAS  PubMed  Google Scholar 

  81. Shirakawa T, Kawazoe Y, Tsujikawa T, Jung D, Sato S, Uesugi M. Deactivation of STAT6 through serine 707 phosphorylation by JNK. J Biol Chem. 2011;286:4003–10.

    Article  CAS  PubMed  Google Scholar 

  82. Pace J, Paladugu P, Das B, He JC, Mallipattu SK. Targeting STAT3 signaling in Kidney Disease. Am J Physiol Renal Physiol. 2019;316:F1151–f1161.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Pesu M, Takaluoma K, Aittomäki S, Lagerstedt A, Saksela K, Kovanen PE, Silvennoinen O. Interleukin-4-induced transcriptional activation by stat6 involves multiple serine/threonine kinase pathways and serine phosphorylation of stat6. Blood. 2000;95:494–502.

    Article  CAS  PubMed  Google Scholar 

  84. Blaszczyk K, Nowicka H, Kostyrko K, Antonczyk A, Wesoly J, Bluyssen HA. The unique role of STAT2 in constitutive and IFN-induced transcription and antiviral responses. Cytokine Growth Factor Rev. 2016;29:71–81.

    Article  CAS  PubMed  Google Scholar 

  85. Begitt A, Meyer T, van Rossum M, Vinkemeier U. Nucleocytoplasmic translocation of Stat1 is regulated by a leucine-rich export signal in the coiled-coil domain. Proc Natl Acad Sci U S A. 2000;97:10418–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Paukku K, Silvennoinen O. STATs as critical mediators of signal transduction and transcription: lessons learned from STAT5. Cytokine Growth Factor Rev. 2004;15:435–55.

    Article  CAS  PubMed  Google Scholar 

  87. Ginger RS, Dalton EC, Ryves WJ, Fukuzawa M, Williams JG, Harwood AJ. Glycogen synthase kinase-3 enhances nuclear export of a Dictyostelium STAT protein. Embo J. 2000;19:5483–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Gupta S, Yan H, Wong LH, Ralph S, Krolewski J, Schindler C. The SH2 domains of Stat1 and Stat2 mediate multiple interactions in the transduction of IFN-alpha signals. Embo J. 1996;15:1075–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Palakurthi B, Fross SR, Guldner IH, Aleksandrovic E, Liu X, Martino AK, Wang Q, Neff RA, Golomb SM, Lewis C, et al. Targeting CXCL16 and STAT1 augments immune checkpoint blockade therapy in triple-negative Breast cancer. Nat Commun. 2023;14:2109.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Steen HC, Kotredes KP, Nogusa S, Harris MY, Balachandran S, Gamero AM. Phosphorylation of STAT2 on serine-734 negatively regulates the IFN-α-induced antiviral response. J Cell Sci. 2016;129:4190–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Bowman T, Garcia R, Turkson J, Jove R. STATs in oncogenesis. Oncogene. 2000;19:2474–88.

    Article  CAS  PubMed  Google Scholar 

  92. Mora LB, Buettner R, Seigne J, Diaz J, Ahmad N, Garcia R, Bowman T, Falcone R, Fairclough R, Cantor A, et al. Constitutive activation of Stat3 in human prostate tumors and cell lines: direct inhibition of Stat3 signaling induces apoptosis of Prostate cancer cells. Cancer Res. 2002;62:6659–66.

    CAS  PubMed  Google Scholar 

  93. Huang S. Regulation of metastases by signal transducer and activator of transcription 3 signaling pathway: clinical implications. Clin Cancer Res. 2007;13:1362–6.

    Article  CAS  PubMed  Google Scholar 

  94. Buettner R, Mora LB, Jove R. Activated STAT signaling in human tumors provides novel molecular targets for therapeutic intervention. Clin Cancer Res. 2002;8:945–54.

    CAS  PubMed  Google Scholar 

  95. Turkson J, Jove R. STAT proteins: novel molecular targets for cancer drug discovery. Oncogene. 2000;19:6613–26.

    Article  CAS  PubMed  Google Scholar 

  96. Turkson J. STAT proteins as novel targets for cancer drug discovery. Expert Opin Ther Targets. 2004;8:409–22.

    Article  CAS  PubMed  Google Scholar 

  97. Alexander WS. Suppressors of cytokine signalling (SOCS) in the immune system. Nat Rev Immunol. 2002;2:410–6.

    Article  CAS  PubMed  Google Scholar 

  98. Yoshimura A, Naka T, Kubo M. SOCS proteins, cytokine signalling and immune regulation. Nat Rev Immunol. 2007;7:454–65.

    Article  CAS  PubMed  Google Scholar 

  99. Alexander WS, Hilton DJ. The role of suppressors of cytokine signaling (SOCS) proteins in regulation of the immune response. Annu Rev Immunol. 2004;22:503–29.

    Article  CAS  PubMed  Google Scholar 

  100. Heppler LN, Frank DA. Targeting oncogenic transcription factors: therapeutic implications of endogenous STAT inhibitors. Trends Cancer. 2017;3:816–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Yagil Z, Nechushtan H, Kay G, Yang CM, Kemeny DM, Razin E. The enigma of the role of protein inhibitor of activated STAT3 (PIAS3) in the immune response. Trends Immunol. 2010;31:199–204.

    Article  CAS  PubMed  Google Scholar 

  102. Nakahira M, Tanaka T, Robson BE, Mizgerd JP, Grusby MJ. Regulation of signal transducer and activator of transcription signaling by the tyrosine phosphatase PTP-BL. Immunity. 2007;26:163–76.

    Article  CAS  PubMed  Google Scholar 

  103. Gurzov EN, Tran M, Fernandez-Rojo MA, Merry TL, Zhang X, Xu Y, Fukushima A, Waters MJ, Watt MJ, Andrikopoulos S, et al. Hepatic oxidative stress promotes insulin-STAT-5 signaling and obesity by inactivating protein tyrosine phosphatase N2. Cell Metab. 2014;20:85–102.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Liu X, Nagy P, Bonfini A, Houtz P, Bing XL, Yang X, Buchon N. Microbes affect gut epithelial cell composition through immune-dependent regulation of intestinal stem cell differentiation. Cell Rep. 2022;38:110572.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Ni Y, Low JT, Silke J, O’Reilly LA. Digesting the role of JAK-STAT and Cytokine Signaling in oral and gastric cancers. Front Immunol. 2022;13:835997.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Gonneaud A, Turgeon N, Boisvert FM, Boudreau F, Asselin C. JAK-STAT pathway inhibition partially restores intestinal homeostasis in Hdac1- and Hdac2-Intestinal epithelial cell-deficient mice. Cells. 2021;10.

  107. Erdogan F, Radu TB, Orlova A, Qadree AK, de Araujo ED, Israelian J, Valent P, Mustjoki SM, Herling M, Moriggl R, Gunning PT. JAK-STAT core cancer pathway: an integrative cancer interactome analysis. J Cell Mol Med. 2022;26:2049–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Wang L, Hu Y, Song B, Xiong Y, Wang J, Chen D. Targeting JAK/STAT signaling pathways in treatment of inflammatory bowel Disease. Inflamm Res. 2021;70:753–64.

