Open Access

Glyoxalase 1 and protein kinase Cλ as potential therapeutic targets for late‑stage breast cancer

  • Authors:
    • Hitomi Motomura
    • Ayaka Ozaki
    • Shoma Tamori
    • Chotaro Onaga
    • Yuka Nozaki
    • Yuko Waki
    • Ryoko Takasawa
    • Kazumi Yoshizawa
    • Yasunari Mano
    • Tsugumichi Sato
    • Kazunori Sasaki
    • Hitoshi Ishiguro
    • Yohei Miyagi
    • Yoji Nagashima
    • Kouji Yamamoto
    • Keiko Sato
    • Takehisa Hanawa
    • Sei-Ichi Tanuma
    • Shigeo Ohno
    • Kazunori Akimoto
  • View Affiliations

  • Published online on: May 24, 2021     https://doi.org/10.3892/ol.2021.12808
  • Article Number: 547
  • Copyright: © Motomura et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

Cancer cells upregulate the expression levels of glycolytic enzymes in order to reach the increased glycolysis required. One such upregulated glycolytic enzyme is glyoxalase 1 (GLO 1), which catalyzes the conversion of toxic methylglyoxal to nontoxic S‑D‑lactoylglutathione. Protein kinase Cλ (PKCλ) is also upregulated in various types of cancer and is involved in cancer progression. In the present study, the association between enhanced glycolysis and PKCλ in breast cancer was investigated. In human breast cancer, high GLO 1 expression was associated with high PKCλ expression at the protein (P<0.01) and mRNA levels (P<0.01). Furthermore, Wilcoxon and Cox regression model analysis revealed that patients with stage III‑IV tumors with high GLO 1 and PKCλ expression had poor overall survival compared with patients expressing lower levels of these genes [P=0.040 (Gehan‑Breslow generalized Wilcoxon test) and P=0.031 (hazard ratio, 2.36; 95% confidence interval, 1.08‑5.16), respectively]. Treatment of MDA‑MB‑157 and MDA‑MB‑468 human basal‑like breast cancer cells with TLSC702 (a GLO 1 inhibitor) and/or aurothiomalate (a PKCλ inhibitor) reduced both cell viability and tumor‑sphere formation. These results suggested that GLO 1 and PKCλ were cooperatively involved in cancer progression and contributed to a poor prognosis in breast cancer. In conclusion, GLO 1 and PKCλ serve as potentially effective therapeutic targets for treatment of late‑stage human breast cancer.

Introduction

Breast cancer is the second most common type of cancer worldwide (1), and the most common type of cancer in women, with an estimated 2.09 million new cases (24.2% of all cases of cancer in women), and 0.6 million cancer-related deaths annually (2). The prognosis for breast cancer patients is generally good; however, patients with late-stage tumors (stages III and IV) have significantly shorter overall survival (OS) (3). This is due to the fact that late-stage breast cancers are often resistant or less responsive to conventional medical approaches, such as conventional surgery, chemotherapy and radiotherapy, and exhibit a high rate of both recurrence and metastasis (3). Thus, novel pharmacological approaches are required to manage late-stage cancer.

Breast cancer can be classified, based on gene expression patterns (PAM 50), into at least six subtypes: Normal-like, luminal A, luminal B, HER2-enriched, claudin-low and basal-like (47). Based on receptor expression status, breast cancer can also be classified immunohistochemically as estrogen receptor (ER)-positive and/or progesterone receptor (PgR)-positive type, HER2-positive type, and triple-negative type (ER-negative, PgR-negative, HER2-negative; TNBC). TNBC has the poorest prognosis amongst the different breast cancer subtypes, and 70–80% of TNBCs are basal-like breast cancer (8).

Metabolic reprogramming leading to increased glycolysis, termed the Warburg effect, is a characteristic feature of cancer cells (9). This enhancement of glycolysis in cancer cells contributes to their proliferation, migration, survival and drug resistance (10). In addition, glyoxalase 1 (GLO 1), which catalyzes the conversion of methylglyoxal (MG), a toxic byproduct of glycolysis, to non-toxic S-D-lactoylglutathione, is upregulated in several types of malignancy, including lung, stomach, colon, liver, prostate, oropharyngeal, skin and breast cancer (1121). GLO 1 is essential for the survival of aldehyde dehydrogenase 1 (ALDH1)-positive breast cancer stem cells, and operates in a caspase-3-dependent manner (21). In addition, HER2/neu signaling regulates GLO 1 expression in HER2-positive tissues and cell lines (22). However, the signal transduction mechanisms of GLO 1 in breast cancer remain unclear.

