Open Access

Roles of protein tyrosine phosphatases in hepatocellular carcinoma progression (Review)

  • Authors:
    • Yi-Li Chen
    • Ching-Chuan Hsieh
    • Pei-Ming Chu
    • Jing-Yi Chen
    • Yu-Chun Huang
    • Cheng-Yi Chen
  • View Affiliations

  • Published online on: January 19, 2023     https://doi.org/10.3892/or.2023.8485
  • Article Number: 48
  • Copyright : © Chen et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY 4.0].

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hepatocellular carcinoma (HCC) represents almost 80% of all liver cancers, is the sixth most common cancer and is the second‑highest cause of cancer‑related deaths worldwide. Protein tyrosine phosphatases (PTPs), which are encoded by the largest family of phosphatase genes, play critical roles in cellular responses and are implicated in various signaling pathways. Moreover, PTPs are dysregulated and involved in various cellular processes in numerous cancers, including HCC. Kinases and phosphatases are coordinators that modulate cell activities and regulate signaling responses. There are multiple interacting signaling networks, and coordination of these signaling networks in response to a stimulus determines the physiological outcome. Numerous issues, such as drug resistance and inflammatory reactions in the tumor microenvironment, are implicated in cancer progression, and the role of PTPs in these processes has not been well elucidated. Therefore, the present review focused on discussing the relationship of PTPs with inflammatory cytokines and chemotherapy/targeted drug resistance, providing detailed information on how PTPs can modulate inflammatory reactions and drug resistance to influence progression in HCC.

Introduction

Hepatocellular carcinoma (HCC) is the most common type of liver cancer, accounting for ~90% of all cases (1). HCC is one of the most common malignancies and the third leading cause of cancer-related death (2). Over 4 million estimated new cases of HCC have been reported, and HCC has been reported to cause over 3 million estimated deaths in USA (3). Multiple factors, including hepatitis B and C, obesity, excess ingestion of alcohol and smoking, lead to HCC progression. Apart from external factors, gene deficiency or mutation is also recognized as a potential cause of liver cancer progression (48). General treatments for HCC are surgery, liver transplantation, and drug therapy. However, the overall efficacy of HCC treatment remains unsatisfactory due to the high recurrence and progression rates (9). Therefore, ongoing studies in HCC are necessary. Complex molecular signaling pathways associated with cell proliferation, metastasis, inflammatory reactions and drug resistance are involved in HCC. The multiple types of molecules involved in these pathways interact with each other and provide various signal transduction pathways that are highly activated in cancers (1013).

HCC is the most common primary hepatic malignant tumor and has been the 2nd most common cause of cancer-related years of life lost worldwide. The prevalence in Taiwan ranks 4th among all malignancies (~29/100,000) and it is the 2nd cause of mortality (14). Most individuals diagnosed with HCC often have chronic liver disease, particularly chronic hepatitis B and C infection, and have not always symptoms in the early stage. ~70% of patients cannot receive curative therapy when they are diagnosed. The average survival time of patients with HCC in Barcelona Clinic Liver Cancer (BCLC) stage C is ~11 months, and that of patients in stage D is ~3 months.

In 2007, sorafenib was approved for HCC treatment. The therapy of HCC entered a new era compared with traditional chemotherapy, and sorafenib became a standard of care in the BCLC stage C group. In the pivotal sorafenib HCC assessment randomized protocol (SHARP) trial, sorafenib in patients with BCLC stage C HCC showed an increased 2.8-month overall survival (OS) (10.7 months) compared with placebo (7.9 months) (15). Before sorafenib, no significant trial had shown a favorable response to treat patients with HCC by traditional chemotherapy. Sorafenib is a multikinase inhibitor (MKI) that suppresses tumor cell proliferation and angiogenesis through different pathways (16). However, in the last 10 years, other TKIs have failed to improve the efficacy of sorafenib, including erlotinib, brivanib, sunitinib and everolimus. Until 2017, regorafenib (REG) showed a significant treatment response in patients with sorafenib-refractory disease compared with placebo. In recent years, several TKIs have shown treatment effects as 2nd-line (REG, cabozantinib, ramuciramab) and 1st-line (lenvatinib) agents, and the development of immune checkpoint inhibitors (ICIs) has revealed another treatment option. Systemic therapy for the BCLC stage C group or BCLC stage B group refractory to transarterial embolization had more medication choices in those years (17).

HCC patients with moderately compromised liver function (Child-Pugh B functional status) have limited curative options due to the risk of liver failure (18). This is particularly evident in HCC cases not suitable for surgery or locoregional treatments. For these patients, the availability of sorafenib differs, which is differentiated by the different international guidelines and local regulatory policies (19). Previously, the potential mechanisms of metronomic capecitabine (MC) have been mentioned, including blockage of tumor angiogenesis, reduced therapeutic resistance, and activation of immune responses (2022). De Lorenzo et al (23) reported that the median OS of 35 MC-treated patients was 7.5 months [95% CI: 3.733-11.267] and 5.1 months [95% CI: 4.098-6.102] in the 70 BSC group (P=0.013) (23). Furthermore, 12 patients (34.3%) treated with MC experienced several adverse events, including fatigue (17.1%), hand-foot syndrome (8.5%), neutropenia (5.7%), and thrombocytopenia (8.5%). However, MC appears to be a safe choice for Child-Pugh B-HCC patients. Its potential antitumor activity warrants prospective evaluations (23). Similarly, MC also displayed a therapeutic alternative for CP-B patients who were resistant to tyrosine kinase inhibitors (TKIs).

Furthermore, radiation, chemotherapy, TKIs and ICIs as adjuvant strategies have not been demonstrated to improve the clinical status of progression-free survival (PFS) or OS in resected early-stage disease (24). Patients with early recurrence risk are usually associated with negative prognostic factors, whereas those at risk of late recurrence are always associated with advanced liver disease, such as tumor formation (25). In fact, some studies have shown that adjuvant systemic therapy can reduce local and distant recurrence rates; however, more recent studies have not demonstrated this effect (26).

Previously, Ohata et al (8) evaluated whether adjuvant systemic treatment could improve survival in early-stage HCC, and the STORM trial evaluated the effect of adjuvant sorafenib in HCC. In this phase 3, double-blind study, 1,114 patients were randomly assigned to placebo or sorafenib-stimulated groups, who were treated with 400 mg twice a day for up to 4 years. However, relapse-free survival was not significantly different between the two groups (27). Based on the evidence, it was considered that the efficacy of anticancer agents cannot translate from the advanced to the adjuvant setting. Unfortunately, effective regimens thus far have not been found. At the same time, because TKIs need an optional ‘target’ to perform their activity, it was suggested that these drugs in the adjuvant setting are probably far from being rational (10). Therefore, immunotherapy is considered to be a promising approach, and patients should be encouraged to enroll in clinical trials evaluating immune-based combinations or ICI monotherapy as adjuvant treatment to reduce the risk of recurrence and improve clinical outcomes.

REG, an orally bioavailable MKI, blocks the activity of several protein kinases, including KIT, BRAF, RAF-1, RET, VEGFR-1, VEGFR-2, VEGFR-3, PDGFR and FGFR, which are associated with tumor microenvironment signaling, tumor angiogenesis, and cell proliferation (28,29). Currently, REG is approved as a single agent for the treatment of HCC at a dose of 160 mg orally once daily on days 1–21 of each 28-day cycle (30). There are currently ongoing trials aimed at evaluating the efficacy of REG as monotherapy or in combination with other anticancer agents, and the number of patients receiving REG is expected to increase in the future (31,32). Therefore, it was suggested that REG dose personalization may improve quality of life, decrease treatment adverse events, and enhance patient outcomes.

ICIs are able to influence immune checkpoint-related molecules, such as programmed cell death-1 (PD-1), cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), and lymphocyte-activation gene 3 (LAG-3) (33,34). However, ICI monotherapy has demonstrated disappointing effects thus far in patients with HCC (35,36). Recently, combined immunotherapy with phase III IMbrave150 and the PD-L1 inhibitor atezolizumab plus anti-angiogenesis bevacizumab showed more convincing effects than monotherapy in advanced HCC patients (37,38). Moreover, patients receiving another combined immunotherapy, including atezolizumab and bevacizumab, had impressive benefits in PFS, OS, objective response rate, and complete response rate, and the median OS could reach 19.2 months (39). However, to date, few molecules have been assessed as predictors, such as programmed death ligand-1 (PD-L1), tumor mutational burden, and gut microbiota, to evaluate whether these therapeutic regimens benefit patients with HCC (40). Hence, it was suggested by the authors that a useful marker is needed to evaluate whether monotherapy or combined immunotherapy benefits patients with HCC.

