Deactivation of glycogen synthase kinase-3β by heat shock‑inducible tumor small protein attenuates hyperthermia‑induced pro‑migratory activity in colorectal cancer cells

  • Authors:
    • Keita Koizumi
    • Takahiro Domoto
    • Toshinari Minamoto
    • Kazuhito Satomura
    • Hideo Nakajima
  • View Affiliations

  • Published online on: June 23, 2023     https://doi.org/10.3892/ijo.2023.5540
  • Article Number: 92
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hyperthermia is a promising approach for improving cancer treatment in combination with chemotherapy, radiotherapy and/or immunotherapy; however, its molecular mechanisms remain unclear. Although heat shock proteins (HSPs) are involved in hyperthermia via antigen presentation and immune activation, major HSPs including HSP90 are associated with cancer progression via tumor cell migration and metastasis. The present study showed that heat shock‑inducible tumor small protein (HITS) could counteract the pro‑migratory effects of HSPs in colorectal cancer (CRC) cells, which represents a novel function. Western blotting analysis revealed that overexpression of HITS increased the protein level of glycogen synthase kinase‑3β (GSK3β) phosphorylated (p) at the serine 9 (pGSK3βS9; inactive form) in HCT 116, RKO and SW480 CRC cells. GSK3βS9 phosphorylation was reported to suppress migration in some cancer types; therefore, by using the wound healing assay, the present study revealed that HITS overexpression decreased the migration activity of CRC cells. Induction of HITS transcription was observed at 12 and 18 h after heat shock (HS) by using semi‑quantitative reverse transcription‑PCR analysis, followed by increased levels of pGSK3βS9 protein at 24 and 30 h in CRC cells in western blotting. Thus, HS induced not only HSPs to promote cell migration, but also HITS to counteract the migratory activity of these HSPs in CRC cells. HITS knockdown in CRC cells subject to HS showed increased cell migration in wound healing assay, which was decreased by the GSK3β inhibitor AR‑A014418, confirming the anti‑migratory effect of HITS via the deactivation of GSK3β. The present findings indicated that the deactivation of GSK3β sufficiently offset the pro‑migratory effect of hyperthermia via major HSPs in CRC.

Introduction

Colorectal cancer (CRC) is the third most commonly diagnosed cancer type and the second leading cause of cancer-related deaths worldwide (1,2). Patients with CRC develop metastatic disease in >50% of cases, especially in the liver, which results in the death of >2/3 of the patients (35). Advances in chemotherapy, radiotherapy and immunotherapy have improved the survival of patients with metastatic CRC (6,7); however, resistance to anticancer drugs, radiation and immune therapies remains a major therapeutic challenge (79).

Increasing evidence has demonstrated that the combination of hyperthermia with the aforementioned therapies can overcome tumor resistance to antitumor treatments and improve their efficacy (1012). In addition, heat shock (HS) proteins (HSPs) are involved in enhancing the cytotoxic activity of natural killer (NK) cells (11,13), inducing maturation and antigen presentation of dendritic cells and activating T-cells (11,14). By contrast, previous studies have indicated that HSPs including HSP90 promote cancer cell proliferation, invasion and metastasis, as well as tumor angiogenesis, which can negatively affect the hyperthermia efficacy (15,16). These major HSPs are highly expressed in some malignant tumor types and are inversely correlated with prognosis, leading to these HSPs being used as therapeutic targets (15,16). Nonetheless, the overall involvement of HSPs in hyperthermia remains unclear.

Heat shock-inducible tumor small protein (HITS) is an 18 kDa protein that was originally identified as a molecule upregulated upon HS treatment in Jurkat cells (17). The induction of HITS protein as well as HSP90 protein has been confirmed in THP-1 cells in vitro and transplanted rat walker 256 sarcoma cells in vivo (17). HITS is highly homologous to downregulated in renal cell carcinoma 1 (DRR1), a putative tumor suppressor that plays important roles in actin and microtubule cytoskeleton organization, and is downregulated in renal cell carcinoma (1719). Our previous studies indicate that HITS expression can be observed in various cancer cells and is downregulated during tumor progression in colon cancer as well as cervix, thyroid and breast cancers compared with the corresponding healthy tissues (17,20). Unlike the major HSPs, overexpression of HITS shows tumor suppressive phenotype in the mouse cervical cancer xenograft model (20).

The present study showed that HITS overexpression increased the levels of glycogen synthase kinase-3β (GSK3β) phosphorylated (p) at serine (S) 9 (pGSK3βS9), resulting in its deactivation in CRC cells (2123). To the best of our knowledge, while GSK3β deactivation has been reported to prevent β-catenin degeneration (23), no previous studies have shown a direct association of cellular GSK3β deactivation with cancer development or progression (22). GSK3β does not participate in the canonical β-catenin destruction complex in the majority of CRCs due to the mutations in either adenomatous polyposis coli (APC; <90% of cases), catenin β-1 (CTNNB1; ~5% of cases) or axis inhibition protein 1 (AXIN1) (22). By contrast, previous studies have demonstrated that pharmacological inhibition of GSK3β activity suppresses cancer progression by attenuating tumor cell migration and invasion in several cancer types, including colorectal, breast and pancreatic cancers, in addition to glioblastoma (2129). The present study showed that HITS upregulation induced by HS exerted an anti-migratory effect via GSK3β deactivation (S9 phosphorylation) in human CRCs, thereby counteracting the pro-migratory effects of HSPs. This novel anti-migratory mechanism may play an important role in reducing the risk of cancer metastasis during hyperthermia.

Materials and methods

Cell culture

HCT 116, RKO and SW480 cells were purchased from American Type Culture Collection and maintained in Dulbecco's Modified Eagle's Medium-high glucose (cat. no. 043-30085; Fujifilm Wako Pure Chemical Corporation) supplemented with 10% fetal bovine serum (FBS) (cat. no. F7524; Sigma-Aldrich; Merck KGaA) in a humidified incubator with 5% CO2 at 37°C. Cells were tested for mycoplasma contamination.

Western blotting

Cells were lysed with 2X sample buffer containing 4% SDS (cat. no. 08933-34; Nacalai Tesque, Inc.), 20% glycerol (cat. no. 17018-25; Nacalai Tesque, Inc.), 0.001% bromophenol blue (cat. no. 05808-61; Nacalai Tesque, Inc.), 0.125 M Tris HCl (cat. no. T1503, Sigma-Aldrich; Merck KGaA) and 10% 2-mercaptoethanol (cat. no. 21418-42; Nacalai Tesque, Inc.) and boiled for 5 min. The protein concentration was measured using BCA Protein Assay Kit (cat. no. 23225; Thermo Fisher Scientific, Inc.). Lysates (20 µg/lane) were separated by SDS-PAGE on an 8.5% gel and transferred onto PVDF membrane (cat. no. IPVH00010; MilliporeSigma). To calculate relative intensity, cell lysates to compare were loaded in the same gel. Membranes were blocked with phosphate-buffered saline [PBT; 0.01 M Na2HPO4; cat. no. 31801-05 and KH2PO4; cat. no. 28721-55; pH 7.4 with 0.15 M NaCl; cat. no. 31320-05; and 0.1% Tween-20 (cat. no. 35624-02; Nacalai Tesque, Inc.)] containing 5% non-fat dry milk or 5% bovine serum albumin (cat. no. 019-21272; Fujifilm Wako Pure Chemical Corporation) for 1 h at room temperature. After washing with PBT, the membranes were incubated with the primary antibodies against GSK3β (1:20,000 or 40,000; cat. no. 9315; Cell Signaling Technology, Inc.), pGSK3βS9 (1:1,000 or 2,000; cat. no. 9336; Cell Signaling Technology, Inc.) and β-tubulin (1:120,000 or 40,000; cat. no. 017-25031; Fujifilm Wako Pure Chemical Corporation) overnight at 4°C. This was followed by incubation with anti-rabbit (1:4,000 dilution; cat. no. 5220-0336; SeraCare Life Sciences, Inc.) and anti-mouse (1:4,000 dilution; cat. no. 5220-0341; SeraCare Life Sciences, Inc.) horseradish peroxidase-conjugated secondary antibodies for 1 h at room temperature. The protein bands were developed using the ECL Prime Western Blotting Detection Reagent (cat. no. RPN2232; Cytiva) and the chemiluminescence was detected using a ChemiDoc Touch MP Imaging System with Image Touch 2.4 software (Bio-Rad Laboratories, Inc.) or C-Digit Blot scanner (LI-COR Biosciences).