It is well established that the majority of cancer cells are derived from epithelial cells, and defects in cell polarity are a characteristic feature of cancer cells (23). One of the atypical protein kinase C (aPKC) isotypes, PKCλ/ι, is known to be involved in cellular responses that include determination of cell polarity, as well as cell proliferation, survival, chemotaxis and migration (2426). PKCλ is overexpressed in several types of cancer, including breast cancer (2742), and is known to be involved in cancer progression, contributing to poor clinical outcomes (3242). In TNBC cells, TGFβ and IL1β induce PKCλ phosphorylation and promote PKCλ-dependent proliferation, invasiveness and metastasis by inducing NF-κB p65 nuclear translocation (30). c-Met and PKCλ are cooperatively involved in cellular viability and tumor formation in basal-like breast cancer cells (42). PKCλ is also essential for the survival of ALDH1-positive breast cancer stem cells in a caspase-3-dependent manner (41,42). EGF, platelet-derived growth factor and insulin collectively activate PKCλ via PI3-kinase (4345), and PKCλ subsequently binds to and regulates p70 S6 kinase (46). PKCλ also activates the Rac1-Pak-Mek1/2-Erk1/2 signaling pathway, which is associated with lung cancer cell proliferation and tumorigenicity (47). PKCλ phosphorylates FoxO1 and modulates the DNA-binding ability of c-Myc, promoting cellular proliferation in angiosarcoma (48). Glucose transporter 1 (GLUT1) facilitates glucose transport and its expression is increased in several types of cancer (including breast cancer), where it is involved in enhanced glycolysis and cancer progression (49,50). PKCλ regulates the translocation of GLUT1 from intracellular vesicles to the plasma membrane in 3T3-L1 adipocytes (51). However, the role of PKCλ in the enhanced glycolysis seen in cancer cells remains unclear.

In the present study, the association between the levels of GLO 1 and PKCλ expression in human breast cancer was investigated, and their impact on the prognoses of patients with late-stage breast cancer was assessed.

Materials and methods

Immunohistochemistry (IHC)

Specimens used for IHC were prepared at the Kanagawa Cancer Center Research Institute from archives of surgically removed and formalin-fixed, paraffin-embedded breast cancer tissues in the Pathology Department. With the approval of the Research Ethics Committee, these prepared specimens were used in the present study through the Kanagawa Cancer Research and Information Association, which has since been dissolved and its duties transferred to the Kanagawa Cancer Center Research Institute Biospecimen Center (approval no. 3-2009). The clinicopathological data of the patients from whom the samples were obtained are summarized in Table SI. TNM stage data is lacking 27% because the data is already anonymized. The research protocol used was also approved by the Institutional Ethics Committees of Tokyo University of Science (approval nos. 13003, 15006 and 16038), and all patients provided consent for the use of their tissue samples for research purposes.

IHC was performed as previously described (29,3135,39,40). Briefly, 4-µm thick paraffin embedded sections were deparaffinized, rehydrated in a descending series of ethanol solutions and autoclaved (120°C for 20 min) in 10 mmol/l citrate buffer (pH 6.0) for antigen retrieval. The semi-serially prepared sections (adjacent sections) were then immersed in 0.3% hydrogen peroxide at room temperature for 30 min to quench the intrinsic peroxidase activity before incubation with a primary antibody at 4°C overnight. The antibodies used in the present study were: Mouse anti-PKCι mAb (1:250; cat. no. 610176; BD Biosciences), mouse anti-GLO 1 mAb (1:2,000; cat. no. NBP1-19015; Novus Biologicals, Inc.), mouse IgG2b κ Isotype Control (eBMG2b; 1:500; cat. no. 14-4732-82; eBioscience; Thermo Fisher Scientific, Inc.) and mouse IgG1 κ Isotype Control (P3.6.2.8.1) (1:1,000; cat. no. 14-4714-82; eBioscience; Thermo Fisher Scientific, Inc.). The labeled antigens were visualized using a Histo Fine kit (Nichirei) and DAB plus (Dako; Agilent Technologies, Inc.). The sections were counterstained with hematoxylin. The antibodies used for double staining were: Mouse anti-PKCι mAb (1:50), rabbit anti-GLO 1 pAb (1:200; cat. no. A1932; ABclonal, Inc.), mouse IgG2b κ Isotype Control and normal rabbit IgG (1:952; cat. no. PM035; MBL). The labeled antigens were visualized using a Histo Fine alkaline phosphatase kit and DAB plus. The sections were counterstained with hematoxylin.