The review entitled ‘The Roles of Protein Tyrosine Phosphatases (PTP) in HCC’ published in 2018, reported both oncogenic and tumor suppressive function of PTPs in HCC. Huang et al (41) mainly reviewed the involvement of PTP and associated signaling pathways in HCC. However, the present review mainly focused on discussing the relationship of PTPs with inflammatory cytokines and chemotherapy/targeted drug resistance, providing detailed information on how PTPs can modulate inflammatory reactions and drug resistance to influence progression in HCC. The roles of inflammatory cytokines and drug resistance modulated by PTPs have never been displayed previously. Therefore, it was suggested that the PTP-regulated inflammatory cytokines and drug resistance are critical in HCC, and PTPs may be therapeutic targets in the future. In the present review, numerous kinds of PTPs associated with cancer progression were discussed, including those associated with inflammatory reactions and drug resistance in HCC.

The PTP family

Class I PTPs include ~100 proteins, which can be separated into two groups based on their interaction residues: classical PTPs and dual-specificity phosphatases (DSPs). The classical PTPs include ~38 proteins; these proteins are dispersed in the cytoplasm (non-transmembrane PTPs) and cellular membrane (receptor-like PTPs), and the classical PTPs are involved in a broad range of pathways connecting intracellular and extracellular components (42).

Tyrosine phosphate groups are the general site of dephosphorylation of classical PTPs. By contrast, DSPs have three phosphate-related areas, including tyrosine, serine and threonine residues. In addition to DSPs having smaller catalytic domains than classical PTPs, there are other major differences between classical PTPs and DSPs. Most DSPs are localized in the cytoplasm, and due to the diverse phosphate residues involved in the dephosphorylation process, DSPs regulate various cellular biogenesis processes, including the cell cycle, metabolism and neuron transduction (43). Currently, DSPs are categorized into several subclasses, such as mitogen-activated protein kinase (MAPK) phosphatases (MPKs, 11 members), protein phosphates slingshot homologs (3 members), phosphatases of regenerating liver (PRLs, 3 members), phosphate and tension homologs (PTEN, 5 members), atypical DSPs (19 members), myotubularins (16 members), and CDC14s (4 members) (Table I) (43,44). To date, certain of these DSP subclasses have been revealed to potentially regulate pathways in multiple diseases, particularly cancer. The MAPK/extracellular signal-regulated kinase (ERK) pathway is one of the most important pathways, and its members have been identified to be overexpressed or mutated in over 90% of cancers. The target phosphatases of this pathway are MPKs. Moreover, cancer cells are able to regulate CDK-induced cell cycle progression through regulation of CDC14s; a previous study has revealed upregulation of certain PRLs and downregulation in certain PTEN members and myotubularins (45). Most class I PTPs induce profound effects in cancers, while the characteristics of other genes and proteins remain unclear (44). Low-molecular-weight phosphatase, also called ACP1, is the only member of the class II PTPs, and it has been demonstrated to be associated with platelet-derived growth factor signaling (46).

Table I.

The subclasses of DSPs.

Table I.

The subclasses of DSPs.

SubclassSymbolMember
Mitogen-activated protein kinase phosphatasesMPKs11
Phosphatases of regenerating liverPRLs  3
Phosphate and tension homologsPTEN  5
Atypical DSPs 19
Myotubularins 16
CDC14s   4

[i] DSPs, dual-specificity phosphatases.

PTP mechanisms

PTPs are multifunctional enzymes mainly localized in the cytoplasm and cellular membrane. These enzymes play an essential role in protein post-translational modification, which affects protein activity and stability (47,48). A common mechanism of PTPs is to recognize the phosphorylation residues on tyrosine, serine, and threonine residues of target proteins to induce the removal of phosphate groups, leading to the dephosphorylation and inactivation of kinases or receptors (49). Two PTP domains participate in this process, the PTP-loop and WPD-loop. In the initiation of the phosphorylation catalytic process, the cysteine catalytic site on the PTP-loop competes for the binding of the phosphate group and the target protein, which results in breaking of the phosphate-oxygen bond and dephosphorylation of the target residue. Concurrently, the WPD loop stabilizes the remaining structure. In the further steps of the catalytic process, the phosphate group is released via cooperation of the P-loop and WPD-loop (50).

Protein tyrosine phosphorylation is a pivotal process for signal transduction in eukaryotic cells and is a reversible regulatory mechanism that is coordinately controlled by protein tyrosine kinases (PTKs) and PTPs (51). Imbalance of the PTK-PTP axis often results in aberrant protein tyrosine phosphorylation in cancers and promotes tumorigenesis, as is observed in HCC (52). PTPs usually play tumor-suppressor roles, whereas PTKs are mainly associated with oncogenic and tumorigenic activities (53). Regulation of RTKs and PTPs, which occurs by reversible alteration of the phosphorylation state of specific tyrosine kinases, leads to various cellular events, such as alterations in the signaling pathway activity and cellular phenotypes (54).

Association between PTPs and cytokines in HCC

Cytokines have been identified as potential factors in cancer progression and are components of the tumor microenvironment. Cytokines include a broad range of extracellular molecules, such as chemokines, interleukins, interferons and tumor necrosis factors (TNFs). They are generally produced in the extracellular environment by immune cells, endothelial cells and stromal cells and regulate downstream cellular signaling by binding to specific receptors (55). Due to the enormous variety of cytokines and their varied characteristics, the final cellular effects of cytokines are usually substantial and long lasting. Cytokines can also increase cancer cell malignancy by modifying cellular proliferation, migration and invasion (55). Several studies have demonstrated that some cytokines and PTPs interact in HCC progression. PTPN6 interacts with gankyrin, an oncoprotein, leading to the secretion of IL-6, which induces signal transducer and activator of transcription 3 (STAT3) phosphorylation and promotes HCC development (56); Sakurai et al (56) reported that gankyrin can interact with PTPN6 to induce STAT3 activation and IL-6 secretion. Moreover, gankyrin can also increase VEGF expression, which leads to HCC progression (Fig. 1A, Table II) (56). however, more studies have identified PTPs as potential candidates for reversing cytokine-induced HCC progression. PTP receptor type delta (PTPRD) is a tumor suppressor that is negatively correlated with PD-L1, an essential molecule in cancer immune escape. Overexpression of PTPRD is able to reduce PD-L1 through the STAT3 pathway activation (Table II) (57). In addition, the effects of combination therapies also involve PTP-cytokine relationships. Induction of SHP-1 through crocin decreases the IL-6-stimulated STAT3 pathway, resulting in HCC cell apoptosis (58). Quercetin enhances IFN-α-induced phosphorylation of STAT1 by downregulating SHP2, leading to an anti-proliferative effect in HCC (59).

Table II.

Association between PTPs and cytokines.

Table II.

Association between PTPs and cytokines.

NameMechanismPathwayEffect
PTPN6Interaction with gankyrin ↑STAT3 phosphorylation ↑IL-6 secretion ↑
PTPRDCorrelation with PD-1 ↓STAT3 pathway ↑PD-L1 ↓
CDC25AIL-6 and IL-1β ↓ Cell cycle arrest
PTPRO JAK2/STAT3 ↓IL-6 secretion ↑ PD-L1 ↓
PTPN11 RAS/ERK pathway/Integrin signaling ↑HCC progression
PTP1BIL-1β, IL-6 and TNF-α ↑NF-κB pathway ↑Inflammatory response ↑

[i] ↑, induction; ↓, reduction; PTP, protein tyrosine phosphatase; HCC, hepatocellular carcinoma; PTPRO, protein tyrosine phosphatase receptor type O.

In addition to classical PTPs, DSPs are a subgroup of tyrosine phosphatases in the PTP family, and these enzymes function in the removal of a wide range of phosphate groups from not only tyrosine residues but also serine/threonine residues (44). Similar to classical PTPs, DSPs are involved in broad signaling transduction in the regulation of the development of both normal and cancer cells, and these proteins also exhibit potential relationships with cytokines (60). Sorafenib, the most common targeted therapy in HCC treatment, induces DSP1 expression and reduces TGF-β expression in macrophages, potentially promoting HCC progression (61). Moreover, knockdown of CDC25A decreases the expression of IL-6 and IL-1β, leading to significant cell cycle arrest in the G1 phase in HCC (62).