RNA extraction and semi-quantitative reverse transcription (RT)-PCR

Total RNA was extracted from cells using TRIzol™ (cat. no. 15596026; Thermo Fisher Scientific, Inc.) and reverse-transcribed to complementary DNA (cDNA) using ReverTra Ace™ (cat. no. FSQ-201; Toyobo Life Science) with Random Primer (cat. no. FSK-301; Toyobo Life Science) according to the manufacturer's instructions. Semi-quantitative RT-PCR was performed using a LifeECO thermal cycler (Yakukensha Co., Ltd.). The reaction mixture comprised 0.25 µl Blend Taq plus (2.5 U/µl; cat. no. BTQ-201; Toyobo Life Science), 2.5 µl 10X buffer, 2.5 µl dNTP (2 mM; cat. no. NTP-501; Toyobo), 0.25 µl primers (10 µM; cat. no. FSK-301; Toyobo) and 1–3 µl of synthesized cDNA. The following primer pairs were used for semi-quantitative RT-PCR: HITS forward, 5′-CCACCTGAGGATATTGACCATAA-3′ and reverse, 5′-TTCTGTGCTTCTTCTTCCTTCTG-3′; matrix metalloproteinase-3 (MMP-3), forward, 5′-CTCAGGAAGCTTGAACCTGAAT-3′ and reverse, 5′-CAGCTCGTACCTCATTTCCTCT-3′; MMP-13, forward, 5′-TTACCAGTCTCCGAGGAGAAAC-3′ and reverse, 5′-TTTTGGAAGACCCAGTTCAGAT-3′; and TATA-box binding protein (TBP) forward, 5′-AGAAAGTGAACATCATGGATCAGA-3′ and reverse, 5′-GTTTACAACCAAGATTCACTGTGG-3′. TBP was amplified as an internal control to normalize the expression of the target gene. The PCR reaction was performed under the following conditions: Initial denaturation at 94°C for 1 min; followed by cycling at 94°C for 20 sec, 55°C (HITS, MMP-13 and TBP) or 53°C (MMP-3) for 20 sec and 72°C for 20 sec; and final extension at 72°C for 1 min. The numbers of cycles were 29 and 30 for HITS, TBP; 36 for MMP-3, 36 for MMP-13 and 33 for TBP. The DNA products were run on a 1.2% agarose gel containing ethidium bromide (cat. no. 312-01193; Nippon Gene Co., Ltd.) and imaged using a ChemiDoc MP Imaging System (Bio-Rad Laboratories, Inc.). The intensity of each band was quantified using ImageJ software, ver. 1.50 (National Institutes of Health).

Plasmid construction and overexpression

The amino acid sequence of the mouse HITS open reading frame (ORF) is 98.47% homologous to the human HITS; therefore, mouse HITS ORF was cloned for transfection (17). The HITS ORF fragment with EcoRV and BamHI ends was amplified from HeLa-Tet-HITS (16) using PCR (initial denaturation at 94°C for 1 min; 25 cycles of 94°C for 20 sec, 55°C for 20 sec and 72°C for 20 sec; and a final extension at 72°C for 1 min) and the following primers: forward, 5′-GATATCATGGCTGAGCCAGACTACATAGAAG-3′, and reverse, 5′-GGATCCCTAGGACTCCTGGGCCTGAGCCACC-3′. The amplified DNA was cloned into PCR2.1 vector using the TOPO™ TA™ Cloning Kit (cat. no. K450002; Thermo Fisher Scientific, Inc.) and subcloned into the EcoRV/BamHI sites of pCAG-IRES-EGFP, which was kindly supplied by Dr T. Kawauchi (30). The generated plasmid (0.5 µg/well), namely pCAG-HITS-IRES-EGFP, and pEGFP (30) were mixed with transfection reagents and kept for 10 min then transfected into HCT 116, RKO and SW480 cells, using Lipofectamine 3000 (cat. no. L3000001; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. The cells were incubated for 36 h and obtained cell lysates for western blotting, wound healing assay or MTT assay as described later. Transfection efficiency was tested using semi-quantitative RT-PCR as described in the previous section (Fig. S1).

RNA interference (RNAi)

The sequences of small interfering RNAs (siRNAs) targeting HITS were as follows: HITS-siRNA#1, 5′-GGAUAUUGACCAUAAGGACUCAUAU-3′; and HITS-siRNA#2, 5′-GCCUCAGAAACUGAUCAAUCCUGUA-3′ (Thermo Fisher Scientific, Inc.). Lipofectamine™ RNAiMAX (cat. no. 13778030; Thermo Fisher Scientific, Inc.) was used to transfect HITS siRNAs and control siRNA (cat. no. 12935300; Thermo Fisher Scientific, Inc.). A total of 10 pmol/well siRNAs were mixed with transfection reagents and kept for 5 min according to the manufacturer's instructions. The cells were incubated for 36 h and obtained cell lysates for western blotting or wound healing assay as described later.

Wound healing assay

HCT 116 cells were seeded at a density of 5×105 cells/well in a 24-well plate and transfected with HITS the following day. At 10 h after transfection, the cells were trypsinized, re-seeded at a density of 8×105 cells/well, and incubated for 18 h (the confluency of the cells reached 80–90%). The cells were then scratched with a 200-µl pipette tip and the medium was replaced to medium containing 1% FBS to prevent the loose cells from settling back down. Images of the cells were captured at 0 and 24 h under a phase-contrast and fluorescence microscope (BZ-X700; Keyence Corp.). Multiple images were captured with the microscope to cover an entire scratched area and merged into one image to compare the same wound gap positions between 0 and 24 h. The wound healing rate was calculated as follows: Wound healing rate (%)=(wound area at 0 h-wound area at 24 h)/(initial wound at 0 h) ×100. For the assay performed on HS-induced cells (HS at 42°C for 1 h), 8.5×105 cells/well were plated. For RNAi followed by HS and inhibitors treatment, the CytoSelect 24-well Wound Healing Assay Kit (Cell Biolabs, Inc.) was used. Plastic inserts provided with the kit were placed onto the wells to create wound gaps where cells were plated at a density of 8.5×105 cells/well. Some groups of cells were treated with the HSP90 inhibitor 17-AAG (cat. no. CS-0161; Funakoshi Co., Ltd.) or the GSK3β inhibitor AR-A014418 (cat. no. A3230; Sigma-Aldrich; Merck KGaA), both of which were dissolved in dimethyl sulfoxide (DMSO) to prepare 10 mM stock solutions. DMSO was added to control and 17-AAG samples to adjust the DMSO concentration to 0.2%. The concentration of 17-AAG was optimized by testing different concentrations, which showed suppression of cell migration but no effect against cell survival (data not shown). The concentration of AR-A014418 was determined based on our previous studies (29).