    Article  CAS  PubMed  Google Scholar 

  109. Wang M, Sun X, Xin H, Wen Z, Cheng Y. SPP1 promotes radiation resistance through JAK2/STAT3 pathway in esophageal carcinoma. Cancer Med. 2022;11:4526–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Jia Z, Xie Y, Wu H, Wang Z, Li A, Li Z, Yang Z, Zhang Z, Xing Z, Zhang X. Phlorizin from sweet tea inhibits the progress of Esophageal cancer by antagonizing the JAK2/STAT3 signaling pathway. Oncol Rep. 2021;46.

  111. Joshi N, Hajizadeh F, Ansari Dezfouli E, Zekiy AO, Nabi Afjadi M, Mousavi SM, Hojjat-Farsangi M, Karpisheh V, Mahmoodpoor A, Hassannia H, et al. Silencing STAT3 enhances sensitivity of cancer cells to doxorubicin and inhibits Tumor progression. Life Sci. 2021;275:119369.

    Article  CAS  PubMed  Google Scholar 

  112. Singh S, Gomez HJ, Thakkar S, Singh SP, Parihar AS. Overcoming Acquired Drug Resistance to Cancer therapies through targeted STAT3 inhibition. Int J Mol Sci. 2023;24.

  113. Nagaraju GP, Mezina A, Shaib WL, Landry J, El-Rayes BF. Targeting the Janus-activated kinase-2-STAT3 signalling pathway in Pancreatic cancer using the HSP90 inhibitor ganetespib. Eur J Cancer. 2016;52:109–19.

    Article  CAS  PubMed  Google Scholar 

  114. Zhang Z, Wang F, Du C, Guo H, Ma L, Liu X, Kornmann M, Tian X, Yang Y. BRM/SMARCA2 promotes the proliferation and chemoresistance of Pancreatic cancer cells by targeting JAK2/STAT3 signaling. Cancer Lett. 2017;402:213–24.

    Article  CAS  PubMed  Google Scholar 

  115. Venkatasubbarao K, Peterson L, Zhao S, Hill P, Cao L, Zhou Q, Nawrocki ST, Freeman JW. Inhibiting signal transducer and activator of transcription-3 increases response to gemcitabine and delays progression of Pancreatic cancer. Mol Cancer. 2013;12:104.

    Article  PubMed  PubMed Central  Google Scholar 

  116. Khatoon E, Hegde M, Kumar A, Daimary UD, Sethi G, Bishayee A, Kunnumakkara AB. The multifaceted role of STAT3 pathway and its implication as a potential therapeutic target in Oral cancer. Arch Pharm Res. 2022;45:507–34.

    Article  CAS  PubMed  Google Scholar 

  117. Ouyang S, Li H, Lou L, Huang Q, Zhang Z, Mo J, Li M, Lu J, Zhu K, Chu Y, et al. Inhibition of STAT3-ferroptosis negative regulatory axis suppresses Tumor growth and alleviates chemoresistance in gastric cancer. Redox Biol. 2022;52:102317.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Du XL, Yang H, Liu SG, Luo ML, Hao JJ, Zhang Y, Lin DC, Xu X, Cai Y, Zhan QM, Wang MR. Calreticulin promotes cell motility and enhances resistance to anoikis through STAT3-CTTN-Akt pathway in esophageal squamous cell carcinoma. Oncogene. 2009;28:3714–22.

    Article  CAS  PubMed  Google Scholar 

  119. Fofaria NM, Srivastava SK. STAT3 induces anoikis resistance, promotes cell invasion and metastatic potential in Pancreatic cancer cells. Carcinogenesis. 2015;36:142–50.

    Article  CAS  PubMed  Google Scholar 

  120. Nassar D, Blanpain C. Cancer Stem cells: Basic concepts and therapeutic implications. Annu Rev Pathol. 2016;11:47–76.

    Article  CAS  PubMed  Google Scholar 

  121. Relier S, Ripoll J, Guillorit H, Amalric A, Achour C, Boissière F, Vialaret J, Attina A, Debart F, Choquet A, et al. FTO-mediated cytoplasmic m(6)A(m) demethylation adjusts stem-like properties in Colorectal cancer cell. Nat Commun. 2021;12:1716.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Vlashi E, Pajonk F. Cancer stem cells, cancer cell plasticity and radiation therapy. Semin Cancer Biol. 2015;31:28–35.

    Article  CAS  PubMed  Google Scholar 

  123. Zheng H, Liu H, Li H, Dou W, Wang J, Zhang J, Liu T, Wu Y, Liu Y, Wang X. Characterization of stem cell landscape and identification of stemness-relevant prognostic gene signature to aid immunotherapy in Colorectal cancer. Stem Cell Res Ther. 2022;13:244.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Khan AQ, Ahmed EI, Elareer NR, Junejo K, Steinhoff M, Uddin S. Role of miRNA-Regulated Cancer Stem cells in the Pathogenesis of Human malignancies. Cells. 2019;8.

  125. Lokau J, Schoeder V, Haybaeck J, Garbers C. Jak-Stat Signaling Induced by Interleukin-6 family cytokines in Hepatocellular Carcinoma. Cancers (Basel) 2019;11.

  126. Guo H, Zhang C, Tang X, Zhang T, Liu Y, Yu H, Li Y, Wang R. HHLA2 activates the JAK/STAT signaling pathway by binding to TMIGD2 in Hepatocellular Carcinoma cells. Inflammation. 2022;45:1585–99.

    Article  CAS  PubMed  Google Scholar 

  127. Yue Y, Zhang Q, Wu S, Wang S, Cui C, Yu M, Sun Z. Identification of key genes involved in JAK/STAT pathway in Colorectal cancer. Mol Immunol. 2020;128:287–97.

    Article  CAS  PubMed  Google Scholar 

  128. Ding X, He M, Chan AWH, Song QX, Sze SC, Chen H, Man MKH, Man K, Chan SL, Lai PBS, et al. Genomic and Epigenomic Features of Primary and Recurrent Hepatocellular Carcinomas. Gastroenterology. 2020;57:1630–45.

    Google Scholar 

  129. Calvisi DF, Ladu S, Gorden A, Farina M, Conner EA, Lee JS, Factor VM, Thorgeirsson SS. Ubiquitous activation of Ras and Jak/Stat pathways in human HCC. Gastroenterology. 2006;130:1117–28.

    Article  CAS  PubMed  Google Scholar 

  130. Sperb N, Tsesmelis M, Wirth T. Crosstalk between Tumor and Stromal cells in pancreatic ductal adenocarcinoma. Int J Mol Sci. 2020;21.