Previously, PTP receptor type O (PTPRO) and its truncated form (PTPROt) were defined as negative regulators of JAK2/STAT3 signaling (63). Hou et al (63) reported that PTPRO downregulates STAT3 activation via the JAK2 and PI3K signaling pathways. Therefore, the effect of PTPRO on HCC development may result from STAT3 activation. Numerous studies have reported that the poor therapeutic effect of PD-1 adjuvant treatment is highly associated with an increased level of IL-6 in serum. Anti-PD-1/PD-L1 antibody combined with anti-IL-6 antibody treatment has demonstrated significant curative effects in animal models (64). Therefore, IL-6 may have a crucial impact on the effect of PD-1/PD-L1 adjuvant therapy. JAK2 has been demonstrated to interact with PTPRO upon IL-6 stimulation by a coimmunoprecipitation assay, indicating that PTPRO may modulate JAK2 (Fig. 1B, Table II). PD-L1 expression in monocytes and macrophages has been demonstrated to be suppressed by PTPRO through downregulation of the JAK2/STAT1 and JAK2/STAT3/c-MYC cascades (65).

PD-L1 expression was significantly increased in PTPRO-expressing macrophages in an IFN-γ-dependent manner after IL-6 treatment for 72 h. This result could indicate that PTPRO expression is essential for IL-6-induced Pd-L1/PD-L1 expression in both Ptpro knockout and PTPRO knockdown macrophages. Moreover, signaling pathway analysis indicated that the action of PTPRO is dysregulated by IL-6 via increased activation of the STAT3/c-MYC/PD-L1 axis in monocytes and macrophages. Treatment of U937- and THP-1-derived macrophages with c-MYC shRNA reversed the IL-6-induced decrease in PTPRO expression (65). IL-6 is secreted by both T cells and macrophages and is a classic proinflammatory cytokine. IL-6 is a key cytokine linking inflammation to tumorigenesis in numerous cancers, including HCC (66,67). Moreover, Naugler et al (68) also reported that IL-6 is an essential cytokine linking inflammation and tumorigenesis. IL-6 has been demonstrated to play an oncogenic role in obesity-related HCC (69).

The nonreceptor PTP SHP2, encoded by PTP, nonreceptor type 11 (PTPN11), is a critical member of the RAS/ERK pathway and most receptor tyrosine kinase, cytokine receptor, and integrin signaling pathways (70). Several lines of evidence have indicated that PTPN11 is involved in HCC progression (71). In addition, several studies have shown that PTPN11 can also play an unexpected tumor suppressor role in HCC (72,73), implying that PTPN11 possesses dual roles in tumorigenesis.

PTPRD, a member of the PTP family, has been reported to act as a tumor suppressor gene and plays a crucial role in controlling numerous cellular processes, including cell proliferation, apoptosis, survival and motility (74). PTPRD is often inactivated via deletion or epigenetic mechanisms in several cancers (75). STAT3, a major transcription factor, is involved in numerous cellular processes, such as cell growth, proliferation, migration, differentiation and death (76). Previously, STAT3 has been demonstrated to positively regulate PD-L1 expression to promote immune escape in cancer (77). Furthermore, Meng et al (57) reported that PTPRD expression was significantly lower in tumor tissues than in normal tissues; however, PD-L1 was significantly overexpressed in cancer tissues compared with normal tissues.

Additional studies have also shown that silencing PTP1B decreases the inflammatory response and levels of associated cytokines, including IL-1β, IL-6 and TNF-α, while overexpression of PTP1B induces inflammation in RAW264.7 cells. Moreover, lipopolysaccharide can activate the NF-κB pathway in RAW264.7 cells, and NF-κB signaling is also affected by dysregulated PTP1B expression (Fig. 1C, Table II) (78).

The nonreceptor PTP Src homology region 2 (SH2) domain-containing phosphatases (SHPs), including SHP-1 (also known as PTPN6) and SHP-2 (also known as PTPN11), are critical modulators of numerous fundamental cellular processes, such as cell proliferation, differentiation and inflammation (79). SHP-2 is a ubiquitously expressed modulator of inflammatory reactions and is implicated in HCC carcinogenesis and progression (80). In addition, SHP-1 is extensively expressed in hematopoietic and epithelial cells and is widely defined as a negative regulator of inflammation (81). Several studies have reported that MKIs, such as sorafenib (82) and dovitinib (83), exert their antitumor effects by enhancing SHP-1 phosphatase activity. TGF-β1-induced STAT3 (Tyr705) phosphorylation and epithelial-to-mesenchymal transition can be abolished with SHP-1 overexpression, which blocks cell migration and invasion of HCC (84). Moreover, SHP-1 has been demonstrated to be overexpressed in non-cancer tissues compared with surrounding cancer tissues, and reduced SHP-1 expression is highly associated with poor prognosis of patients with HCC (85). Collectively, SHP-1 can be defined as a tumor suppressor that prevents the initiation and progression of HCC in animal models (85). SHP-1 can also inhibit the activation of various signaling pathways, such as the STAT3, NF-kB, and AKT pathways, to suppress hepatocarcinogenesis and the malignant phenotype of HCC (85). Several drugs, including sorafenib, dovitinib, and SC-2001, induce cell apoptosis and inhibit the growth of HCC cells by enhancing the activity of SHP-1 tyrosine phosphatase (82,86). SHP-1 and SHP-2, cytoplasmic PTPs, share similar sequences, containing two Src homology 2 (SH2) NH2-terminal domains and a C-terminal protein-tyrosine phosphatase domain (87). SHP-2 reduces STAT3 phosphorylation via the JAK/STAT pathway to suppress HCC initiation. By contrast, SHP-2 coordinately activates the Ras/Raf/Erk and PI3K/Akt/mTOR cascades to promote the progression of HCC (80). Liver inflammation, a primary oncogenic factor, is highly associated with HCC (88,89). The inflammatory cytokines induced by liver injury activate inflammatory signaling pathways, including the JAK/STAT and NF-kB signaling pathways, which in turn induce the expression of IL6, TGF-β, and TNF-α (90,91).

The relationship between PTPs and drug resistance

SHP2 is related to the stress sensor DNA damage 45G (GADD45G), which is involved in multiple biological processes and downregulated in various cancers. GADD45G has been demonstrated to induce senescence in HCC and reduce tumor growth in vivo. Moreover, GADD45G-induced senescence can be efficiently counteracted with Shp2 silencing. GADD45G expression is negatively correlated with the phosphorylation status of STAT3 in tumor cells of clinical HCC specimens (Table III) (92); this result is related to the relationship of SHP2 with STAT3 (93,94). Dovitinib downregulation of p-STAT3 and induction of apoptosis can be abolished by using an SHP-1 inhibitor or silencing SHP-1 with RNA interference, suggesting that SHP-1, a PTP, modulates the effects of dovitinib. In addition, dovitinib reduced STAT3 activation to induce cell apoptosis in two sorafenib-resistant cell lines, and sorafenib-resistant cells showed significant activation of STAT3, indicating that STAT3 may be a useful target to overcome drug resistance in HCC (54). JAK/STAT3 signaling is inactivated by several PTPs, including the SH2 domain-containing cytosolic phosphatases SHP-1 and SHP-2 (95,96). Furthermore, SHP-1 has been demonstrated to be involved in the dovitinib-mediated effect on kinase inhibition, phosphorylated (p)-STAT3 and apoptosis in HCC (54). Additionally, dovitinib suppressed tumor growth in both Huh-7 and PLC5 ×enograft tumors in vivo, suggesting the potential utility of dovitinib in the clinical practice. Therefore, an understanding of the mechanism of SHP-1-mediated STAT3 inhibition provides a potential target for future HCC molecular therapy (Fig. 2A, Table III) (54).

Table III.

The relationship between PTPs and drug resistance.

Table III.

The relationship between PTPs and drug resistance.

PTPsMarkersPathwayEffect
PTPN6p-JAK1, p-JAK2, Mcl-1, and cyclin D1JAK/STAT3 pathway ↑Apoptosis ↑ HCC progression ↑
PTPN11GADD45GSTAT3 pathway ↑Senescence ↑
MEK/ERK pathways ↑Tumor growth ↓
NF-κB pathway ↑
PI3K/AKT/mTOR pathway ↑
PTPRO STAT3 pathway ↓HCC development ↓
LC3II/I, p62PI3K pathway ↓Autophagy ↓

[i] ↑, induction; ↓, reduction; PTP, protein tyrosine phosphatase; p-, phosphorylated; HCC, hepatocellular carcinoma.