MTT assay

HCT 116 cells were seeded at a density of 1×104 cells/well in a 96-well plate. The expression vectors pCAG-HITS-IRES-EGFP and pEGFP were transfected into the cells the following day, as previously described. The MTT assay (cat. no. 11465007001; Sigma-Aldrich; Merck KGaA) was performed 2 days after transfection, according to the manufacturer's instructions. Methanol was used to dissolve formazan. Colorimetric detection was done at a wavelength of 550 nm and reference wavelength was 690 nm.

Statistical analysis

Data are expressed as the mean ± standard error. Statistical analysis was performed using the unpaired Student's t-test to compare two groups of data and one-way analysis of variance (ANOVA) followed by Dunnett's test or two-way ANOVA followed by Tukey's test for multiple comparisons. P<0.05 was considered to indicate a statistically significant difference.

Results

HITS promotes the phosphorylation of GSK3β S9 in CRC cells

The effects of HITS on GSK3β, which plays important roles in tumorigenesis (21,22), were investigated to study the role of HITS in cancer progression. Overexpression of HITS in HCT 116, RKO and SW480 cells were followed by a significant increase in pGSK3βS9 level, while no significant changes were observed in the expression of GSK3β (Fig. 1A-C) nor in the levels of GSK3β phosphorylation at tyrosine (Y)216 (pGSK3βY216; active form; data not shown).

HCT 116 is one of the human CRC cell lines that were established to extrapolate clinical CRC (31,32). Our previous studies showed the distinct and common tumor-promoting roles of active GSK3β (lower and higher phosphorylation of its S9 and Y216 residues, respectively) in multiple human CRC cell lines including HCT 116 cells (29,3335). In addition, no biological association between the pathological (tumor-promoting) property of deregulated GSK3β and the pathways mediated by activated β-catenin and phosphoinositide 3-kinase (PI3K)/Akt in human CRC cell lines including HCT 116 cells as well as in clinical CRC tumors (29). Therefore, the present study used HCT 116 cells for the subsequent analyses.

To confirm the effects of HITS on the expression and phosphorylation of GSK3β, HITS RNAi was performed on CRC cells. Semi-quantitative RT-PCR indicated that transfection with either HITS siRNA#1 or HITS siRNA#2 significantly reduced HITS mRNA expression by >90% compared with the that in cells transfected with control siRNA (Fig. S2). Transfection of these HITS siRNAs in HCT 116 cells significantly decreased the level of pGSK3βS9 compared with that in cells transfected with control siRNA, but did not affect the GSK3β expression (Fig. 1D-F). The present data demonstrated that HITS promoted the phosphorylation of GSK3β at S9 in CRC cells.

HS induces GSK3β S9 phosphorylation via HITS upregulation

Our previous study showed that HS induced HITS protein expression both in vitro and in vivo (17). To study the transcriptional response to HS in detail, the levels of HITS mRNA were monitored in HCT 116 cells in response to HS (42°C, 1 h). Semi-quantitative RT-PCR analysis showed that the level of HITS mRNA increased relatively slowly following HS, with its expression peaking at 12 h after HS (Fig. 2A). The level of pGSK3βS9 in the cells, measured using western blotting, increased significantly at 24 and 30 h after HS compared with their respective controls (Fig. 2B and C). This was likely caused by HITS upregulation in response to HS, since HITS knockdown in the heat-shocked cells significantly decreased the level of pGSK3βS9 at 24 h after HS compared with the control group (Fig. 2E and F). Significantly increased expression of GSK3β was observed 18 h after HS compared with the control (Fig. 2B and D); however, HITS knockdown induced no significant change in the GSK3β expression (Fig. 2E and G).

HITS suppresses cell migration but not proliferation

It is hypothesized that HITS is involved in suppressing tumor progression via phosphorylation-dependent deactivation of GSK3β. To investigate the effects of HITS on the migration of CRC cells, a wound healing assay was performed on HCT 116 cells transfected with a pEGFP control vector expressing GFP or pCAG-HITS-IRES-EGFP vector expressing both HITS and GFP. Compared with that in the control cells, cells overexpressing HITS showed a significant suppression of cell migration (Fig. 3A and B). This was not due to a cell proliferation suppression caused by HITS because no significant changes were observed in cell proliferation upon HITS overexpression (Fig. 3C).

MMPs are key proteases involved in cancer cell migration, invasion and metastasis by degrading the extracellular matrix (ECM) (36). MMP-3 and MMP-13 have been reported to be regulated by GSK3β (37); therefore, the present study investigated if HITS caused any changes in the expression of MMPs. Semi-quantitative RT-PCR and western blotting analysis revealed significant downregulation of MMP-3 and MMP-13 mRNA and protein in the HITS overexpressing cells (Fig. 3D-F), indicating that HITS suppressed cell motility, at least in part, by downregulating MMP-3 and MMP-13.

Anti-migratory effect of the HITS-GSK3β pathway offsets the pro-migratory effects of HSPs after HS

Previous studies have indicated that, while HS increases HITS expression, HS also induces expression of HSPs, including HSP90 (17,38,39), which in turn promotes the migration of cancer cells (15,16,4042). Therefore, it remains unclear whether the overall effects of HS on CRC cell migration are promotive or suppressive. Thus, a wound healing assay was performed to compare the migration of cells with and without HS. To cover all the time points where the S9 phosphorylation or the protein expression of GSK3β was upregulated by HS, HCT 116 cells were scratched and observed at 14 and 38 h (0 and 24 h in the wound healing timeframe, respectively) after HS. No significant change was observed in the migration of these cells (Fig. 4A), suggesting that the anti-migratory effect of the HITS-GSK3β pathway offset the pro-migratory activities of HSPs after HS. To understand the HITS-GSK3β effect, HITS siRNAs were transfected into HCT 116 cells under the same HS conditions, and a wound healing assay was performed following the time schedule shown in Fig. 4B. The wound healing rate was significantly increased in cells transfected with HITS siRNAs compared with that in the control, demonstrating that HITS decreased the migration of the cells induced with HS (Fig. 4C and D). Subsequently, the pro-migratory effect of HSP90 was further demonstrated using 17-AAG which is metabolized by NAD(P)H:quinone oxidoreductase 1 (NQO1) and selectively inhibits HSP90 (43). 17-AAG decreased the wound healing rate of HS-treated cells (Fig. 4C and D). Moreover, the migratory effect of the HITS knockdown was not observed in cells treated with 17-AAG, which suggested that the pro-migratory pathway mediated by HSP90 was the target of HITS. The function of HITS was mediated by GSK3β because the treatment with the specific GSK3β inhibitor, AR-A014418 (44) counteracted the effects of the HITS knockdown on cell migration (Fig. 4C and D). Taken together, the present findings indicated that HS not only induced the pro-migratory pathways mediated by HSPs, but also induced the anti-migratory pathway involving HITS and GSK3β, and that HITS-mediated GSK3β deactivation sufficiently offset the pro-migratory activity in CRC cells.