IHC scoring

To evaluate the expression of GLO 1 and PKCλ proteins using IHC, ImageJ version 1.51u was used (National Institutes of Health) with the IHC Profiler plugin (52). The scoring system was based on the classification calculated from the IHC Profiler (+3, high-positive; +2, positive; +1, low-positive; and 0, negative). Signal intensity of GLO 1 was classified into color density as follows; +3, High positive; +2, Positive; and +1, Low positive. Signal intensity of PKCλ was classified into color density as follows: +3, High positive; +2, Positive; +1, Low positive; and 0, Negative. Signal intensities were categorized as high (+3 or +2) or low (+1 or 0). H-scores of the scatter plot data were based on calculated values from the IHC Profiler.

Analysis of gene expression in the breast cancer dataset from the molecular taxonomy of breast cancer international consortium (METABRIC)

Gene expression data was downloaded from cBioportal and analyzed as previously described (21,41,42,53,54). Briefly, the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) dataset (55,56) was downloaded from cBioPortal (cbioportal.org/; last entry, 25th November 2019) (57,58). The clinicopathological data of the patients are summarized in Table SII. The median age at the time of diagnosis was 61.8 years (age range, 21.9–96.3 years). Gene expression levels were classified as high if they were in the top 25% of Z-scores; or otherwise, they were classed as low.

Analysis of gene expression in the breast cancer dataset from The Cancer Genome Atlas (TCGA)

Briefly, gene expression microarray datasets from TCGA were downloaded from Oncomine (oncomine.org; Compendia Bioscience, 28th January 2021) (59,60), and the breast cancer dataset (n=459) was obtained. Levels of GLO 1 (reporter, A_32_P53822) and PKCλ (reporter, A_23_P18392) mRNA expression are presented using log2 median-centered ratio boxplots for normal vs. cancerous tissues.

Cell culture

The MCF-10A human normal-like (non-transformed) mammary epithelial cell line and the MDA-MB-157 and MDA-MB-468 human basal-like breast cancer cell lines were obtained from the American Type Culture Collection. MCF-10A cells were grown in mammary epithelial cell growth medium (MEGM; Lonza Group, Ltd.) according to instructions from ATCC. The cancer cell lines were cultured as previously described (21,41,42,53). Mycoplasma testing was performed on all the cell lines used.

Inhibitory compounds

3-(1,3-Benzothiazol-2-yl)-4- (4-methoxyphenyl) but-3-enoic acid (TLSC702) was purchased from Namiki Shoji Co., Ltd. and dissolved in DMSO. Aurothiomalate (ATM) was purchased from Calbiochem (Merck KGaA) and dissolved in water.

Immunoblot analysis

Immunoblotting was performed as previously described (21,41,42,53). The primary antibodies used were: Mouse anti-PKCι mAb (1:5,000; cat. no. 610176; BD Biosciences), mouse anti-GLO 1 mAb (1:2,000; cat. no. sc-133144, Santa Cruz Biotechnology, Inc.) and mouse anti-β-actin mAb (1:20,000; cat. no. 60008-1-Ig, ProteinTech Group Inc.). The secondary antibody used was a goat anti-mouse IgG horseradish peroxidase-conjugate (1:5,000; cat. no. 7076S; Cell Signaling Technology, Inc.).

WST-8 assay

WST-8 assays were performed according to the manufacturer's protocol, and as previously described (21,42,53). Briefly, cells (5×103/well) were seeded into 96-well plates (Sigma-Aldrich; Merck KGaA) and incubated for 24 h. Inhibitors were then added to the culture medium, and the cells were incubated for an additional 3 days, after which cell viability was assessed using a Cell Counting Kit-8 assay (Dojindo Molecular Technologies, Inc.). The formazan dye formed was measured using Sunrise Remote (Tecan Group, Ltd.) at 450 nm. Assays with MCF-10A cells were performed in MEGM supplemented with 10% FBS. Assays using the cancer cell lines were performed in DMEM supplemented with 10% FBS. Numerical values of the test groups were expressed relative to the control cell (no drug).