SHP2, encoded by PTPN11, was found to not only be overexpressed in HCC (97) but also serve as a potential predictive biomarker for sorafenib response and patient survival (97). Moreover, SHP2 has been defined as a downstream effector of numerous RTKs, and SHP2 blockade may be a possible mechanism causing RTK activation, resulting in the development of acquired resistance to sorafenib in HCC (98). Collectively, sorafenib-induced reactivation of the RTK-mediated AKT and MEK/ERK pathways can be significantly induced by SHP099 (98). Targeting SHP2 with SHP099 combined with sorafenib treatment may be a novel and safe therapeutic strategy against HCC (99). Previously, Kang et al (100) reported that the RNA level of SHP2 is upregulated through the NF-κB signaling pathway in HBX-transfected HCC cells. Mechanistically, SHP2 expression is induced by direct binding of NF-κB to its promoter. Since NF-κB signaling has been implicated in HCC progression (100) and sorafenib resistance (101), its activation may be a potent mechanism leading to SHP2 upregulation in both parental and sorafenib-resistant HCC cells. Previously, SHP2 was identified as an oncogenic tyrosine phosphatase that contains two Src-homology 2 domains (102). SHP2 has been reported to be a critical component of multiple RTK signaling pathways activated in response to numerous growth factors, including FGFR, EGFR, PDGFR, and VEGFR, and this activation leads to induction of the PI3K/AKT/mTOR pathway and ERK signaling based on genetic and biochemical evidence (103,104). SHP2 is required for RTK-evoked RAS activation, which results in the activation of the MEK/ERK and AKT pathways (Fig. 2B, Table III) (105).

SHP-1 is a PTP that is largely expressed in hematopoietic cells. To date, several studies have addressed the role of SHP-1 in tumor progression, and a few studies have suggested that SHP1 plays a potential tumor suppressor role in various cancer types (106). Moreover, impaired function of SHP-1 has been shown to induce cancer progression by downregulating intracellular signaling transmembrane receptors, such as growth factor and cytokine receptors, leading to abnormal pathologies (107). Upregulation of SHP-1 activity induces cell apoptosis both in vitro and in vivo (54). In addition, Tai et al (54) reported that STAT3-related kinases or downstream effectors, including p-JAK1, p-JAK2, Mcl-1, and cyclin D1, are also induced in sorafenib-resistant cells. Evidence has shown that the JAK/STAT3 signaling pathway is a crucial modulator of the efficacy of sorafenib. Notably, decreased expression of SHP-1 was also observed in sorafenib-resistant cells. Collectively, the role of SHP-1-related STAT3 signaling in HCC has been verified; therefore, the SHP-1/STAT3 pathway may be an effective target for HCC treatment (54). SHP-1-mediated dephosphorylation of PKM2 at Y105 results in increased activity, and tetrameric PKM2 has reduced nuclear localization, which leads to the downregulation of the expression of oncogenic molecules, such as c-Myc and cyclin D1. Furthermore, constitutively active SHP-1 (D61A) can increase the percentage of tetrameric PKM2 and phosphorylation of PKM2 (Y105F), suggesting that SHP-1 determines the levels of dimeric/tetramer PKM2 and the subsequent nuclear localization via PKM2 Y105 dephosphorylation (108). SHP-1 (PTPN6), first identified in hematopoietic cells, is implicated in various hematopoietic signaling processes, including integration of immunoreceptor tyrosine-based activation motif-mediated inhibitory signals (109) and B-cell and natural killer (NK)-cell development (110,111). Furthermore, gankyrin is upregulated in chronic inflammation and induces STAT3 activation and IL-6 secretion by interacting with SHP-1 in non-parenchymal cells. Such proinflammatory interactions may induce the levels of stem cell markers in the tumor microenvironment and eventually promote HCC progression. Thus, the expression of gankyrin is defined as a promising predictor of the efficacy of advanced treatment for patients with HCC (56). Previously, gankyrin was demonstrated to enhance hepatocarcinogenesis via STAT3 activation through SHP-1 inhibition and IL-6 upregulation in the tumor microenvironment. Thus, STAT3/IL-6 signaling may involve gankyrin-regulated crosstalk between tumor cells and nonparenchymal cells (56).

PTPRO is a receptor type of PTP that has been defined as an integral membrane protein in numerous parenchymal cells (including lung, liver, and breast cells) (112,113). It has been previously demonstrated by the authors that PTPRO can suppress STAT3 activation, leading to reduced development of HCC. PTPRO can negatively regulate important pathways related to autophagy, such as the PI3K signaling pathway (63,114). Moreover, Zhang et al (115) reported that ptpro−/− hepatocytes lead to the development of steatosis and induce tumorigenesis in mice fed a high-fat diet (HFD). PTPRO deletion significantly augmented obesity-reduced autophagy, as evidenced by increased p62 expression and a reduced LC3II/I ratio, as revealed by western blotting. Collectively, evidence confirmed that PTPRO deletion promotes obesity-related hyperinsulinemia and autophagy deficiency in the liver (Fig. 2C, Table III) (115). Furthermore, evidence suggested that PTPRO increases the cytoplasmic accumulation of p53 through the PI3K/Akt/MDM axis (115). Additionally, the expression of PTPRO has been demonstrated to be significantly reduced in HCC compared with normal tissues (63). PTPRO expression is suppressed in vivo in mice fed an HFD compared with that in mice fed a normal diet (116,117). PTPRO regulates autophagy and lipid metabolism in obesity and steatohepatitis (115). Furthermore, PTPRO regulates lipid metabolism through reduced expression of lipogenesis genes and induction of β-oxidation-related gene expression, and obesity significantly induces tumorigenesis in the liver in ptpro−/− mice (115).

In the present review, extensive information was provided discussing whether PTPs play a critical role in inflammatory reactions and drug resistance to influence cancer progression in HCC. Numerous inflammatory cytokines/chemokines modulated by PTPs and several chemotherapeutic and targeted therapeutic drugs were illustrated, that are likely related to PTPs that play critical roles in numerous cellular mechanisms and signaling pathways. A total of 3 PTPs involved in HCC drug resistance were listed, including PTPN6, PTPN11 and PTPRO. In addition, there are six PTPs, PTPN6, PTPRD, CDC25A, PTPRO, PTPN11 and PTP1B. Among these, it was found that the three PTPs PTPN6, PTPN11 and PTPRO both induce drug resistance and alter inflammatory cytokine regulation, and these molecules can influence tumor growth and HCC progression. Hence, it was suggested that the three PTPs PTPN6, PTPN11 and PTPRO play equally important roles in HCC progression. The present review provided practical information for researchers to understand in an improved way the roles and functions of PTPs in cancer progression and hence may aid the identification of further therapeutic options to cure cancer.

According to findings from other groups about the roles of PTPs, it was suggested by the authors that PTPs play roles in inflammatory cytokines and drug resistance. In the future, it is considered by the authors that the effects of PTPs may be applied to clinical practice to evaluate whether PTP molecules could be useful predictors in patients with HCC. It was hypothesized that the levels of PTPs and inflammatory cytokines, such as IL-1β and IL-6, in HCC patients with chemotherapy/targeted drug resistance could be detected in tissues and plasma by immunohistochemistry and ELISA, respectively. Finally, the correlations between the levels of PTPs and IL-1β and IL-6 can also be analyzed to determine whether the correlations and these molecules could be markers to predict the prognosis and survival rate of drug-resistant patients.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Ministry of Science and Technology of the Republic of China (grant nos. MOST 109-2320-B-006-067. and MOST 110-2320-B-039-044).

Availability of data and materials

Not applicable.

Authors' contributions

Y-LC, J-YC, Y-CH and C-YC performed formal analysis. Y-LC, C-CH, J-YC and C-YC prepared the original draft of the manuscript. C-CH, P-MC, Y-CH and C-Y wrote, reviewed and edited the manuscript. P-MC and C-YC acquired funding. All authors have read and approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Ringelhan M, Pfister D, O'Connor T, Pikarsky E and Heikenwalder M: The immunology of hepatocellular carcinoma. Nat Immunol. 19:222–232. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Yang JD and Roberts LR: Hepatocellular carcinoma: A global view. Nat Rev Gastroenterol Hepatol. 7:448–458. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Bosch FX, Ribes J, Diaz M and Cleries R: Primary liver cancer: Worldwide incidence and trends. Gastroenterology. 127 (5 Suppl 1):S5–S16. 2004. View Article : Google Scholar : PubMed/NCBI

4 

Morgan TR, Mandayam S and Jamal MM: Alcohol and hepatocellular carcinoma. Gastroenterology. 127 (5 Suppl 1):S87–S96. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Caldwell SH, Crespo DM, Kang HS and Al-Osaimi AM: Obesity and hepatocellular carcinoma. Gastroenterology. 127 (5 Suppl 1):S97–S103. 2004. View Article : Google Scholar : PubMed/NCBI

6 

Ming L, Thorgeirsson SS, Gail MH, Lu P, Harris CC, Wang N, Shao Y, Wu Z, Liu G, Wang X and Sun Z: Dominant role of hepatitis B virus and cofactor role of aflatoxin in hepatocarcinogenesis in Qidong, China. Hepatology. 36:1214–1220. 2002. View Article : Google Scholar : PubMed/NCBI