Discussion

The present study explored the effects of HS on CRC cell migration by focusing on HITS and its putative downstream target GSK3β. The level of pGSK3βS9 (GSK3β inactive form) was significantly increased in CRC cells overexpressing HITS, whereas its knockdown showed an opposite effect. These data demonstrated that HITS expression suppressed GSK3β activity by increasing the levels of pGSK3βS9. Our previous studies have indicated that the deactivation of GSK3β by S9 phosphorylation suppresses tumor progression in various CRC cells such as RKO, SW480, SW48, SW620 and HT29, as well as HCT 116 in vitro (29) and xenograft in vivo (35). The wound healing assay showed that overexpression of HITS in the CRC cells significantly suppressed cell migration compared with that of the control cells. This was most likely mediated by GSK3β deactivation because the positive effect of HITS knockdown on cell migration was abolished by treatment with the GSK3β inhibitor AR-A014418.

The molecular mechanisms underlying the anti-migratory and anti-invasive effects of GSK3β inhibition are of interest. Previous studies have indicated that the inhibition of GSK3β suppresses F-actin assembly and decreases the formation of lamellipodia and invadopodia via suppression of the phosphorylation-dependent activity of focal adhesion kinase (FAK) (27,28,45), c-Jun N-terminal kinase (JNK) (27) and adenylyl cyclase-associated protein 1 (CAP1) (46,47), the deactivation of guanine nucleotide-exchange factors (GEF) and Ras-related C3 botulinum toxin substrate 1 (RAC1) (27,28,48) and the degradation of nuclear factor of activated T cells (NFAT) (49). Potential involvement of these functional molecules in suppression of CRC cells migration upon the S9 phosphorylation-mediated deactivation of GSK3β is an aim of our future studies to investigate mechanisms by which HITS regulates tumor cell migration. The deactivation of GEF/RAC1 and JNK pathways reduces the expression of MMP-2 and membrane type 1 (MT1)-MMP, resulting in the suppression of invadopodia formation in glioblastoma and pancreatic cancer cells (27,28). The present study showed that HITS decreased the expression of MMP-3 and MMP-13 in CRC cells in association with attenuated cell migration (50,51). Therefore, the anti-migratory effect of HITS in cancer cells might be mediated by suppressing these MMPs.

A previous study reported that DRR1, which shares a highly homologous region with HITS but is not induced by HS (18,19,52), can directly bind actin (53). This study elucidated the two actin-binding regions of DRR1, of which the N-terminal region is highly homologous to HITS (70% amino acid identity) and is necessary for suppressing F-actin elongation in HeLa cells. It is possible that HITS also directly binds to actin to suppress F-actin assembly. Guo et al (54) indicated that the downregulation of HITS by S100A4, a pro-metastatic protein, increases the migration of gastric cancer MGC803 cells. Considering that S100A4 has been reported to bind F-actin and myosin II heavy chain to promote cytoskeletal formation (55,56), the direct binding of HITS to actin may disturb the interaction between S100A4 and the actin/myosin II heavy chain, resulting in the suppression of cell migration. In conjunction with these previous studies, the present findings suggested that HITS was involved in suppressing actin cytoskeleton formation directly and/or via S9 phosphorylation-mediated deactivation of GSK3β, which suppresses the formation of lamellipodia and invadopodia to prevent the CRC cell migration that is necessary for invasion (Fig. 5).

A number of previous studies have indicated that HSPs promote the migration, invasion and metastasis of cancer cells (15,16,5759). Among the HSPs, HSP90 was shown to act as a molecular chaperone able to stabilize and maintain oncoproteins involved in CRC progression such as mutated TP53 (gain-of-function), HER2 and BRAF, especially in stress conditions caused by chemotherapy, radiotherapy and hyperthermia (60,61). GSK3β has also been shown to be one of the client proteins of HSP90 (6264), which in turn phosphorylates HSP90 and HSP70 to facilitate cancer progression (65). The upregulation of HSP90 in HCT 116 cells was indicated in a previous report (66). Consistently, the increased expression of GSK3β observed after HS might be due to the chaperoning function of HSP90 and other HSPs. It has been indicated that HSP90 chaperoning function is required for maintaining the expression and/or function of FAK, NFAT, JNK and MMPs (6771) that promote the cell migration under the control of GSK3β, as discussed above. The present study indicated that GSK3β was involved in the major pro-migratory pathways mediated by HSP90 after HS, and that the inhibition of GSK3β caused by HITS played a critical role in attenuating the pro-migratory effect of GSK3β (Fig. 5). Indeed, HS did not induce significant changes in the migration of HCT 116 cells, although HSP90 was activated and showed pro-migratory activity. This was most likely due to the deactivation of GSK3β caused by HITS because HITS knockdown, under the same HS condition, showed an increase in wound healing rate, which in turn was decreased by treatment with AR-A014418. Treatment with 17-AAG counteracted the effect of the HITS knockdown, confirming the HSP90 chaperoning effect on GSK3β and its downstream pathways (Fig. 5). Other studies have also reported the anti-migratory and anti-metastatic effects of hyperthermia in vitro or in animal models, although the underlying mechanisms remain unclear (7274). In these reports, the HITS-GSK3β pathway might have played a critical role in suppressing cell migration in these reports.

The single or combined use of HSPs inhibitors with other therapies were proposed as attractive strategies for cancer therapy (75,76). Several studies have proposed to use hyperthermia in combination with HSP inhibitors, such as HSP90 inhibitors, which provides successful in vitro results (7779). However, HSPs are involved in the activation of the immune response, including the cytotoxicity of NK cells, maturation and antigen presentation of dendritic cells, and activation of T cells (11,13,14). Previous studies have shown that CRC is a heterogeneous disease consisting of the right-side colon cancer and the left-side colorectal cancer with different molecular, biological and clinicopathological properties (8082). Consistently, the antitumor immunity activated by HSPs would be beneficial to prevent peritoneal metastasis of the right-sided (proximal) colon cancer with highly immunogenic and intensive lymphocytic infiltration (81,82). Therefore, the combined use of HSPs inhibitors with hyperthermia may reduce the clinical benefits of hyperthermia. The present findings showed that the HITS-GSK3β pathway prevented hyperthermia-induced cancer cell migration caused by HSP90. One concern of the present study is that, in certain cancer types, HS may not induce HITS considering that the endogenous HITS expression has been reported to be suppressed in various cancer types (17,20,83,84) possibly due to epigenetic modification (8486). The present data showed that treatment with a GSK3β inhibitor reinforced the anti-migratory effect via HITS, while other studies also showed the benefit of GSK3β inhibitors to activate NK cells and dendritic cells and suppress the immune checkpoint protein PD-1 to enhance CD8+ T cells activity (8790). Grassilli et al (91) also indicated another benefit of GSK3β inhibitors to overcome drug resistance of p53 null colon carcinoma by inducing necroptosis. Accordingly, as well as an investigation of the putative roles of the pathways mediated by HITS and GSK3β in CRC biology as discussed in the former paragraph, the combination use of GSK3β inhibitors with hyperthermia would be an aim of our future studies.

Hyperthermia in cancer treatment shows clinical benefits because of its antitumor effects exerted through several processes, such as direct heat-induced cell death, induction of oxidative stress and molecular damage, antitumor immunity boosting, radio- and chemo-sensitivity enhancement, tumor microenvironment modifications, and inhibition of epithelial-to-mesenchymal transition (1014). While the HSF1-HSPs system, major cellular responses to HS, has long been considered to have a promigratory function that leads to cancer progression, the present study clarified that HITS could counterbalance the migratory effects caused by major HSPs (15,16), such as HSP90 in CRC cells. Further investigation is required to fully understand the molecular mechanisms underlying the antitumor effects of hyperthermia.