  131. Lin W, Noel P, Borazanci EH, Lee J, Amini A, Han IW, Heo JS, Jameson GS, Fraser C, Steinbach M, et al. Single-cell transcriptome analysis of Tumor and stromal compartments of pancreatic ductal adenocarcinoma primary tumors and metastatic lesions. Genome Med. 2020;12:80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Lu M, Zou Y, Fu P, Li Y, Wang P, Li G, Luo S, Chen Y, Guan G, Zhang S, Chen L. The tumor-stroma ratio and the immune microenvironment improve the prognostic prediction of pancreatic ductal adenocarcinoma. Discov Oncol. 2023;14:124.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Huang A, Zhao X, Yang XR, Li FQ, Zhou XL, Wu K, Zhang X, Sun QM, Cao Y, Zhu HM et al. Corrigendum to “Circumventing intratumoral heterogeneity to identify potential therapeutic targets in hepatocellular carcinoma” [J Hepatol 67 (2017) 293–301]. J Hepatol. 2017;67:1123.

  134. Schürch CM, Bhate SS, Barlow GL, Phillips DJ, Noti L, Zlobec I, Chu P, Black S, Demeter J, McIlwain DR, et al. Coordinated Cellular neighborhoods Orchestrate Antitumoral immunity at the Colorectal Cancer Invasive Front. Cell. 2020;182:1341–1359e1319.

    Article  PubMed  PubMed Central  Google Scholar 

  135. Huang A, Zhao X, Yang XR, Li FQ, Zhou XL, Wu K, Zhang X, Sun QM, Cao Y, Zhu HM, et al. Circumventing intratumoral heterogeneity to identify potential therapeutic targets in hepatocellular carcinoma. J Hepatol. 2017;67:293–301.

    Article  CAS  PubMed  Google Scholar 

  136. Wu L, Yan J, Bai Y, Chen F, Zou X, Xu J, Huang A, Hou L, Zhong Y, Jing Z, et al. An invasive zone in human Liver cancer identified by stereo-seq promotes hepatocyte-tumor cell crosstalk, local immunosuppression and Tumor progression. Cell Res. 2023;33:585–603.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Xu W, Zhao M, Fu X, Hou J, Wang Y, Shi F, Hu S. Molecular mechanisms underlying macrophage immunomodulatory activity of Rubus Chingii Hu polysaccharides. Int J Biol Macromol. 2021;185:907–16.

    Article  CAS  PubMed  Google Scholar 

  138. Kelly PA, Djiane J, Postel-Vinay MC, Edery M. The prolactin/growth hormone receptor family. Endocr Rev. 1991;12:235–51.

    Article  CAS  PubMed  Google Scholar 

  139. Assy N, Pruzansky Y, Gaitini D, Shen Orr Z, Hochberg Z, Baruch Y. Growth hormone-stimulated IGF-1 generation in Cirrhosis reflects hepatocellular dysfunction. J Hepatol. 2008;49:34–42.

    Article  CAS  PubMed  Google Scholar 

  140. Perry JK, Wu ZS, Mertani HC, Zhu T, Lobie PE. Tumour-derived human growth hormone as a therapeutic target in Oncology. Trends Endocrinol Metab. 2017;28:587–96.

    Article  CAS  PubMed  Google Scholar 

  141. Zhou C, Jiao Y, Wang R, Ren SG, Wawrowsky K, Melmed S. STAT3 upregulation in pituitary somatotroph adenomas induces growth hormone hypersecretion. J Clin Invest. 2015;125:1692–702.

    Article  PubMed  PubMed Central  Google Scholar 

  142. Wennbo H, Törnell J. The role of prolactin and growth hormone in Breast cancer. Oncogene. 2000;19:1072–6.

    Article  CAS  PubMed  Google Scholar 

  143. Haque A, Sahu V, Lombardo JL, Xiao L, George B, Wolff RA, Morris JS, Rashid A, Kopchick JJ, Kaseb AO, Amin HM. Disruption of growth hormone receptor signaling abrogates Hepatocellular Carcinoma Development. J Hepatocell Carcinoma. 2022;9:823–37.

    Article  PubMed  PubMed Central  Google Scholar 

  144. González L, Díaz ME, Miquet JG, Sotelo AI, Dominici FP. Growth hormone modulation of hepatic epidermal growth factor receptor signaling. Trends Endocrinol Metab. 2021;32:403–14.

    Article  PubMed  Google Scholar 

  145. Snibson KJ. Hepatocellular kinetics and the expression of growth hormone (GH) in the livers and liver tumours of GH-transgenic mice. Tissue Cell. 2002;34:88–97.

    Article  CAS  PubMed  Google Scholar 

  146. García-Caballero T, Mertani HM, Lambert A, Gallego R, Fraga M, Pintos E, Forteza J, Chevallier M, Lobie PE, Vonderhaar BK, et al. Increased expression of growth hormone and prolactin receptors in hepatocellular carcinomas. Endocrine. 2000;12:265–71.

    Article  PubMed  Google Scholar 

  147. Abu El-Makarem MA, Kamel MF, Mohamed AA, Ali HA, Mohamed MR, Mohamed AEM, El-Said AM, Ameen MG, Hassnine AA, Hassan HA. Down-regulation of hepatic expression of GHR/STAT5/IGF-1 signaling pathway fosters development and aggressiveness of HCV-related hepatocellular carcinoma: crosstalk with Snail-1 and type 2 transforming growth factor-beta receptor. PLoS ONE. 2022;17:e0277266.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Kaseb AO, Haque A, Vishwamitra D, Hassan MM, Xiao L, George B, Sahu V, Mohamed YI, Carmagnani Pestana R, Lombardo JL, et al. Blockade of growth hormone receptor signaling by using pegvisomant: a functional therapeutic strategy in hepatocellular carcinoma. Front Oncol. 2022;12:986305.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Friedbichler K, Themanns M, Mueller KM, Schlederer M, Kornfeld JW, Terracciano LM, Kozlov AV, Haindl S, Kenner L, Kolbe T, et al. Growth-hormone-induced signal transducer and activator of transcription 5 signaling causes gigantism, inflammation, and premature death but protects mice from aggressive Liver cancer. Hepatology. 2012;55:941–52.

    Article  CAS  PubMed  Google Scholar 

  150. MacDougald OA, Hwang CS, Fan H, Lane MD. Regulated expression of the obese gene product (leptin) in white adipose tissue and 3T3-L1 adipocytes. Proc Natl Acad Sci U S A. 1995;92:9034–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Sierra-Honigmann MR, Nath AK, Murakami C, García-Cardeña G, Papapetropoulos A, Sessa WC, Madge LA, Schechner JS, Schwabb MB, Polverini PJ, Flores-Riveros JR. Biological action of leptin as an angiogenic factor. Science. 1998;281:1683–6.

    Article  CAS  PubMed  Google Scholar 

  152. Grossmann ME, Ray A, Dogan S, Mizuno NK, Cleary MP. Balance of adiponectin and leptin modulates Breast cancer cell growth. Cell Res. 2008;18:1154–6.

    Article  CAS  PubMed  Google Scholar 

  153. Feng H, Zhang Q, Zhao Y, Zhao L, Shan B. Leptin acts on mesenchymal stem cells to promote chemoresistance in osteosarcoma cells. Aging. 2020;12:6340–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Sharma D, Wang J, Fu PP, Sharma S, Nagalingam A, Mells J, Handy J, Page AJ, Cohen C, Anania FA, Saxena NK. Adiponectin antagonizes the oncogenic actions of leptin in hepatocellular carcinogenesis. Hepatology. 2010;52:1713–22.