7 

Yu MC and Yuan JM: Environmental factors and risk for hepatocellular carcinoma. Gastroenterology. 127 (5 Suppl 1):S72–S78. 2004. View Article : Google Scholar : PubMed/NCBI

8 

Ohata K, Hamasaki K, Toriyama K, Matsumoto K, Saeki A, Yanagi K, Abiru S, Nakagawa Y, Shigeno M, Miyazoe S, et al: Hepatic steatosis is a risk factor for hepatocellular carcinoma in patients with chronic hepatitis C virus infection. Cancer. 97:3036–3043. 2003. View Article : Google Scholar : PubMed/NCBI

9 

Spangenberg HC, Thimme R and Blum HE: Targeted therapy for hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol. 6:423–432. 2009. View Article : Google Scholar : PubMed/NCBI

10 

El-Serag HB and Rudolph KL: Hepatocellular carcinoma: Epidemiology and molecular carcinogenesis. Gastroenterology. 132:2557–2576. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Suriawinata A and Xu R: An update on the molecular genetics of hepatocellular carcinoma. Semin Liver Dis. 24:77–88. 2004. View Article : Google Scholar : PubMed/NCBI

12 

Satyanarayana A, Manns MP and Rudolph KL: Telomeres and telomerase: A dual role in hepatocarcinogenesis. Hepatology. 40:276–283. 2004. View Article : Google Scholar : PubMed/NCBI

13 

Brechot C: Pathogenesis of hepatitis B virus-related hepatocellular carcinoma: Old and new paradigms. Gastroenterology. 127 (5 Suppl 1):S56–S61. 2004. View Article : Google Scholar : PubMed/NCBI

14 

Ahn JH, Kim SJ, Park WS, Cho SY, Ha JD, Kim SS, Kang SK, Jeong DG, Jung SK, Lee SH, et al: Synthesis and biological evaluation of rhodanine derivatives as PRL-3 inhibitors. Bioorg Med Chem Lett. 16:2996–2999. 2006. View Article : Google Scholar : PubMed/NCBI

15 

Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, de Oliveira AC, Santoro A, Raoul JL, Forner A, et al: Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 359:378–390. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Zhu YJ, Zheng B, Wang HY and Chen L: New knowledge of the mechanisms of sorafenib resistance in liver cancer. Acta Pharmacol Sin. 38:614–622. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Villanueva A: Hepatocellular Carcinoma. N Engl J Med. 380:1450–1462. 2019. View Article : Google Scholar : PubMed/NCBI

18 

Granito A and Bolondi L: Non-transplant therapies for patients with hepatocellular carcinoma and Child-Pugh-Turcotte class B cirrhosis. Lancet Oncol. 18:e101–e112. 2017. View Article : Google Scholar : PubMed/NCBI

19 

Tovoli F, Negrini G and Bolondi L: Comparative analysis of current guidelines for the treatment of hepatocellular carcinoma. Hepat Oncol. 3:119–136. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Tovoli F, Lorenzo S, Barbera MA, Garajova I, Frega G, Palloni A, Pantaleo MA, Biasco G and Brandi G: Postsorafenib systemic treatments for hepatocellular carcinoma: Questions and opportunities after the regorafenib trial. Future Oncol. 13:1893–1905. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Pasquier E, Kavallaris M and Andre N: Metronomic chemotherapy: New rationale for new directions. Nat Rev Clin Oncol. 7:455–465. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Kareva I, Waxman DJ and Lakka Klement G: Metronomic chemotherapy: An attractive alternative to maximum tolerated dose therapy that can activate anti-tumor immunity and minimize therapeutic resistance. Cancer Lett. 358:100–106. 2015. View Article : Google Scholar : PubMed/NCBI

23 

De Lorenzo S, Tovoli F, Barbera MA, Garuti F, Palloni A, Frega G, Garajovà I, Rizzo A, Trevisani F and Brandi G: Metronomic capecitabine vs. best supportive care in Child-Pugh B hepatocellular carcinoma: A proof of concept. Sci Rep. 8:99972018. View Article : Google Scholar : PubMed/NCBI

24 

Personeni N and Rimassa L: Hepatocellular carcinoma: A global disease in need of individualized treatment strategies. J Oncol Pract. 13:368–369. 2017. View Article : Google Scholar : PubMed/NCBI

25 

Akateh C, Black SM, Conteh L, Miller ED, Noonan A, Elliott E, Pawlik TM, Tsung A and Cloyd JM: Neoadjuvant and adjuvant treatment strategies for hepatocellular carcinoma. World J Gastroenterol. 25:3704–3721. 2019. View Article : Google Scholar : PubMed/NCBI

26 

Rizzo A, Mollica V, Ricci AD, Maggio I, Massucci M, Rojas Limpe FL, Fabio FD and Ardizzoni A: Third- and later-line treatment in advanced or metastatic gastric cancer: A systematic review and meta-analysis. Future Oncol. 16:4409–4418. 2020. View Article : Google Scholar : PubMed/NCBI

27 

Bruix J, Takayama T, Mazzaferro V, Chau GY, Yang J, Kudo M, Cai J, Poon RT, Han KH, Tak WY, et al: Adjuvant sorafenib for hepatocellular carcinoma after resection or ablation (STORM): A phase 3, randomised, double-blind, placebo-controlled trial. Lancet Oncol. 16:1344–1354. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Wilhelm SM, Dumas J, Adnane L, Lynch M, Carter CA, Schütz G, Thierauch KH and Zopf D: Regorafenib (BAY 73-4506): A new oral multikinase inhibitor of angiogenic, stromal and oncogenic receptor tyrosine kinases with potent preclinical antitumor activity. Int J Cancer. 129:245–255. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Abou-Elkacem L, Arns S, Brix G, Gremse F, Zopf D, Kiessling F and Lederle W: Regorafenib inhibits growth, angiogenesis, and metastasis in a highly aggressive, orthotopic colon cancer model. Mol Cancer Ther. 12:1322–1331. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Rimassa L, Pressiani T, Personeni N and Santoro A: Regorafenib for the treatment of unresectable hepatocellular carcinoma. Expert Rev Anticancer Ther. 17:567–576. 2017. View Article : Google Scholar : PubMed/NCBI

31 

Cerrito L, Ponziani FR, Garcovich M, Tortora A, Annicchiarico BE, Pompili M, Siciliano M and Gasbarrini A: Regorafenib: A promising treatment for hepatocellular carcinoma. Expert Opin Pharmacother. 19:1941–1948. 2018. View Article : Google Scholar : PubMed/NCBI

32 

Liu Z, Lin Y, Zhang J, Zhang Y, Li Y, Liu Z, Li Q, Luo M, Liang R and Ye J: Molecular targeted and immune checkpoint therapy for advanced hepatocellular carcinoma. J Exp Clin Cancer Res. 38:4472019. View Article : Google Scholar : PubMed/NCBI

33 

Ingles Garces AH, Au L, Mason R, Thomas J and Larkin J: Building on the anti-PD1/PD-L1 backbone: Combination immunotherapy for cancer. Expert Opin Investig Drugs. 28:695–708. 2019. View Article : Google Scholar : PubMed/NCBI

34 

Alsaab HO, Sau S, Alzhrani R, Tatiparti K, Bhise K, Kashaw SK and Iyer AK: PD-1 and PD-L1 checkpoint signaling inhibition for cancer immunotherapy: Mechanism, combinations, and clinical outcome. Front Pharmacol. 8:5612017. View Article : Google Scholar : PubMed/NCBI

35 

Cheng AL, Hsu C, Chan SL, Choo SP and Kudo M: Challenges of combination therapy with immune checkpoint inhibitors for hepatocellular carcinoma. J Hepatol. 72:307–319. 2020. View Article : Google Scholar : PubMed/NCBI

36 

Pinter M, Jain RK and Duda DG: The current landscape of immune checkpoint blockade in hepatocellular carcinoma: A review. JAMA Oncol. 7:113–123. 2021. View Article : Google Scholar : PubMed/NCBI

37 

Finn RS, Qin S, Ikeda M, Galle PR, Ducreux M, Kim TY, Kudo M, Breder V, Merle P, Kaseb AO, et al: Atezolizumab plus bevacizumab in unresectable hepatocellular carcinoma. N Engl J Med. 382:1894–1905. 2020. View Article : Google Scholar : PubMed/NCBI

38 

Kelley RK: Atezolizumab plus Bevacizumab-A landmark in liver cancer. N Engl J Med. 382:1953–1955. 2020. View Article : Google Scholar : PubMed/NCBI