Supplementary Material

Supporting Data

Acknowledgements

The authors thank Dr Keiko Nakao (Saitama Medical University, Moroyama, Japan) for providing DNA vectors and technical support.

Funding

This study was supported in part by the Extramural Collaborative Research Grant of Cancer Research Institute, Kanazawa University. This study was also supported by a fund from Ageo Central General Hospital.

Availability of data and materials

The datasets used and/or analyzed during of current study are available from the corresponding author on reasonable request.

Authors' contributions

KK conceptualized the study, designed the experiments, acquired data and wrote the primary manuscript. TD acquired data and resources, designed the methodology and critically revised and proofread the manuscript. TM and KS acquired resources, designed methodology and critically revised and proofread the manuscript. HN conceptualized the study, acquired resources, designed methodology and critically revised and proofread the manuscript. All authors read and approved the final version of the manuscript. KK and HN confirm the authenticity of all the raw data.

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 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021. View Article : Google Scholar : PubMed/NCBI

3 

Leporrier J, Maurel J, Chiche L, Bara S, Segol P and Launoy G: A population-based study of the incidence, management and prognosis of hepatic metastases from colorectal cancer. Br J Surg. 93:465–474. 2006. View Article : Google Scholar : PubMed/NCBI

4 

Zarour LR, Anand S, Billingsley KG, Bisson WH, Cercek A, Clarke MF, Coussens LM, Gast CE, Geltzeiler CB, Hansen L, et al: Colorectal cancer liver metastasis: Evolving paradigms and future directions. Cell Mol Gastroenterol Hepatol. 3:163–173. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Dillekås H, Rogers MS and Straume O: Are 90% of deaths from cancer caused by metastases? Cancer Med. 8:5574–5576. 2019. View Article : Google Scholar : PubMed/NCBI

6 

Chow FC and Chok KS: Colorectal liver metastases: An update on multidisciplinary approach. World J Hepatol. 11:150–172. 2019. View Article : Google Scholar : PubMed/NCBI

7 

Ganesh K, Stadler ZK, Cercek A, Mendelsohn RB, Shia J, Segal NH and Diaz LA Jr: Immunotherapy in colorectal cancer: rationale, challenges and potential. Nat Rev Gastroenterol Hepatol. 16:361–375. 2019. View Article : Google Scholar : PubMed/NCBI

8 

Van der Jeught K, Xu HC, Li YJ, Lu XB and Ji G: Drug resistance and new therapies in colorectal cancer. World J Gastroenterol. 24:3834–3848. 2018. View Article : Google Scholar : PubMed/NCBI

9 

George TJ, Franke AJ, Chakravarthy AB, Das P, Dasari A, El-Rayes BF, Hong TS, Kinsella TJ, Landry JC, Lee JJ, et al: National Cancer Institute (NCI) state of the science: Targeted radiosensitizers in colorectal cancer. Cancer. 125:2732–2746. 2019.PubMed/NCBI

10 

Hettinga JV, Konings AW and Kampinga HH: Reduction of cellular cisplatin resistance by hyperthermia-a review. Int J Hyperthermia. 13:439–457. 1997. View Article : Google Scholar : PubMed/NCBI

11 

Yagawa Y, Tanigawa K, Kobayashi Y and Yamamoto M: Cancer immunity and therapy using hyperthermia with immunotherapy, radiotherapy, chemotherapy, and surgery. J Cancer Metastasis Treat. 3:218–230. 2017. View Article : Google Scholar

12 

Vassos N and Piso P: Metastatic colorectal cancer to the peritoneum: Current treatment options. Curr Treat Options Oncol. 19:492018. View Article : Google Scholar : PubMed/NCBI

13 

Dayanc BE, Beachy SH, Ostberg JR and Repasky EA: Dissecting the role of hyperthermia in natural killer cell mediated anti-tumor responses. Int J Hyperthermia. 24:41–56. 2008. View Article : Google Scholar : PubMed/NCBI

14 

Tsan MF and Gao B: Heat shock proteins and immune system. J Leukoc Biol. 85:905–910. 2009. View Article : Google Scholar : PubMed/NCBI

15 

Ciocca DR and Calderwood SK: Heat shock proteins in cancer: Diagnostic, prognostic, predictive, and treatment implications. Cell Stress Chaperones. 10:86–103. 2005. View Article : Google Scholar : PubMed/NCBI

16 

Boroumand N, Saghi H, Avan A, Bahreyni A, Ryzhikov M, Khazaei M and Hassanian SM: Therapeutic potency of heat-shock protein-90 pharmacological inhibitors in the treatment of gastrointestinal cancer, current status and perspectives. J Pharm Pharmacol. 70:151–158. 2018. View Article : Google Scholar : PubMed/NCBI

17 

Nakajima H, Ishigaki Y, Xia QS, Ikeda T, Yoshitake Y, Yonekura H, Nojima T, Tanaka T, Umehara H, Tomosugi N, et al: Induction of HITS, a newly identified family with sequence similarity 107 protein (FAM107B), in cancer cells by heat shock stimulation. Int J Oncol. 37:583–593. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Liu Q, Zhao XY, Bai RZ, Liang SF, Nie CL, Yuan Z, Wang CT, Wu Y, Chen LJ and Wei YQ: Induction of tumor inhibition and apoptosis by a candidate tumor suppressor gene DRR1 on 3p21.1. Oncol Rep. 22:1069–1075. 2009.PubMed/NCBI

19 

Schmidt MV, Schülke JP, Liebl C, Stiess M, Avrabos C, Bock J, Wochnik GM, Davies HA, Zimmermann N, Scharf SH, et al: Tumor suppressor down-regulated in renal cell carcinoma 1 (DRR1) is a stress-induced actin bundling factor that modulates synaptic efficacy and cognition. Proc Natl Acad Sci USA. 108:17213–17218. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Nakajima H, Koizumi K, Tanaka T, Ishigaki Y, Yoshitake Y, Yonekura H, Sakuma T, Fukushima T, Umehara H, Ueno S, et al: Loss of HITS (FAM107B) expression in cancers of multiple organs: Tissue microarray analysis. Int J Oncol. 41:1347–1357. 2012. View Article : Google Scholar : PubMed/NCBI

21 

Domoto T, Pyko IV, Furuta T, Miyashita K, Uehara M, Shimasaki T, Nakada M and Minamoto T: Glycogen synthase kinase-3β is a pivotal mediator of cancer invasion and resistance to therapy. Cancer Sci. 107:1363–1372. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Domoto T, Uehara M, Bolidong D and Minamoto T: Glycogen synthase kinase 3β in cancer biology and treatment. Cells. 9:13882020. View Article : Google Scholar : PubMed/NCBI

23 

Beurel E, Grieco SF and Jope RS: Glycogen synthase kinase-3 (GSK3): Regulation, actions, and diseases. Pharmacol Ther. 148:114–131. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Turano M, Costabile V, Cerasuolo A, Duraturo F, Liccardo R, Delrio P, Pace U, Rega D, Dodaro CA, Milone M, et al: Characterisation of mesenchymal colon tumour-derived cells in tumourspheres as a model for colorectal cancer progression. Int J Oncol. 53:2379–2396. 2018.PubMed/NCBI

25 

Kazi A, Xiang S, Yang H, Delitto D, Trevino J, Jiang RHY, Ayaz M, Lawrence HR, Kennedy P and Sebti SM: GSK3 suppression upregulates β-catenin and c-Myc to abrogate KRas-dependent tumors. Nat Commun. 9:51542018. View Article : Google Scholar : PubMed/NCBI