    Article  CAS  PubMed  Google Scholar 

  155. Babic A, Bao Y, Qian ZR, Yuan C, Giovannucci EL, Aschard H, Kraft P, Amundadottir LT, Stolzenberg-Solomon R, Morales-Oyarvide V, et al. Pancreatic Cancer Risk Associated with Prediagnostic plasma levels of leptin and Leptin Receptor Genetic Polymorphisms. Cancer Res. 2016;76:7160–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Saxena NK, Sharma D, Ding X, Lin S, Marra F, Merlin D, Anania FA. Concomitant activation of the JAK/STAT, PI3K/AKT, and ERK signaling is involved in leptin-mediated promotion of invasion and migration of hepatocellular carcinoma cells. Cancer Res. 2007;67:2497–507.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Lin XL, Liu M, Liu Y, Hu H, Pan Y, Zou W, Fan X, Hu X. Transforming growth factor β1 promotes migration and invasion in HepG2 cells: epithelial–to–mesenchymal transition via JAK/STAT3 signaling. Int J Mol Med. 2018;41:129–36.

    CAS  PubMed  Google Scholar 

  158. Xiong Q, Wu S, Wang J, Zeng X, Chen J, Wei M, Guan H, Fan C, Chen L, Guo D, Sun G. Hepatitis B virus promotes cancer cell migration by downregulating mir-340-5p expression to induce STAT3 overexpression. Cell Biosci. 2017;7:16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Xu F, Song H, Xiao Q, Li N, Zhang H, Cheng G, Tan G. Type III interferon-induced CBFβ inhibits HBV replication by Hijacking HBx. Cell Mol Immunol. 2019;16:357–66.

    Article  CAS  PubMed  Google Scholar 

  160. Brook MG, McDonald JA, Karayiannis P, Caruso L, Forster G, Harris JR, Thomas HC. Randomised controlled trial of interferon alfa 2A (rbe) (Roferon-A) for the treatment of chronic Hepatitis B virus (HBV) Infection: factors that influence response. Gut. 1989;30:1116–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Perrillo R. Benefits and risks of interferon therapy for Hepatitis B. Hepatology. 2009;49:103–11.

    Article  Google Scholar 

  162. Hung WC, Chuang LY. Sodium butyrate enhances STAT 1 expression in PLC/PRF/5 hepatoma cells and augments their responsiveness to interferon-alpha. Br J Cancer. 1999;80:705–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Hung JH, Li CH, Yeh CH, Huang PC, Fang CC, Chen YF, Lee KJ, Chou CH, Cheng HY, Huang HD, et al. MicroRNA-224 down-regulates Glycine N-methyltransferase gene expression in Hepatocellular Carcinoma. Sci Rep. 2018;8:12284.

    Article  PubMed  PubMed Central  Google Scholar 

  164. Wang YC, Chen YM, Lin YJ, Liu SP, Chiang EP. GNMT expression increases hepatic folate contents and folate-dependent methionine synthase-mediated homocysteine remethylation. Mol Med. 2011;17:486–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Chen PM, Tsai CH, Huang CC, Hwang HH, Li JR, Liu CC, Ko HA, Chiang EI. Downregulation of methionine cycle genes MAT1A and GNMT enriches protein-Associated translation process and worsens Hepatocellular Carcinoma Prognosis. Int J Mol Sci. 2022;23.

  166. Kant R, Yen CH, Hung JH, Lu CK, Tung CY, Chang PC, Chen YH, Tyan YC, Chen YA. Induction of GNMT by 1,2,3,4,6-penta-O-galloyl-beta-D-glucopyranoside through proteasome-independent MYC downregulation in hepatocellular carcinoma. Sci Rep. 2019;9:1968.

  167. Chang MM, Lin CN, Fang CC, Chen M, Liang PI, Li WM, Yeh BW, Cheng HC, Huang BM, Wu WJ, Chen YA. Glycine N-methyltransferase inhibits aristolochic acid Nephropathy by increasing CYP3A44 and decreasing NQO1 expression in female mouse hepatocytes. Sci Rep. 2018;8:6960.

    Article  PubMed  PubMed Central  Google Scholar 

  168. Martínez-Chantar ML, Vázquez-Chantada M, Ariz U, Martínez N, Varela M, Luka Z, Capdevila A, Rodríguez J, Aransay AM, Matthiesen R, et al. Loss of the glycine N-methyltransferase gene leads to steatosis and hepatocellular carcinoma in mice. Hepatology. 2008;47:1191–9.

    Article  PubMed  Google Scholar 

  169. Liu ZK, Li C, Zhang RY, Wei D, Shang YK, Yong YL, Kong LM, Zheng NS, Liu K, Lu M, et al. EYA2 suppresses the progression of hepatocellular carcinoma via SOCS3-mediated blockade of JAK/STAT signaling. Mol Cancer. 2021;20:79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Wen LZ, Ding K, Wang ZR, Ding CH, Lei SJ, Liu JP, Yin C, Hu PF, Ding J, Chen WS, et al. SHP-1 acts as a Tumor suppressor in Hepatocarcinogenesis and HCC Progression. Cancer Res. 2018;78:4680–91.

    Article  CAS  PubMed  Google Scholar 

  171. Toh TB, Lim JJ, Hooi L, Rashid M, Chow EK. Targeting Jak/Stat pathway as a therapeutic strategy against SP/CD44 + tumorigenic cells in Akt/β-catenin-driven hepatocellular carcinoma. J Hepatol. 2020;72:104–18.

    Article  CAS  PubMed  Google Scholar 

  172. Cafferkey C, Chau I. Novel STAT 3 inhibitors for treating gastric cancer. Expert Opin Investig Drugs. 2016;25:1023–31.

    Article  CAS  PubMed  Google Scholar 

  173. Denson LA. Adding fuel to the Fire: STAT3 priming of gastric tumorigenesis. Gastroenterology. 2006;131:1342–4.

    Article  PubMed  Google Scholar 

  174. Deng JY, Sun D, Liu XY, Pan Y, Liang H. STAT-3 correlates with lymph node Metastasis and cell survival in gastric cancer. World J Gastroenterol. 2010;16:5380–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Judd LM, Bredin K, Kalantzis A, Jenkins BJ, Ernst M, Giraud AS. STAT3 activation regulates growth, inflammation, and vascularization in a mouse model of gastric tumorigenesis. Gastroenterology. 2006;131:1073–85.

    Article  CAS  PubMed  Google Scholar 

  176. Kanda N, Seno H, Konda Y, Marusawa H, Kanai M, Nakajima T, Kawashima T, Nanakin A, Sawabu T, Uenoyama Y, et al. STAT3 is constitutively activated and supports cell survival in association with survivin expression in gastric cancer cells. Oncogene. 2004;23:4921–9.