39 

Rizzo A, Ricci AD and Brandi G: Atezolizumab in advanced hepatocellular carcinoma: Good things come to those who wait. Immunotherapy. 13:637–644. 2021. View Article : Google Scholar : PubMed/NCBI

40 

Sangro B, Sarobe P, Hervas-Stubbs S and Melero I: Advances in immunotherapy for hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol. 18:525–543. 2021. View Article : Google Scholar : PubMed/NCBI

41 

Huang Y, Zhang Y, Ge L, Lin Y and Kwok HF: The roles of protein tyrosine phosphatases in hepatocellular carcinoma. Cancers (Basel). 10:822018. View Article : Google Scholar : PubMed/NCBI

42 

Hendriks WJ, Elson A, Harroch S, Pulido R, Stoker A and den Hertog J: Protein tyrosine phosphatases in health and disease. FEBS J. 280:708–730. 2013. View Article : Google Scholar : PubMed/NCBI

43 

Pulido R and Hooft van Huijsduijnen R: Protein tyrosine phosphatases: Dual-specificity phosphatases in health and disease. FEBS J. 275:848–866. 2008. View Article : Google Scholar : PubMed/NCBI

44 

Alonso A, Sasin J, Bottini N, Friedberg I, Friedberg I, Osterman A, Godzik A, Hunter T, Dixon J and Mustelin T: Protein tyrosine phosphatases in the human genome. Cell. 117:699–711. 2004. View Article : Google Scholar : PubMed/NCBI

45 

Buj-Bello A, Laugel V, Messaddeq N, Zahreddine H, Laporte J, Pellissier JF and Mandel JL: The lipid phosphatase myotubularin is essential for skeletal muscle maintenance but not for myogenesis in mice. Proc Natl Acad Sci USA. 99:15060–15065. 2002. View Article : Google Scholar : PubMed/NCBI

46 

Chiarugi P, Cirri P, Marra F, Raugei G, Fiaschi T, Camici G, Manao G, Romanelli RG and Ramponi G: The Src and signal transducers and activators of transcription pathways as specific targets for low molecular weight phosphotyrosine-protein phosphatase in platelet-derived growth factor signaling. J Biol Chem. 273:6776–6785. 1998. View Article : Google Scholar : PubMed/NCBI

47 

Hunter T and Sefton BM: Transforming gene product of Rous sarcoma virus phosphorylates tyrosine. Proc Natl Acad Sci USA. 77:1311–1315. 1980. View Article : Google Scholar : PubMed/NCBI

48 

Cohen P: Protein kinases-the major drug targets of the twenty-first century? Nat Rev Drug Discov. 1:309–315. 2002. View Article : Google Scholar : PubMed/NCBI

49 

Tanner JJ, Parsons ZD, Cummings AH, Zhou H and Gates KS: Redox regulation of protein tyrosine phosphatases: Structural and chemical aspects. Antioxid Redox Signal. 15:77–97. 2011. View Article : Google Scholar : PubMed/NCBI

50 

Zhang ZY: Protein tyrosine phosphatases: Structure and function, substrate specificity, and inhibitor development. Annu Rev Pharmacol Toxicol. 42:209–234. 2002. View Article : Google Scholar : PubMed/NCBI

51 

He RJ, Yu ZH, Zhang RY and Zhang ZY: Protein tyrosine phosphatases as potential therapeutic targets. Acta Pharmacol Sin. 35:1227–1246. 2014. View Article : Google Scholar : PubMed/NCBI

52 

Wang ZC, Gao Q, Shi JY, Guo WJ, Yang LX, Liu XY, Liu LZ, Ma LJ, Duan M, Zhao YJ, et al: Protein tyrosine phosphatase receptor S acts as a metastatic suppressor in hepatocellular carcinoma by control of epithermal growth factor receptor-induced epithelial-mesenchymal transition. Hepatology. 62:1201–1214. 2015. View Article : Google Scholar : PubMed/NCBI

53 

Meeusen B and Janssens V: Tumor suppressive protein phosphatases in human cancer: Emerging targets for therapeutic intervention and tumor stratification. Int J Biochem Cell Biol. 96:98–134. 2018. View Article : Google Scholar : PubMed/NCBI

54 

Tai WT, Cheng AL, Shiau CW, Liu CY, Ko CH, Lin MW, Chen PJ and Chen KF: Dovitinib induces apoptosis and overcomes sorafenib resistance in hepatocellular carcinoma through SHP-1-mediated inhibition of STAT3. Mol Cancer Ther. 11:452–463. 2012. View Article : Google Scholar : PubMed/NCBI

55 

Wang CI, Chu PM, Chen YL, Lin YH and Chen CY: Chemotherapeutic drug-regulated cytokines might influence therapeutic efficacy in HCC. Int J Mol Sci. 22:136272021. View Article : Google Scholar : PubMed/NCBI

56 

Sakurai T, Yada N, Hagiwara S, Arizumi T, Minaga K, Kamata K, Takenaka M, Minami Y, Watanabe T, Nishida N and Kudo M: Gankyrin induces STAT3 activation in tumor microenvironment and sorafenib resistance in hepatocellular carcinoma. Cancer Sci. 108:1996–2003. 2017. View Article : Google Scholar : PubMed/NCBI

57 

Meng Q, Tian J, Qin F, Huang X, Zhu D, Xiang B and Dong D: Protein tyrosine phosphatase receptor type delta (PTPRD) suppresses the expression of PD-L1 in human hepatocellular carcinoma by down-regulating STAT3. Transl Cancer Res. 9:5574–5584. 2020. View Article : Google Scholar : PubMed/NCBI

58 

Kim B and Park B: Saffron carotenoids inhibit STAT3 activation and promote apoptotic progression in IL-6-stimulated liver cancer cells. Oncol Rep. 39:1883–1891. 2018.PubMed/NCBI

59 

Igbe I, Shen XF, Jiao W, Qiang Z, Deng T, Li S, Liu WL, Liu HW, Zhang GL and Wang F: Dietary quercetin potentiates the antiproliferative effect of interferon-α in hepatocellular carcinoma cells through activation of JAK/STAT pathway signaling by inhibition of SHP2 phosphatase. Oncotarget. 8:113734–113748. 2017. View Article : Google Scholar : PubMed/NCBI

60 

Hammer M, Mages J, Dietrich H, Servatius A, Howells N, Cato AC and Lang R: Dual specificity phosphatase 1 (DUSP1) regulates a subset of LPS-induced genes and protects mice from lethal endotoxin shock. J Exp Med. 203:15–20. 2006. View Article : Google Scholar : PubMed/NCBI

61 

Wei X, Tang C, Lu X, Liu R, Zhou M, He D, Zheng D, Sun C and Wu Z: MiR-101 targets DUSP1 to regulate the TGF-β secretion in sorafenib inhibits macrophage-induced growth of hepatocarcinoma. Oncotarget. 6:18389–18405. 2015. View Article : Google Scholar : PubMed/NCBI

62 

Chen S, Tang Y, Yang C, Li K, Huang X and Cao J: Silencing CDC25A inhibits the proliferation of liver cancer cells by downregulating IL6 in vitro and in vivo. Int J Mol Med. 45:743–752. 2020.PubMed/NCBI

63 

Hou J, Xu J, Jiang R, Wang Y, Chen C, Deng L, Huang X, Wang X and Sun B: Estrogen-sensitive PTPRO expression represses hepatocellular carcinoma progression by control of STAT3. Hepatology. 57:678–688. 2013. View Article : Google Scholar : PubMed/NCBI

64 

Tsukamoto H, Fujieda K, Miyashita A, Fukushima S, Ikeda T, Kubo Y, Senju S, Ihn H, Nishimura Y and Oshiumi H: Combined Blockade of IL6 and PD-1/PD-L1 signaling abrogates mutual regulation of their immunosuppressive effects in the tumor microenvironment. Cancer Res. 78:5011–5022. 2018. View Article : Google Scholar : PubMed/NCBI

65 

Zhang W, Liu Y, Yan Z, Yang H, Sun W, Yao Y, Chen Y and Jiang R: IL-6 promotes PD-L1 expression in monocytes and macrophages by decreasing protein tyrosine phosphatase receptor type O expression in human hepatocellular carcinoma. J Immunother Cancer. 8:e0002852020. View Article : Google Scholar : PubMed/NCBI

66 

He G and Karin M: NF-κB and STAT3-key players in liver inflammation and cancer. Cell Res. 21:159–168. 2011. View Article : Google Scholar : PubMed/NCBI

67 

Wang K and Karin M: Tumor-Elicited inflammation and colorectal cancer. Adv Cancer Res. 128:173–196. 2015. View Article : Google Scholar : PubMed/NCBI