26 

Yoshino Y, Suzuki M, Takahashi H and Ishioka C: Inhibition of invasion by glycogen synthase kinase-3 beta inhibitors through dysregulation of actin re-organisation via down-regulation of WAVE2. Biochem Biophys Res Commun. 464:275–280. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Chikano Y, Domoto T, Furuta T, Sabit H, Kitano-Tamura A, Pyko IV, Takino T, Sai Y, Hayashi Y, Sato H, et al: Glycogen synthase kinase 3β sustains invasion of glioblastoma via the focal adhesion kinase, Rac1, and c-Jun N-terminal kinase-mediated pathway. Mol Cancer Ther. 14:564–574. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Kitano A, Shimasaki T, Chikano Y, Nakada M, Hirose M, Higashi T, Ishigaki Y, Endo Y, Takino T, Sato H, et al: Aberrant glycogen synthase kinase 3β is involved in pancreatic cancer cell invasion and resistance to therapy. PLoS One. 8:e552892013. View Article : Google Scholar : PubMed/NCBI

29 

Shakoori A, Ougolkov A, Yu ZW, Zhang B, Modarressi MH, Billadeau DD, Mai M, Takahashi Y and Minamoto T: Deregulated GSK3β activity in colorectal cancer: Its association with tumor cell survival and proliferation. Biochem Biophys Res Commun. 334:1365–1373. 2005. View Article : Google Scholar : PubMed/NCBI

30 

Kawauchi T, Chihama K, Nabeshima Y and Hoshino M: The in vivo roles of STEF/Tiam1, Rac1 and JNK in cortical neuronal migration. EMBO J. 22:4190–4201. 2003. View Article : Google Scholar : PubMed/NCBI

31 

Gayet J, Zhou XP, Duval A, Rolland S, Hoang JM, Cottu P and Hamelin R: Extensive characterization of genetic alterations in a series of human colorectal cancer cell lines. Oncogene. 20:5025–5032. 2001. View Article : Google Scholar : PubMed/NCBI

32 

Ahmed D, Eide PW, Eilertsen IA, Danielsen SA, Eknæs M, Hektoen M, Lind GE and Lothe RA: Epigenetic and genetic features of 24 colon cancer cell lines. Oncogenesis. 2:e712013. View Article : Google Scholar : PubMed/NCBI

33 

Mai W, Miyashita K, Shakoori A, Zhang B, Yu ZW, Takahashi Y, Motoo Y, Kawakami K and Minamoto T: Detection of active fraction of GSK3β in cancer cells by nonradioisotopic in vitro kinase assay. Oncology. 71:297–305. 2006. View Article : Google Scholar : PubMed/NCBI

34 

Shakoori A, Mai W, Miyashita K, Yasumoto K, Takahashi Y, Ooi A, Kawakami K and Minamoto T: Inhibition of GSK-3β activity attenuates proliferation of human colon cancer cells in rodents. Cancer Sci. 98:1388–1393. 2007. View Article : Google Scholar : PubMed/NCBI

35 

Mai W, Kawakami K, Shakoori A, Kyo S, Miyashita K, Yokoi K, Jin MJ, Shimasaki T, Motoo Y and Minamoto T: Deregulated glycogen synthase kinase 3β sustains gastrointestinal cancer cells survival by modulating human telomerase reverse transcriptase and telomerase. Clin Cancer Res. 15:6810–6819. 2009. View Article : Google Scholar : PubMed/NCBI

36 

Turunen SP, Tatti-Bugaeva O and Lehti K: Membrane-type matrix metalloproteases as diverse effectors of cancer progression. Biochim Biophys Acta Mol Cell Res. 1864:1974–1988. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Ning Q, Gan YH, Shi RR and Meng JH: Effects of HDAC4 on IL-1β-induced matrix metalloproteinase expression regulated partially through the WNT3A/β-catenin pathway. Chin Med J (Engl). 134:963–970. 2021. View Article : Google Scholar : PubMed/NCBI

38 

Tu Y, Tian Y, Wu Y and Cui S: Clinical significance of heat shock proteins in gastric cancer following hyperthermia stress: Indications for hyperthermic intraperitoneal chemoperfusion therapy. Oncol Lett. 15:9385–9391. 2018.PubMed/NCBI

39 

Grimmig T, Moll EM, Kloos K, Thumm R, Moench R, Callies S, Kreckel J, Vetterlein M, Pelz J, Polat B, et al: Upregulated heat shock proteins after hyperthermic chemotherapy point to induced cell survival mechanisms in affected tumor cells from peritoneal carcinomatosis. Cancer Growth Metastasis. 10:11790644177305592017. View Article : Google Scholar : PubMed/NCBI

40 

Chen JS, Hsu YM, Chen CC, Chen LL, Lee CC and Huang TS: Secreted heat shock protein 90α induces colorectal cancer cell invasion through CD91/LRP-1 and NF-κB-mediated integrin αV expression. J Biol Chem. 285:25458–25466. 2010. View Article : Google Scholar : PubMed/NCBI

41 

Song D, Guo M, Xu S, Song X, Bai B, Li Z, Chen J, An Y, Nie Y, Wu K, et al: HSP90-dependent PUS7 overexpression facilitates the metastasis of colorectal cancer cells by regulating LASP1 abundance. J Exp Clin Cancer Res. 40:1702021. View Article : Google Scholar : PubMed/NCBI

42 

Sims JD, McCready J and Jay DG: Extracellular heat shock protein (HSP)70 and HSP90α assist in matrix metalloproteinase-2 activation and breast cancer cell migration and invasion. PLoS One. 6:e188482011. View Article : Google Scholar : PubMed/NCBI

43 

Guo W, Reigan P, Siegel D, Zirrolli J, Gustafson D and Ross D: Formation of 17-allylamino-demethoxygeldanamycin (17-AAG) hydroquinone by NAD(P)H:quinone oxidoreductase 1: Role of 17-AAG hydroquinone in heat shock protein 90 inhibition. Cancer Res. 65:10006–10015. 2005. View Article : Google Scholar : PubMed/NCBI

44 

Bhat R, Xue Y, Berg S, Hellberg S, Ormö M, Nilsson Y, Radesäter AC, Jerning E, Markgren PO, Borgegård T, et al: Structural insights and biological effects of glycogen synthase kinase 3-specific inhibitor AR-A014418. J Biol Chem. 278:45937–45945. 2003. View Article : Google Scholar : PubMed/NCBI

45 

John JK, Paraiso KH, Rebecca VW, Cantini LP, Abel EV, Pagano N, Meggers E, Mathew R, Krepler C, Izumi V, et al: GSK3β inhibition blocks melanoma cell/host interactions by downregulating N-cadherin expression and decreasing FAK phosphorylation. J Invest Dermatol. 132:2818–2827. 2012. View Article : Google Scholar : PubMed/NCBI

46 

Wu H, Hasan R, Zhang H, Gray J, Williams D, Miller M, Allen F, Lee V, Kelly T and Zhou GL: Phosphorylation regulates CAP1 (cyclase-associated protein 1) functions in the motility and invasion of pancreatic cancer cells. Sci Rep. 9:49252019. View Article : Google Scholar : PubMed/NCBI

47 

Zhou GL, Zhang H, Wu H, Ghai P and Field J: Phosphorylation of the cytoskeletal protein CAP1 controls its association with cofilin and actin. J Cell Sci. 127:5052–5065. 2014.PubMed/NCBI