    Article  CAS  PubMed  Google Scholar 

  177. Tye H, Kennedy CL, Najdovska M, McLeod L, McCormack W, Hughes N, Dev A, Sievert W, Ooi CH, Ishikawa TO, et al. STAT3-driven upregulation of TLR2 promotes gastric tumorigenesis Independent of Tumor inflammation. Cancer Cell. 2012;22:466–78.

    Article  CAS  PubMed  Google Scholar 

  178. Clyne M, Dillon P, Daly S, O’Kennedy R, May FE, Westley BR, Drumm B. Helicobacter pylori interacts with the human single-domain trefoil protein TFF1. Proc Natl Acad Sci U S A. 2004;101:7409–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Soutto M, Belkhiri A, Piazuelo MB, Schneider BG, Peng D, Jiang A, Washington MK, Kokoye Y, Crowe SE, Zaika A, et al. Loss of TFF1 is associated with activation of NF-κB-mediated inflammation and gastric neoplasia in mice and humans. J Clin Invest. 2011;121:1753–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Soutto M, Peng D, Katsha A, Chen Z, Piazuelo MB, Washington MK, Belkhiri A, Correa P, El-Rifai W. Activation of β-catenin signalling by TFF1 loss promotes cell proliferation and gastric tumorigenesis. Gut. 2015;64:1028–39.

    Article  CAS  PubMed  Google Scholar 

  181. Soutto M, Chen Z, Bhat AA, Wang L, Zhu S, Gomaa A, Bates A, Bhat NS, Peng D, Belkhiri A, et al. Activation of STAT3 signaling is mediated by TFF1 silencing in gastric neoplasia. Nat Commun. 2019;10:3039.

    Article  PubMed  PubMed Central  Google Scholar 

  182. Cassetta L, Pollard JW. A timeline of tumour-associated macrophage biology. Nat Rev Cancer. 2023;23:238–57.

    Article  CAS  PubMed  Google Scholar 

  183. Zhang M, Pan X, Fujiwara K, Jurcak N, Muth S, Zhou J, Xiao Q, Li A, Che X, Li Z, Zheng L. Pancreatic cancer cells render tumor-associated macrophages metabolically reprogrammed by a GARP and DNA methylation-mediated mechanism. Signal Transduct Target Ther. 2021;6:366.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Wang Y, Tiruthani K, Li S, Hu M, Zhong G, Tang Y, Roy S, Zhang L, Tan J, Liao C, Liu R. mRNA delivery of a bispecific single-domain antibody to Polarize Tumor-Associated macrophages and Synergize Immunotherapy against Liver malignancies. Adv Mater. 2021;33:e2007603.

    Article  PubMed  PubMed Central  Google Scholar 

  185. Piao H, Fu L, Wang Y, Liu Y, Wang Y, Meng X, Yang D, Xiao X, Zhang J. A positive feedback loop between gastric cancer cells and tumor-associated macrophage induces malignancy progression. J Exp Clin Cancer Res. 2022;41:174.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Miao L, Qi J, Zhao Q, Wu QN, Wei DL, Wei XL, Liu J, Chen J, Zeng ZL, Ju HQ, et al. Targeting the STING pathway in tumor-associated macrophages regulates innate immune sensing of gastric cancer cells. Theranostics. 2020;10:498–515.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Coombes JL, Powrie F. Dendritic cells in intestinal immune regulation. Nat Rev Immunol. 2008;8:435–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Palucka K, Banchereau J. Cancer immunotherapy via dendritic cells. Nat Rev Cancer. 2012;12:265–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Pulendran B, Banchereau J, Maraskovsky E, Maliszewski C. Modulating the immune response with dendritic cells and their growth factors. Trends Immunol. 2001;22:41–7.

    Article  CAS  PubMed  Google Scholar 

  190. Bai X, Wong CC, Pan Y, Chen H, Liu W, Zhai J, Kang W, Shi Y, Yamamoto M, Tsukamoto T et al. Loss of YTHDF1 in gastric tumors restores sensitivity to antitumor immunity by recruiting mature dendritic cells. J Immunother Cancer. 2022;10.

  191. Caruso RA, Bellocco R, Pagano M, Bertoli G, Rigoli L, Inferrera C. Prognostic value of intratumoral neutrophils in advanced gastric carcinoma in a high-risk area in northern Italy. Mod Pathol. 2002;15:831–7.

    Article  PubMed  Google Scholar 

  192. Zhang H, Liu H, Shen Z, Lin C, Wang X, Qin J, Qin X, Xu J, Sun Y. Tumor-infiltrating neutrophils is prognostic and predictive for postoperative adjuvant Chemotherapy Benefit in patients with gastric Cancer. Ann Surg. 2018;267:311–8.

    Article  PubMed  Google Scholar 

  193. Zhao JJ, Pan K, Wang W, Chen JG, Wu YH, Lv L, Li JJ, Chen YB, Wang DD, Pan QZ, et al. The prognostic value of tumor-infiltrating neutrophils in gastric adenocarcinoma after resection. PLoS ONE. 2012;7:e33655.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Shan ZG, Zhao YL, Zhang JY, Yan ZB, Wang TT, Mao FY, Teng YS, Peng LS, Chen WY, Wang P, et al. FasL(+) PD-L2(+) identifies a Novel Immunosuppressive Neutrophil Population in Human gastric Cancer that promotes Disease Progression. Adv Sci (Weinh). 2022;9:e2103543.

    Article  PubMed  Google Scholar 

  195. Wang TT, Zhao YL, Peng LS, Chen N, Chen W, Lv YP, Mao FY, Zhang JY, Cheng P, Teng YS, et al. Tumour-activated neutrophils in gastric cancer foster immune suppression and Disease progression through GM-CSF-PD-L1 pathway. Gut. 2017;66:1900–11.

    Article  CAS  PubMed  Google Scholar 

  196. Wu H, Liang J. Contributions of NFKB1 -94insertion/deletion ATTG polymorphism to the susceptibility of gastrointestinal cancers: a meta-analysis. J Cell Mol Med. 2021;25:10674–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Vangsted AJ, Klausen TW, Gimsing P, Andersen NF, Abildgaard N, Gregersen H, Vogel U. A polymorphism in NFKB1 is associated with improved effect of interferon-{alpha} maintenance treatment of patients with Multiple Myeloma after high-dose treatment with stem cell support. Haematologica. 2009;94:1274–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Companioni O, Bonet C, García N, Ramírez-Lázaro MJ, Lario S, Mendoza J, Adrados MM, Poves E, Espinosa L, Pozo-Kreilinger JJ, et al. Genetic variation analysis in a follow-up study of gastric cancer precursor lesions confirms the association of MUC2 variants with the evolution of the lesions and identifies a significant association with NFKB1 and CD14. Int J Cancer. 2018;143:2777–86.

    Article  CAS  PubMed  Google Scholar 

  199. Grivennikov SI, Karin M. Dangerous liaisons: STAT3 and NF-kappaB collaboration and crosstalk in cancer. Cytokine Growth Factor Rev. 2010;21:11–9.