68 

Naugler WE, Sakurai T, Kim S, Maeda S, Kim K, Elsharkawy AM and Karin M: Gender disparity in liver cancer due to sex differences in MyD88-dependent IL-6 production. Science. 317:121–124. 2007. View Article : Google Scholar : PubMed/NCBI

69 

Park EJ, Lee JH, Yu GY, He G, Ali SR, Holzer RG, Osterreicher CH, Takahashi H and Karin M: Dietary and genetic obesity promote liver inflammation and tumorigenesis by enhancing IL-6 and TNF expression. Cell. 140:197–208. 2010. View Article : Google Scholar : PubMed/NCBI

70 

Grossmann KS, Rosario M, Birchmeier C and Birchmeier W: The tyrosine phosphatase Shp2 in development and cancer. Adv Cancer Res. 106:53–89. 2010. View Article : Google Scholar : PubMed/NCBI

71 

Mohi MG and Neel BG: The role of Shp2 (PTPN11) in cancer. Curr Opin Genet Dev. 17:23–30. 2007. View Article : Google Scholar : PubMed/NCBI

72 

Bard-Chapeau EA, Li S, Ding J, Zhang SS, Zhu HH, Princen F, Fang DD, Han T, Bailly-Maitre B, Poli V, et al: Ptpn11/Shp2 acts as a tumor suppressor in hepatocellular carcinogenesis. Cancer Cell. 19:629–639. 2011. View Article : Google Scholar : PubMed/NCBI

73 

Jiang C, Hu F, Tai Y, Du J, Mao B, Yuan Z, Wang Y and Wei L: The tumor suppressor role of Src homology phosphotyrosine phosphatase 2 in hepatocellular carcinoma. J Cancer Res Clin Oncol. 138:637–646. 2012. View Article : Google Scholar : PubMed/NCBI

74 

Chan TA and Heguy A: The protein tyrosine phosphatase receptor D, a broadly inactivated tumor suppressor regulating STAT function. Cell Cycle. 8:3063–3064. 2009. View Article : Google Scholar : PubMed/NCBI

75 

Ostman A, Hellberg C and Bohmer FD: Protein-tyrosine phosphatases and cancer. Nat Rev Cancer. 6:307–320. 2006. View Article : Google Scholar : PubMed/NCBI

76 

McFarland BC and Benveniste EN: Reactive astrocytes foster brain metastases via STAT3 signaling. Ann Transl Med. 7 (Suppl 3):S832019. View Article : Google Scholar : PubMed/NCBI

77 

Chen J, Jiang CC, Jin L and Zhang XD: Regulation of PD-L1: A novel role of pro-survival signalling in cancer. Ann Oncol. 27:409–416. 2016. View Article : Google Scholar : PubMed/NCBI

78 

Yang L, Sun YY, Liu YR, Yin NN, Bu FT, Yu HX, Du XS, Li J and Huang C: PTP1B promotes macrophage activation by regulating the NF-κB pathway in alcoholic liver injury. Toxicol Lett. 319:11–21. 2020. View Article : Google Scholar : PubMed/NCBI

79 

Chong ZZ and Maiese K: The Src homology 2 domain tyrosine phosphatases SHP-1 and SHP-2: Diversified control of cell growth, inflammation, and injury. Histol Histopathol. 22:1251–1267. 2007.PubMed/NCBI

80 

Han T, Xiang DM, Sun W, Liu N, Sun HL, Wen W, Shen WF, Wang RY, Chen C, Wang X, et al: PTPN11/Shp2 overexpression enhances liver cancer progression and predicts poor prognosis of patients. J Hepatol. 63:651–660. 2015. View Article : Google Scholar : PubMed/NCBI

81 

An H, Hou J, Zhou J, Zhao W, Xu H, Zheng Y, Yu Y, Liu S and Cao X: Phosphatase SHP-1 promotes TLR- and RIG-I-activated production of type I interferon by inhibiting the kinase IRAK1. Nat Immunol. 9:542–550. 2008. View Article : Google Scholar : PubMed/NCBI

82 

Tai WT, Shiau CW, Chen PJ, Chu PY, Huang HP, Liu CY, Huang JW and Chen KF: Discovery of novel Src homology region 2 domain-containing phosphatase 1 agonists from sorafenib for the treatment of hepatocellular carcinoma. Hepatology. 59:190–201. 2014. View Article : Google Scholar : PubMed/NCBI

83 

Chen KF, Tai WT, Hsu CY, Huang JW, Liu CY, Chen PJ, Kim I and Shiau CW: Blockade of STAT3 activation by sorafenib derivatives through enhancing SHP-1 phosphatase activity. Eur J Med Chem. 55:220–227. 2012. View Article : Google Scholar : PubMed/NCBI

84 

Fan LC, Shiau CW, Tai WT, Hung MH, Chu PY, Hsieh FS, Lin H, Yu HC and Chen KF: SHP-1 is a negative regulator of epithelial-mesenchymal transition in hepatocellular carcinoma. Oncogene. 34:5252–5263. 2015. View Article : Google Scholar : PubMed/NCBI

85 

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. 78:4680–4691. 2018. View Article : Google Scholar : PubMed/NCBI

86 

Su JC, Tseng PH, Hsu CY, Tai WT, Huang JW, Ko CH, Lin MW, Liu CY, Chen KF and Shiau CW: RFX1-dependent activation of SHP-1 induces autophagy by a novel obatoclax derivative in hepatocellular carcinoma cells. Oncotarget. 5:4909–4919. 2014. View Article : Google Scholar : PubMed/NCBI

87 

Qian H, Deng X, Huang ZW, Wei J, Ding CH, Feng RX, Zeng X, Chen YX, Ding J, Qiu L, et al: An HNF1α-regulated feedback circuit modulates hepatic fibrogenesis via the crosstalk between hepatocytes and hepatic stellate cells. Cell Res. 25:930–945. 2015. View Article : Google Scholar : PubMed/NCBI

88 

Zhang J, Li Z, Liu L, Wang Q, Li S, Chen D, Hu Z, Yu T, Ding J, Li J, et al: Long noncoding RNA TSLNC8 is a tumor suppressor that inactivates the interleukin-6/STAT3 signaling pathway. Hepatology. 67:171–187. 2018. View Article : Google Scholar : PubMed/NCBI

89 

Duran A, Hernandez ED, Reina-Campos M, Castilla EA, Subramaniam S, Raghunandan S, Roberts LR, Kisseleva T, Karin M, Diaz-Meco MT and Moscat J: p62/SQSTM1 by binding to vitamin D receptor inhibits hepatic stellate cell activity, fibrosis, and liver cancer. Cancer Cell. 30:595–609. 2016. View Article : Google Scholar : PubMed/NCBI

90 

Yuan JH, Yang F, Wang F, Ma JZ, Guo YJ, Tao QF, Liu F, Pan W, Wang TT, Zhou CC, et al: A long noncoding RNA activated by TGF-β promotes the invasion-metastasis cascade in hepatocellular carcinoma. Cancer Cell. 25:666–681. 2014. View Article : Google Scholar : PubMed/NCBI

91 

Li N, Zhou ZS, Shen Y, Xu J, Miao HH, Xiong Y, Xu F, Li BL, Luo J and Song BL: Inhibition of the sterol regulatory element-binding protein pathway suppresses hepatocellular carcinoma by repressing inflammation in mice. Hepatology. 65:1936–1947. 2017. View Article : Google Scholar : PubMed/NCBI

92 

Zhang L, Yang Z, Ma A, Qu Y, Xia S, Xu D, Ge C, Qiu B, Xia Q, Li J and Liu Y: Growth arrest and DNA damage 45G down-regulation contributes to Janus kinase/signal transducer and activator of transcription 3 activation and cellular senescence evasion in hepatocellular carcinoma. Hepatology. 59:178–189. 2014. View Article : Google Scholar : PubMed/NCBI

93 

Gough DJ, Corlett A, Schlessinger K, Wegrzyn J, Larner AC and Levy DE: Mitochondrial STAT3 supports Ras-dependent oncogenic transformation. Science. 324:1713–1716. 2009. View Article : Google Scholar : PubMed/NCBI

94 

Wang YQ, Zhang F, Tian R, Ji W, Zhou Y, Sun XM, Liu Y, Wang ZY and Niu RF: Tyrosine 23 phosphorylation of annexin A2 promotes proliferation, invasion, and Stat3 phosphorylation in the nucleus of human breast cancer SK-BR-3 Cells. Cancer Biol Med. 9:248–253. 2012.PubMed/NCBI

95 

Yamada S, Shiono S, Joo A and Yoshimura A: Control mechanism of JAK/STAT signal transduction pathway. FEBS Lett. 534:190–196. 2003. View Article : Google Scholar : PubMed/NCBI