48 

Rom S, Fan S, Reichenbach N, Dykstra H, Ramirez SH and Persidsky Y: Glycogen synthase kinase 3β inhibition prevents monocyte migration across brain endothelial cells via Rac1-GTPase suppression and down-regulation of active integrin conformation. Am J Pathol. 181:1414–1425. 2012. View Article : Google Scholar : PubMed/NCBI

49 

Yoeli-Lerner M, Chin YR, Hansen CK and Toker A: Akt/protein kinase B and glycogen synthase kinase-3β signaling pathway regulates cell migration through the NFAT1 transcription factor. Mol Cancer Res. 7:425–432. 2009. View Article : Google Scholar : PubMed/NCBI

50 

Zhao J, Xu J, Zhao J and Zhang R: EFEMP2 promotes colon cancer cell invasion and growth through the ERK1/2 signaling pathway. Int J Clin Exp Pathol. 12:851–856. 2019.PubMed/NCBI

51 

Rath T, Stöckle J, Roderfeld M, Tschuschner A, Graf J and Roeb E: Matrix metalloproteinase-13 is regulated by toll-like receptor-9 in colorectal cancer cells and mediates cellular migration. Oncol Lett. 2:483–488. 2011. View Article : Google Scholar : PubMed/NCBI

52 

Nakajima H and Koizumi K: Family with sequence similarity 107: A family of stress responsive small proteins with diverse functions in cancer and the nervous system (Review). Biomed Rep. 2:321–325. 2014. View Article : Google Scholar : PubMed/NCBI

53 

Kretzschmar A, Schülke JP, Masana M, Dürre K, Müller MB, Bausch AR and Rein T: The stress-inducible protein DRR1 exerts distinct effects on actin dynamics. Int J Mol Sci. 19:39932018. View Article : Google Scholar : PubMed/NCBI

54 

Guo J, Bian Y, Wang Y, Chen L, Yu A and Sun X: FAM107B is regulated by S100A4 and mediates the effect of S100A4 on the proliferation and migration of MGC803 gastric cancer cells. Cell Biol Int. 41:1103–1109. 2017. View Article : Google Scholar : PubMed/NCBI

55 

Allgöwer C, Kretz AL, von Karstedt S, Wittau M, Henne-Bruns D and Lemke J: Friend or foe: S100 proteins in cancer. Cancers (Basel). 12:20372020. View Article : Google Scholar : PubMed/NCBI

56 

Chen M, Bresnick AR and O'Connor KL: Coupling S100A4 to rhotekin alters Rho signaling output in breast cancer cells. Oncogene. 32:3754–3764. 2013. View Article : Google Scholar : PubMed/NCBI

57 

Huang CY, Wei PL, Chen WY, Chang WC and Chang YJ: Silencing heat shock protein 27 inhibits the progression and metastasis of colorectal cancer (CRC) by maintaining the stability of stromal interaction molecule 1 (STIM1) proteins. Cells. 7:2622018. View Article : Google Scholar : PubMed/NCBI

58 

Lin Y, Peng N, Zhuang H, Zhang D, Wang Y and Hua ZC: Heat shock proteins HSP70 and MRJ cooperatively regulate cell adhesion and migration through urokinase receptor. BMC Cancer. 14:6392014. View Article : Google Scholar : PubMed/NCBI

59 

Moser C, Lang SA, Kainz S, Gaumann A, Fichtner-Feigl S, Koehl GE, Schlitt HJ, Geissler EK and Stoeltzing O: Blocking heat shock protein-90 inhibits the invasive properties and hepatic growth of human colon cancer cells and improves the efficacy of oxaliplatin in p53-deficient colon cancer tumors in vivo. Mol Cancer Ther. 6:2868–2878. 2007. View Article : Google Scholar : PubMed/NCBI

60 

Hagn F, Lagleder S, Retzlaff M, Rohrberg J, Demmer O, Richter K, Buchner J and Kessler H: Structural analysis of the interaction between Hsp90 and the tumor suppressor protein p53. Nat Struct Mol Biol. 18:1086–1093. 2011. View Article : Google Scholar : PubMed/NCBI

61 

Lacey T and Lacey H: Linking hsp90′s role as an evolutionary capacitator to the development of cancer. Cancer Treat Res Commun. 28:1004002021. View Article : Google Scholar : PubMed/NCBI

62 

Dou F, Chang X and Ma D: Hsp90 maintains the stability and function of the Tau phosphorylating kinase GSK3β. Int J Mol Sci. 8:51–60. 2007. View Article : Google Scholar

63 

Banz VM, Medová M, Keogh A, Furer C, Zimmer Y, Candinas D and Stroka D: Hsp90 transcriptionally and post-translationally regulates the expression of NDRG1 and maintains the stability of its modifying kinase GSK3β. Biochim Biophys Acta. 1793:1597–1603. 2009. View Article : Google Scholar : PubMed/NCBI

64 

Tang W, Wu Y, Qi X, Yu R, Lu Z, Chen A, Fan X and Li J: PGK1-coupled HSP90 stabilizes GSK3β expression to regulate the stemness of breast cancer stem cells. Cancer Biol Med. 19:486–503. 2022. View Article : Google Scholar : PubMed/NCBI

65 

Muller P, Ruckova E, Halada P, Coates PJ, Hrstka R, Lane DP and Vojtesek B: C-terminal phosphorylation of Hsp70 and Hsp90 regulates alternate binding to co-chaperones CHIP and HOP to determine cellular protein folding/degradation balances. Oncogene. 32:3101–3110. 2013. View Article : Google Scholar : PubMed/NCBI

66 

Zhang C, Li S and Zhao Z: β-Elemene promotes apoptosis induced by hyperthermia via inhibiting HSP70. Dis Markers. 2022:73130262022.PubMed/NCBI

67 

Schwock J, Dhani N, Cao MP, Zheng J, Clarkson R, Radulovich N, Navab R, Horn LC and Hedley DW: Targeting focal adhesion kinase with dominant-negative FRNK or Hsp90 inhibitor 17-DMAG suppresses tumor growth and metastasis of SiHa cervical xenografts. Cancer Res. 69:4750–4759. 2009. View Article : Google Scholar : PubMed/NCBI

68 

Taiyab A and Rao ChM: HSP90 modulates actin dynamics: Inhibition of HSP90 leads to decreased cell motility and impairs invasion. Biochim Biophys Acta. 1813:213–221. 2011. View Article : Google Scholar : PubMed/NCBI

69 

Liu Z, Li H, He L, Xiang Y, Tian C, Li C, Tan P, Jing J, Tian Y, Du L, et al: Discovery of small-molecule inhibitors of the HSP90-calcineurin-NFAT pathway against glioblastoma. Cell Chem Biol. 26:352–365.e7. 2019. View Article : Google Scholar : PubMed/NCBI

70 

Lu C, Chen D, Zhang Z, Fang F, Wu Y, Luo L and Yin Z: Heat shock protein 90 regulates the stability of c-Jun in HEK293 Cells. Mol Cells. 24:210–214. 2007.PubMed/NCBI

71 

Stellas D, El Hamidieh A and Patsavoudi E: Monoclonal antibody 4C5 prevents activation of MMP2 and MMP9 by disrupting their interaction with extracellular HSP90 and inhibits formation of metastatic breast cancer cell deposits. BMC Cell Biol. 11:512010. View Article : Google Scholar : PubMed/NCBI