    Article  CAS  PubMed  Google Scholar 

  200. O’Reilly LA, Putoczki TL, Mielke LA, Low JT, Lin A, Preaudet A, Herold MJ, Yaprianto K, Tai L, Kueh A, et al. Loss of NF-κB1 causes gastric Cancer with aberrant inflammation and expression of Immune Checkpoint regulators in a STAT-1-Dependent manner. Immunity. 2018;48:570–583e578.

    Article  PubMed  Google Scholar 

  201. Low JT, Christie M, Ernst M, Dumoutier L, Preaudet A, Ni Y, Griffin MDW, Mielke LA, Strasser A, Putoczki TL, O’Reilly LA. Loss of NFKB1 results in expression of Tumor Necrosis Factor and Activation of Signal Transducer and activator of transcription 1 to promote gastric tumorigenesis in mice. Gastroenterology. 2020;159:1444–1458e1415.

    Article  CAS  PubMed  Google Scholar 

  202. Silva VR, Santos LS, Dias RB, Quadros CA, Bezerra DP. Emerging agents that target signaling pathways to eradicate Colorectal cancer stem cells. Cancer Commun (Lond). 2021;41:1275–313.

    Article  PubMed  Google Scholar 

  203. Chang TC, Yeh CT, Adebayo BO, Lin YC, Deng L, Rao YK, Huang CC, Lee WH, Wu AT, Hsiao M, et al. 4-Acetylantroquinonol B inhibits Colorectal cancer tumorigenesis and suppresses cancer stem-like phenotype. Toxicol Appl Pharmacol. 2015;288:258–68.

    Article  CAS  PubMed  Google Scholar 

  204. Farooqi AA, de la Roche M, Djamgoz MBA, Siddik ZH. Overview of the oncogenic signaling pathways in Colorectal cancer: mechanistic insights. Semin Cancer Biol. 2019;58:65–79.

    Article  CAS  PubMed  Google Scholar 

  205. Schmitt M, Greten FR. The inflammatory pathogenesis of Colorectal cancer. Nat Rev Immunol. 2021;21:653–67.

    Article  CAS  PubMed  Google Scholar 

  206. Park JH, Watt DG, Roxburgh CS, Horgan PG, McMillan DC. Colorectal Cancer, systemic inflammation, and Outcome: staging the Tumor and staging the host. Ann Surg. 2016;263:326–36.

    Article  PubMed  Google Scholar 

  207. Suzuki Y, Okabayashi K, Hasegawa H, Tsuruta M, Shigeta K, Kondo T, Kitagawa Y. Comparison of Preoperative Inflammation-based Prognostic scores in patients with Colorectal Cancer. Ann Surg. 2018;267:527–31.

    Article  PubMed  Google Scholar 

  208. Yasui K, Shida D, Nakamura Y, Ahiko Y, Tsukamoto S, Kanemitsu Y. Postoperative, but not preoperative, inflammation-based prognostic markers are prognostic factors in stage III Colorectal cancer patients. Br J Cancer. 2021;124:933–41.

    Article  CAS  PubMed  Google Scholar 

  209. Park JH, van Wyk H, McMillan DC, Quinn J, Clark J, Roxburgh CS, Horgan PG, Edwards J. Signal Transduction and activator of Transcription-3 (STAT3) in patients with Colorectal Cancer: associations with the phenotypic features of the Tumor and host. Clin Cancer Res. 2017;23:1698–709.

    Article  CAS  PubMed  Google Scholar 

  210. Corvinus FM, Orth C, Moriggl R, Tsareva SA, Wagner S, Pfitzner EB, Baus D, Kaufmann R, Huber LA, Zatloukal K, et al. Persistent STAT3 activation in colon Cancer is associated with enhanced cell proliferation and Tumor growth. Neoplasia. 2005;7:545–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Campbell H, Fleming N, Roth I, Mehta S, Wiles A, Williams G, Vennin C, Arsic N, Parkin A, Pajic M, et al. ∆133p53 isoform promotes tumour invasion and Metastasis via interleukin-6 activation of JAK-STAT and RhoA-ROCK signalling. Nat Commun. 2018;9:254.

    Article  PubMed  PubMed Central  Google Scholar 

  212. Zheng X, Song J, Yu C, Zhou Z, Liu X, Yu J, Xu G, Yang J, He X, Bai X, et al. Single-cell transcriptomic profiling unravels the adenoma-initiation role of protein tyrosine kinases during colorectal tumorigenesis. Signal Transduct Target Ther. 2022;7:60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  213. Bian Z, Zhang J, Li M, Feng Y, Wang X, Zhang J, Yao S, Jin G, Du J, Han W, et al. LncRNA-FEZF1-AS1 promotes Tumor Proliferation and Metastasis in Colorectal Cancer by regulating PKM2 signaling. Clin Cancer Res. 2018;24:4808–19.

    Article  CAS  PubMed  Google Scholar 

  214. Iliopoulos D, Jaeger SA, Hirsch HA, Bulyk ML, Struhl K. STAT3 activation of miR-21 and miR-181b-1 via PTEN and CYLD are part of the epigenetic switch linking inflammation to cancer. Mol Cell. 2010;39:493–506.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  215. Xu DD, Zhou PJ, Wang Y, Zhang L, Fu WY, Ruan BB, Xu HP, Hu CZ, Tian L, Qin JH, et al. Reciprocal activation between STAT3 and miR-181b regulates the proliferation of Esophageal cancer stem-like cells via the CYLD pathway. Mol Cancer. 2016;15:40.

    Article  PubMed  PubMed Central  Google Scholar 

  216. Liu S, Sun X, Wang M, Hou Y, Zhan Y, Jiang Y, Liu Z, Cao X, Chen P, Liu Z, et al. A microRNA 221- and 222-mediated feedback loop maintains constitutive activation of NFκB and STAT3 in Colorectal cancer cells. Gastroenterology. 2014;147:847–859e811.

    Article  CAS  PubMed  Google Scholar 

  217. Wang N, He X, Zhou R, Jia G, Qiao Q. STAT3 induces colorectal carcinoma progression through a novel mir-572-MOAP-1 pathway. Onco Targets Ther. 2018;11:3475–84.

    Article  PubMed  PubMed Central  Google Scholar 

  218. Ma J, Yang Y, Fu Y, Guo F, Zhang X, Xiao S, Zhu W, Huang Z, Zhang J, Chen J. PIAS3-mediated feedback loops promote chronic colitis-associated malignant transformation. Theranostics. 2018;8:3022–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  219. Nagathihalli NS, Castellanos JA, Shi C, Beesetty Y, Reyzer ML, Caprioli R, Chen X, Walsh AJ, Skala MC, Moses HL, Merchant NB. Signal Transducer and Activator of Transcription 3, Mediated Remodeling of the Tumor Microenvironment Results in Enhanced Tumor Drug Delivery in a Mouse Model of Pancreatic Cancer. Gastroenterology. 2015;149:1932–1943.e1939.