96 

Zhang Z, Shen K, Lu W and Cole PA: The role of C-terminal tyrosine phosphorylation in the regulation of SHP-1 explored via expressed protein ligation. J Biol Chem. 278:4668–4674. 2003. View Article : Google Scholar : PubMed/NCBI

97 

Xiang D, Cheng Z, Liu H, Wang X, Han T, Sun W, Li X, Yang W, Chen C, Xia M, et al: Shp2 promotes liver cancer stem cell expansion by augmenting beta-catenin signaling and predicts chemotherapeutic response of patients. Hepatology. 65:1566–1580. 2017. View Article : Google Scholar : PubMed/NCBI

98 

Chen YN, LaMarche MJ, Chan HM, Fekkes P, Garcia-Fortanet J, Acker MG, Antonakos B, Chen CH, Chen Z, Cooke VG, et al: Allosteric inhibition of SHP2 phosphatase inhibits cancers driven by receptor tyrosine kinases. Nature. 535:148–152. 2016. View Article : Google Scholar : PubMed/NCBI

99 

Leung CON, Tong M, Chung KPS, Zhou L, Che N, Tang KH, Ding J, Lau EYT, Ng IOL, Ma S and Lee TKW: Overriding adaptive resistance to sorafenib through combination therapy with Src homology 2 domain-containing phosphatase 2 blockade in hepatocellular carcinoma. Hepatology. 72:155–168. 2020. View Article : Google Scholar : PubMed/NCBI

100 

Kang HJ, Chung DH, Sung CO, Yoo SH, Yu E, Kim N, Lee SH, Song JY, Kim CJ and Choi J: SHP2 is induced by the HBx-NF-κB pathway and contributes to fibrosis during human early hepatocellular carcinoma development. Oncotarget. 8:27263–27276. 2017. View Article : Google Scholar : PubMed/NCBI

101 

Lo J, Lau EY, Ching RH, Cheng BY, Ma MK, Ng IO and Lee TK: Nuclear factor kappa B-mediated CD47 up-regulation promotes sorafenib resistance and its blockade synergizes the effect of sorafenib in hepatocellular carcinoma in mice. Hepatology. 62:534–545. 2015. View Article : Google Scholar : PubMed/NCBI

102 

Mohi MG, Williams IR, Dearolf CR, Chan G, Kutok JL, Cohen S, Morgan K, Boulton C, Shigematsu H, Keilhack H, et al: Prognostic, therapeutic, and mechanistic implications of a mouse model of leukemia evoked by Shp2 (PTPN11) mutations. Cancer Cell. 7:179–191. 2005. View Article : Google Scholar : PubMed/NCBI

103 

Zhang SQ, Yang W, Kontaridis MI, Bivona TG, Wen G, Araki T, Luo J, Thompson JA, Schraven BL, Philips MR and Neel BG: Shp2 regulates SRC family kinase activity and Ras/Erk activation by controlling Csk recruitment. Mol Cell. 13:341–355. 2004. View Article : Google Scholar : PubMed/NCBI

104 

Yang X, Tang C, Luo H, Wang H and Zhou X: Shp2 confers cisplatin resistance in small cell lung cancer via an AKT-mediated increase in CA916798. Oncotarget. 8:23664–23674. 2017. View Article : Google Scholar : PubMed/NCBI

105 

Ran H, Tsutsumi R, Araki T and Neel BG: Sticking it to cancer with molecular glue for SHP2. Cancer Cell. 30:194–196. 2016. View Article : Google Scholar : PubMed/NCBI

106 

Wu C, Sun M, Liu L and Zhou GW: The function of the protein tyrosine phosphatase SHP-1 in cancer. Gene. 306:1–12. 2003. View Article : Google Scholar : PubMed/NCBI

107 

Barr AJ, Ugochukwu E, Lee WH, King ON, Filippakopoulos P, Alfano I, Savitsky P, Burgess-Brown NA, Müller S and Knapp S: Large-scale structural analysis of the classical human protein tyrosine phosphatome. Cell. 136:352–363. 2009. View Article : Google Scholar : PubMed/NCBI

108 

Tai WT, Hung MH, Chu PY, Chen YL, Chen LJ, Tsai MH, Chen MH, Shiau CW, Boo YP and Chen KF: SH2 domain-containing phosphatase 1 regulates pyruvate kinase M2 in hepatocellular carcinoma. Oncotarget. 7:22193–22205. 2016. View Article : Google Scholar : PubMed/NCBI

109 

Pfirsch-Maisonnas S, Aloulou M, Xu T, Claver J, Kanamaru Y, Tiwari M, Launay P, Monteiro RC and Blank U: Inhibitory ITAM signaling traps activating receptors with the phosphatase SHP-1 to form polarized ‘inhibisome’ clusters. Sci Signal. 4:ra242011. View Article : Google Scholar : PubMed/NCBI

110 

Alsadeq A, Hobeika E, Medgyesi D, Klasener K and Reth M: The role of the Syk/Shp-1 kinase-phosphatase equilibrium in B cell development and signaling. J Immunol. 193:268–276. 2014. View Article : Google Scholar : PubMed/NCBI

111 

Viant C, Fenis A, Chicanne G, Payrastre B, Ugolini S and Vivier E: SHP-1-mediated inhibitory signals promote responsiveness and anti-tumour functions of natural killer cells. Nat Commun. 5:51082014. View Article : Google Scholar : PubMed/NCBI

112 

Motiwala T, Kutay H, Ghoshal K, Bai S, Seimiya H, Tsuruo T, Suster S, Morrison C and Jacob ST: Protein tyrosine phosphatase receptor-type O (PTPRO) exhibits characteristics of a candidate tumor suppressor in human lung cancer. Proc Natl Acad Sci USA. 101:13844–13849. 2004. View Article : Google Scholar : PubMed/NCBI

113 

Motiwala T, Ghoshal K, Das A, Majumder S, Weichenhan D, Wu YZ, Holman K, James SJ, Jacob ST and Plass C: Suppression of the protein tyrosine phosphatase receptor type O gene (PTPRO) by methylation in hepatocellular carcinomas. Oncogene. 22:6319–6331. 2003. View Article : Google Scholar : PubMed/NCBI

114 

Klionsky DJ, Abeliovich H, Agostinis P, Agrawal DK, Aliev G, Askew DS, Baba M, Baehrecke EH, Bahr BA, Ballabio A, et al: Guidelines for the use and interpretation of assays for monitoring autophagy in higher eukaryotes. Autophagy. 4:151–175. 2008. View Article : Google Scholar : PubMed/NCBI

115 

Zhang W, Hou J, Wang X, Jiang R, Yin Y, Ji J, Deng L, Huang X, Wang K and Sun B: PTPRO-mediated autophagy prevents hepatosteatosis and tumorigenesis. Oncotarget. 6:9420–9433. 2015. View Article : Google Scholar : PubMed/NCBI

116 

Mareninova OA, Hermann K, French SW, O'Konski MS, Pandol SJ, Webster P, Erickson AH, Katunuma N, Gorelick FS, Gukovsky I and Gukovskaya AS: Impaired autophagic flux mediates acinar cell vacuole formation and trypsinogen activation in rodent models of acute pancreatitis. J Clin Invest. 119:3340–3355. 2009.PubMed/NCBI

117 

Klionsky DJ, Abdalla FC, Abeliovich H, Abraham RT, Acevedo-Arozena A, Adeli K, Agholme L, Agnello M, Agostinis P, Aguirre-Ghiso JA, et al: Guidelines for the use and interpretation of assays for monitoring autophagy. Autophagy. 8:445–544. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2023
Volume 49 Issue 3

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen Y, Hsieh C, Chu P, Chen J, Huang Y and Chen C: Roles of protein tyrosine phosphatases in hepatocellular carcinoma progression (Review). Oncol Rep 49: 48, 2023
APA
Chen, Y., Hsieh, C., Chu, P., Chen, J., Huang, Y., & Chen, C. (2023). Roles of protein tyrosine phosphatases in hepatocellular carcinoma progression (Review). Oncology Reports, 49, 48. https://doi.org/10.3892/or.2023.8485
MLA
Chen, Y., Hsieh, C., Chu, P., Chen, J., Huang, Y., Chen, C."Roles of protein tyrosine phosphatases in hepatocellular carcinoma progression (Review)". Oncology Reports 49.3 (2023): 48.
Chicago
Chen, Y., Hsieh, C., Chu, P., Chen, J., Huang, Y., Chen, C."Roles of protein tyrosine phosphatases in hepatocellular carcinoma progression (Review)". Oncology Reports 49, no. 3 (2023): 48. https://doi.org/10.3892/or.2023.8485