72 

Jia D, Rao W, Wang C, Jin C, Wang S, Chen D, Zhang M, Guo J, Chang Z and Liu J: Inhibition of B16 murine melanoma metastasis and enhancement of immunity by fever-range whole body hyperthermia. Int J Hyperthermia. 27:275–285. 2011. View Article : Google Scholar : PubMed/NCBI

73 

Byun YH, Gwak HS, Kwon JW, Song MK, Shin SH, Jo YH, Yoo H and Lee SH: Local recurrence of brain metastasis reduced by intra-operative hyperthermia treatment. Int J Hyperthermia. 35:168–175. 2019. View Article : Google Scholar : PubMed/NCBI

74 

Zhao J, Lv Y, Cai Y, Wei W, Yin C, Wang X, Hao Z, Shen C and Wang H: Hyperthermic carbon dioxide pneumoperitoneum reinforces the inhibition of 5-FU on the proliferation and invasion of colon cancer. Oncol Rep. 37:492–500. 2017. View Article : Google Scholar : PubMed/NCBI

75 

Kumar S, Stokes J III, Singh UP, Scissum Gunn K, Acharya A, Manne U and Mishra M: Targeting HSP70: A possible therapy for cancer. Cancer Lett. 374:156–166. 2016. View Article : Google Scholar : PubMed/NCBI

76 

Kryeziu K, Bruun J, Guren TK, Sveen A and Lothe RA: Combination therapies with HSP90 inhibitors against colorectal cancer. Biochim Biophys Acta Rev Cancer. 1871:240–247. 2019. View Article : Google Scholar : PubMed/NCBI

77 

Zhou L, Zhang M, Fu Q, Li J and Sun H: Targeted near infrared hyperthermia combined with immune stimulation for optimized therapeutic efficacy in thyroid cancer treatment. Oncotarget. 7:6878–6890. 2016. View Article : Google Scholar : PubMed/NCBI

78 

Vriend LEM, van den Tempel N, Oei AL, L'Acosta M, Pieterson FJ, Franken NAP, Kanaar R and Krawczyk PM: Boosting the effects of hyperthermia-based anticancer treatments by HSP90 inhibition. Oncotarget. 8:97490–97503. 2017. View Article : Google Scholar : PubMed/NCBI

79 

Daunys S, Matulis D and Petrikaitė V: Synergistic activity of HSP90 inhibitors and anticancer agents in pancreatic cancer cell cultures. Sci Rep. 9:161772019. View Article : Google Scholar : PubMed/NCBI

80 

Baran B, Mert Ozupek N, Yerli Tetik N, Acar E, Bekcioglu O and Baskin Y: Difference between left-sided and right-sided colorectal cancer: A focused review of literature. Gastroenterology Res. 11:264–273. 2018. View Article : Google Scholar : PubMed/NCBI

81 

de Vries NL, Swets M, Vahrmeijer AL, Hokland M and Kuppen PJ: The immunogenicity of colorectal cancer in relation to tumor development and treatment. Int J Mol Sci. 17:10302016. View Article : Google Scholar : PubMed/NCBI

82 

Picard E, Verschoor CP, Ma GW and Pawelec G: Relationships between immune landscapes, genetic subtypes and responses to immunotherapy in colorectal cancer. Front Immunol. 11:3692020. View Article : Google Scholar : PubMed/NCBI

83 

Zhang H, Du Y, Wang Z, Lou R, Wu J and Feng J: Integrated analysis of oncogenic networks in colorectal cancer identifies GUCA2A as a molecular marker. Biochem Res Int. 2019:64694202019. View Article : Google Scholar : PubMed/NCBI

84 

Mah WC, Thurnherr T, Chow PK, Chung AY, Ooi LL, Toh HC, Teh BT, Saunthararajah Y and Lee CG: Methylation profiles reveal distinct subgroup of hepatocellular carcinoma patients with poor prognosis. PLoS One. 9:e1041582014. View Article : Google Scholar : PubMed/NCBI

85 

Song MA, Tiirikainen M, Kwee S, Okimoto G, Yu H and Wong LL: Elucidating the landscape of aberrant DNA methylation in hepatocellular carcinoma. PLoS One. 8:e557612013. View Article : Google Scholar : PubMed/NCBI

86 

Kan S, Chai S, Chen W and Yu B: DNA methylation profiling identifies potentially significant epigenetically-regulated genes in glioblastoma multiforme. Oncol Lett. 18:1679–1688. 2019.PubMed/NCBI

87 

Parameswaran R, Ramakrishnan P, Moreton SA, Xia Z, Hou Y, Lee DA, Gupta K, deLima M, Beck RC and Wald DN: Repression of GSK3 restores NK cell cytotoxicity in AML patients. Nat Commun. 7:111542016. View Article : Google Scholar : PubMed/NCBI

88 

Cichocki F, Valamehr B, Bjordahl R, Zhang B, Rezner B, Rogers P, Gaidarova S, Moreno S, Tuininga K, Dougherty P, et al: GSK3 inhibition drives maturation of NK cells and enhances their antitumor activity. Cancer Res. 77:5664–5675. 2017. View Article : Google Scholar : PubMed/NCBI

89 

Noh KT, Son KH, Jung ID, Kang TH, Choi CH and Park YM: Glycogen synthase kinase-3β (GSK-3β) inhibition enhances dendritic cell-based cancer vaccine potency via suppression of interferon-γ-induced indoleamine 2,3-dioxygenase expression. J Biol Chem. 290:12394–12402. 2015. View Article : Google Scholar : PubMed/NCBI

90 

Taylor A, Harker JA, Chanthong K, Stevenson PG, Zuniga EI and Rudd CE: Glycogen synthase kinase 3 inactivation drives T-bet-mediated downregulation of co-receptor PD-1 to enhance CD8(+) cytolytic T cell responses. Immunity. 44:274–286. 2016. View Article : Google Scholar : PubMed/NCBI

91 

Grassilli E, Narloch R, Federzoni E, Ianzano L, Pisano F, Giovannoni R, Romano G, Masiero L, Leone BE, Bonin S, et al: Inhibition of GSK3B bypass drug resistance of p53-null colon carcinomas by enabling necroptosis in response to chemotherapy. Clin Cancer Res. 19:3820–3831. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2023
Volume 63 Issue 2

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Koizumi K, Domoto T, Minamoto T, Satomura K and Nakajima H: Deactivation of glycogen synthase kinase-3β by heat shock‑inducible tumor small protein attenuates hyperthermia‑induced pro‑migratory activity in colorectal cancer cells. Int J Oncol 63: 92, 2023.
APA
Koizumi, K., Domoto, T., Minamoto, T., Satomura, K., & Nakajima, H. (2023). Deactivation of glycogen synthase kinase-3β by heat shock‑inducible tumor small protein attenuates hyperthermia‑induced pro‑migratory activity in colorectal cancer cells. International Journal of Oncology, 63, 92. https://doi.org/10.3892/ijo.2023.5540
MLA
Koizumi, K., Domoto, T., Minamoto, T., Satomura, K., Nakajima, H."Deactivation of glycogen synthase kinase-3β by heat shock‑inducible tumor small protein attenuates hyperthermia‑induced pro‑migratory activity in colorectal cancer cells". International Journal of Oncology 63.2 (2023): 92.
Chicago
Koizumi, K., Domoto, T., Minamoto, T., Satomura, K., Nakajima, H."Deactivation of glycogen synthase kinase-3β by heat shock‑inducible tumor small protein attenuates hyperthermia‑induced pro‑migratory activity in colorectal cancer cells". International Journal of Oncology 63, no. 2 (2023): 92. https://doi.org/10.3892/ijo.2023.5540