  220. Qiu Z, Huang C, Sun J, Qiu W, Zhang J, Li H, Jiang T, Huang K, Cao J. RNA interference-mediated signal transducers and activators of transcription 3 gene silencing inhibits invasion and Metastasis of human Pancreatic cancer cells. Cancer Sci. 2007;98:1099–106.

    Article  CAS  PubMed  Google Scholar 

  221. Huang C, Yang G, Jiang T, Huang K, Cao J, Qiu Z. Effects of IL-6 and AG490 on regulation of Stat3 signaling pathway and invasion of human Pancreatic cancer cells in vitro. J Exp Clin Cancer Res. 2010;29:51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  222. Mace TA, Bloomston M, Lesinski GB. Pancreatic cancer-associated stellate cells: a viable target for reducing immunosuppression in the Tumor microenvironment. Oncoimmunology. 2013;2:e24891.

    Article  PubMed  PubMed Central  Google Scholar 

  223. Yoo HB, Moon JW, Kim HR, Lee HS, Miyabayashi K, Park CH, Ge S, Zhang A, Tae YK, Sub Y, et al. A TEAD2-Driven endothelial-like Program shapes basal-like differentiation and Metastasis of Pancreatic Cancer. Gastroenterology. 2023;165:133–148e117.

    Article  CAS  PubMed  Google Scholar 

  224. Guerra C, Schuhmacher AJ, Cañamero M, Grippo PJ, Verdaguer L, Pérez-Gallego L, Dubus P, Sandgren EP, Barbacid M. Chronic Pancreatitis is essential for induction of pancreatic ductal adenocarcinoma by K-Ras oncogenes in adult mice. Cancer Cell. 2007;11:291–302.

    Article  CAS  PubMed  Google Scholar 

  225. Almoguera C, Shibata D, Forrester K, Martin J, Arnheim N, Perucho M. Most human carcinomas of the exocrine pancreas contain mutant c-K-ras genes. Cell. 1988;53:549–54.

    Article  CAS  PubMed  Google Scholar 

  226. Kanda M, Matthaei H, Wu J, Hong SM, Yu J, Borges M, Hruban RH, Maitra A, Kinzler K, Vogelstein B, Goggins M. Presence of somatic mutations in most early-stage pancreatic intraepithelial neoplasia. Gastroenterology. 2012;142:730–733e739.

    Article  CAS  PubMed  Google Scholar 

  227. Gruber R, Panayiotou R, Nye E, Spencer-Dene B, Stamp G, Behrens A. YAP1 and TAZ control Pancreatic Cancer initiation in mice by direct Up-regulation of JAK-STAT3 signaling. Gastroenterology. 2016;151:526–39.

    Article  CAS  PubMed  Google Scholar 

  228. Perusina Lanfranca M, Zhang Y, Girgis A, Kasselman S, Lazarus J, Kryczek I, Delrosario L, Rhim A, Koneva L, Sartor M, et al. Interleukin 22 signaling regulates Acinar Cell plasticity to promote pancreatic Tumor Development in mice. Gastroenterology. 2020;158:1417–1432e1411.

    Article  CAS  PubMed  Google Scholar 

  229. Caraglia M, Tagliaferri P, Marra M, Giuberti G, Budillon A, Gennaro ED, Pepe S, Vitale G, Improta S, Tassone P, et al. EGF activates an inducible survival response via the RAS-> Erk-1/2 pathway to counteract interferon-alpha-mediated apoptosis in epidermoid cancer cells. Cell Death Differ. 2003;10:218–29.

    Article  CAS  PubMed  Google Scholar 

  230. Caraglia M, Abbruzzese A, Leardi A, Pepe S, Budillon A, Baldassare G, Selleri C, Lorenzo SD, Fabbrocini A, Giuberti G, et al. Interferon-alpha induces apoptosis in human KB cells through a stress-dependent mitogen activated protein kinase pathway that is antagonized by epidermal growth factor. Cell Death Differ. 1999;6:773–80.

    Article  CAS  PubMed  Google Scholar 

  231. Motomura W, Nagamine M, Tanno S, Sawamukai M, Takahashi N, Kohgo Y, Okumura T. Inhibition of cell invasion and morphological change by troglitazone in human Pancreatic cancer cells. J Gastroenterol. 2004;39:461–8.

    Article  CAS  PubMed  Google Scholar 

  232. Motomura W, Okumura T, Takahashi N, Obara T, Kohgo Y. Activation of peroxisome proliferator-activated receptor gamma by troglitazone inhibits cell growth through the increase of p27KiP1 in human. Pancreatic carcinoma cells. Cancer Res. 2000;60:5558–64.

    CAS  PubMed  Google Scholar 

  233. Farrow B, O’Connor KL, Hashimoto K, Iwamura T, Evers BM. Selective activation of PPARgamma inhibits Pancreatic cancer invasion and decreases expression of tissue plasminogen activator. Surgery. 2003;134:206–12.

    Article  PubMed  Google Scholar 

  234. Wang D, Zhou Y, Lei W, Zhang K, Shi J, Hu Y, Shu G, Song J. Signal transducer and activator of transcription 3 (STAT3) regulates adipocyte differentiation via peroxisome-proliferator-activated receptor gamma (PPARgamma). Biol Cell. 2009;102:1–12.

    Article  PubMed  Google Scholar 

  235. Vitale G, Zappavigna S, Marra M, Dicitore A, Meschini S, Condello M, Arancia G, Castiglioni S, Maroni P, Bendinelli P, et al. The PPAR-γ agonist troglitazone antagonizes survival pathways induced by STAT-3 in recombinant interferon-β treated Pancreatic cancer cells. Biotechnol Adv. 2012;30:169–84.

    Article  CAS  PubMed  Google Scholar 

  236. Dandawate P, Kaushik G, Ghosh C, Standing D, Ali Sayed AA, Choudhury S, Subramaniam D, Manzardo A, Banerjee T, Santra S, et al. Diphenylbutylpiperidine antipsychotic Drugs inhibit prolactin receptor signaling to reduce growth of pancreatic ductal adenocarcinoma in mice. Gastroenterology. 2020;158:1433–1449e1427.

    Article  CAS  PubMed  Google Scholar 

  237. Ma RJ, Ma C, Hu K, Zhao MM, Zhang N, Sun ZG. Molecular mechanism, regulation, and therapeutic targeting of the STAT3 signaling pathway in Esophageal cancer (review). Int J Oncol. 2022;61.

  238. Leu CM, Wong FH, Chang C, Huang SF, Hu CP. Interleukin-6 acts as an antiapoptotic factor in human esophageal carcinoma cells through the activation of both STAT3 and mitogen-activated protein kinase pathways. Oncogene. 2003;22:7809–18.

    Article  CAS  PubMed  Google Scholar 

  239. Lei YY, Feng YF, Zeng B, Zhang W, Xu Q, Cheng F, Lan J, Luo HH, Zou JY, Chen ZG, et al. Exogenous H(2)S promotes cancer progression by activating JAK2/STAT3 signaling pathway in esophageal EC109 cells. Int J Clin Exp Pathol. 2018;11:3247–56.

    PubMed  PubMed Central  Google Scholar 

  240. Strebhardt K. Multifaceted polo-like kinases: drug targets and antitargets for cancer therapy. Nat Rev Drug Discov. 2010;9:643–60.

    Article  CAS  PubMed  Google Scholar 

  241. Raab CA, Raab M, Becker S, Strebhardt K. Non-mitotic functions of polo-like kinases in cancer cells. Biochim Biophys Acta Rev Cancer. 2021;1875:188467.

    Article  CAS  PubMed  Google Scholar 

  242. Helmke C, Becker S, Strebhardt K. The role of Plk3 in oncogenesis. Oncogene. 2016;35:135–47.

    Article  CAS  PubMed  Google Scholar 

  243. Feng YB, Lin DC, Shi ZZ, Wang XC, Shen XM, Zhang Y, Du XL, Luo ML, Xu X, Han YL, et al. Overexpression of PLK1 is associated with poor survival by inhibiting apoptosis via enhancement of survivin level in esophageal squamous cell carcinoma. Int J Cancer. 2009;124:578–88.

    Article  CAS  PubMed  Google Scholar 

  244. Zhang Y, Du XL, Wang CJ, Lin DC, Ruan X, Feng YB, Huo YQ, Peng H, Cui JL, Zhang TT, et al. Reciprocal activation between PLK1 and Stat3 contributes to survival and proliferation of Esophageal cancer cells. Gastroenterology. 2012;142:521–530e523.

    Article  CAS  PubMed  Google Scholar 

  245. Andl CD, Mizushima T, Oyama K, Bowser M, Nakagawa H, Rustgi AK. EGFR-induced cell migration is mediated predominantly by the JAK-STAT pathway in primary esophageal keratinocytes. Am J Physiol Gastrointest Liver Physiol. 2004;287:G1227–1237.

    Article  CAS  PubMed  Google Scholar 

  246. Yu N, Xue M, Wang W, Xia D, Li Y, Zhou X, Pang D, Lu K, Hou J, Zhang A, et al. RNF168 facilitates proliferation and invasion of esophageal carcinoma, possibly via stabilizing STAT1. J Cell Mol Med. 2019;23:1553–61.

    Article  CAS  PubMed  Google Scholar 

  247. Tan R, Liu J, Wang J, Zhang W, He M, Zhang Y. Long noncoding RNA SNHG6 silencing sensitized Esophageal cancer cells to 5-FU via EZH2/STAT pathway. Sci Rep. 2023;13:5363.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  248. Hu X, Li J, Fu M, Zhao X, Wang W. The JAK/STAT signaling pathway: from bench to clinic. Signal Transduct Target Ther. 2021;6:402.

    Article  PubMed  PubMed Central  Google Scholar 

  249. Krasilnikov M, Ivanov VN, Dong J, Ronai Z. ERK and PI3K negatively regulate STAT-transcriptional activities in human Melanoma cells: implications towards sensitization to apoptosis. Oncogene. 2003;22:4092–101.

    Article  CAS  PubMed  Google Scholar 

  250. Wu Q, Wu W, Fu B, Shi L, Wang X, Kuca K. JNK signaling in cancer cell survival. Med Res Rev. 2019;39:2082–104.

    Article  CAS  PubMed  Google Scholar 

  251. He G, Karin M. NF-κB and STAT3 - key players in liver inflammation and cancer. Cell Res. 2011;21:159–68.

    Article  CAS  PubMed  Google Scholar 

  252. Lu X, Wo G, Li B, Xu C, Wu J, Jiang C, Wei J. The anti-inflammatory NHE-06 restores antitumor immunity by targeting NF-κB/IL-6/STAT3 signaling in hepatocellular carcinoma. Biomed Pharmacother. 2018;102:420–7.

    Article  CAS  PubMed  Google Scholar 

  253. Zhou Q, Tian W, Jiang Z, Huang T, Ge C, Liu T, Zhao F, Chen T, Cui Y, Li H, et al. A positive Feedback Loop of AKR1C3-Mediated activation of NF-κB and STAT3 facilitates proliferation and Metastasis in Hepatocellular Carcinoma. Cancer Res. 2021;81:1361–74.

    Article  CAS  PubMed  Google Scholar 

  254. van de Stolpe A. Quantitative measurement of functional activity of the PI3K Signaling Pathway in Cancer. Cancers (Basel). 2019;11.

  255. Zhu YJ, Zheng B, Wang HY, Chen L. New knowledge of the mechanisms of sorafenib resistance in Liver cancer. Acta Pharmacol Sin. 2017;38:614–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  256. Guillén Díaz-Maroto N, Sanz-Pamplona R, Berdiel-Acer M, Cimas FJ, García E, Gonçalves-Ribeiro S, Albert N, Garcia-Vicién G, Capella G, Moreno V, et al. Noncanonical TGFβ pathway relieves the blockade of IL1β/TGFβ-Mediated crosstalk between Tumor and Stroma: TGFBR1 and TAK1 inhibition in Colorectal Cancer. Clin Cancer Res. 2019;25:4466–79.

    Article  PubMed  Google Scholar 

  257. Morgensztern D, McLeod HL. PI3K/Akt/mTOR pathway as a target for cancer therapy. Anticancer Drugs. 2005;16:797–803.

    Article  CAS  PubMed  Google Scholar 

  258. Abusaliya A, Jeong SH, Bhosale PB, Kim HH, Park MY, Kim E, Won CK, Park KI, Heo JD, Kim HW et al. Mechanistic action of cell cycle arrest and intrinsic apoptosis via inhibiting Akt/mTOR and activation of p38-MAPK signaling pathways in Hep3B Liver Cancer cells by Prunetrin-A flavonoid with therapeutic potential. Nutrients. 2023;15.

  259. Zhang Z, Zhou X, Shen H, Wang D, Wang Y. Phosphorylated ERK is a potential predictor of sensitivity to sorafenib when treating hepatocellular carcinoma: evidence from an in vitro study. BMC Med. 2009;7:41.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This study was funded by the National Nature Science Foundation of China (82100640 and 82200673), and the Independent Project Fund of the State Key Laboratory for.

Diagnosis and Treatment of Infectious Diseases, the National Key Research and Development Program of China (zz202306).

Author information

Authors and Affiliations

Authors

Contributions

Bing Ruan and Juan Lu conceptualized the work, Ruihong Zhao, Zhangmin Hu, and Xiaoli Zhang wrote the manuscript. Shujuan Huang, Guodong Yu, and Wei Yu helped with reference collection and draw the figures. All authors reviewed and approved the final manuscript.

Corresponding authors

Correspondence to Juan Lu or Bing Ruan.

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 4.0 International License, which permits use, sharing, adaptation, 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 changes were made. 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/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhao, R., Hu, Z., Zhang, X. et al. The oncogenic mechanisms of the Janus kinase-signal transducer and activator of transcription pathway in digestive tract tumors. Cell Commun Signal 22, 68 (2024). https://doi.org/10.1186/s12964-023-01421-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12964-023-01421-9

Keywords