Tumor-sphere culture

Tumor-spheres were grown as previously described (21,41,42,53). Briefly, cells (1×103/well) were cultured in 96-well ultralow attachment plates (Greiner Bio-One) and treated with inhibitors for 6 days. Images were taken through an inverted microscope (Leica Microsystems, Inc.), and the numbers of tumor-spheres ≥50 µm in diameter were counted. Numerical values of the test groups are shown relative to the untreated cells. Cell Titer-Glo® luminescence assays (Promega Corporation) were performed according to the manufacturer's protocol, and as previously described (21,53). Values for test groups are shown relative to cells in the absence of the drug.

Statistical analysis

For correlation between protein GLO 1 and PKCλ expression, statistical significance was calculated using the χ2-test with Yates' correction. H-scores of the scatter plot data were based on calculated values from the IHC Profiler. Spearman's rank correlation coefficients (r) and P-value are indicated. In the analysis of gene expression, Pearson's correlation coefficients (r) and P-value are indicated. P-values were calculated using a test for non-correlation. In the analysis of gene expression, survival curves were plotted using the Kaplan-Meier method, and P-values were calculated using the Gehan-Breslow generalized Wilcoxon test to weight early death points. Multiplicity was adjusted using the Holm's method for post-hoc analysis. A multivariable Cox regression model was used to evaluate the effect of gene expression and to estimate the adjusted hazard ratios (HRs) with age as a confounding factor. P-values for comparison of gene expression are presented using the Kruskal-Wallis test with the Steel-Dwass test. Statistical analysis was performed using BellCurve for Excel version 2.11 (SSRI). Data for the WST-8 assay is presented as the mean ± standard deviation of three independent experiments. Differences between groups were compared using Tukey's test. Data for the tumor-sphere assay is presented as the mean ± standard error of the mean of three independent experiments. Data for the Cell Titer-Glo® luminescence assay is presented as the mean ± standard deviation of three independent experiments. Statistical significance was calculated using one-way ANOVA followed by Dunnett's test. For any of the analyses above, α-level was fixed at 0.05, and P<0.05 was considered to indicate a statistically significant difference.

Results

GLO 1 expression is positively correlated with PKCλ expression in breast cancer

GLO 1 protein expression in both the cytosol and nucleus of breast cancer cells was detected, and the results were consistent with earlier observations (Fig. 1A) (20). There were no 0 image samples for GLO 1 protein. GLO 1 protein expression was detected in all breast cancer samples, and the results were consistent with a previous report (20). PKCλ was also localized in the cytosol and nucleus of breast cancer cells (Fig. 1B) as reported in our previous study (29). Double IHC staining showed that GLO 1 and PKCλ were colocalized in breast cancer cells (Fig. 1C). To evaluate the relationship between GLO 1 and PKCλ, their signal intensities were quantified (Fig. 1A and B). High expression of GLO 1 was significantly correlated with high expression of PKCλ in breast cancer tissue (Fig. 1D, P<0.01, χ2-test; Fig. 1E, r=0.71, P<0.01).

GLO 1 expression is correlated with PKCλ expression at the mRNA level in human breast cancer

To further examine the relationship between GLO 1 and PKCλ at the mRNA level, mRNA expression data from the METABRIC breast cancer dataset (n=1,904) was downloaded from cBioPortal (Fig. 2A). In our previous study, it was shown that GLO 1 expression is higher in breast cancer compared with normal tissue samples (21). In addition, GLO 1 gene expression was enhanced at all breast cancer tumor stages compared with normal tissue samples. Similarly, PKCλ expression was significantly higher in breast cancer tissues and at all tumor stages compared with the normal tissues (41) (Fig. S1). Scatter plot analysis indicated that GLO 1 expression was weakly correlated with PKCλ expression in all patients and in patients with early-stage (stage 0-II) tumors (Fig. 2A), but was not correlated with PKCλ expression in patients with late-stage (stage III–IV) tumors (Fig. 2A). As shown in Table SIII, none of the clinicopathological parameters were correlated with high GLO 1 and PKCλ gene expression. Nonetheless, these results, together with the IHC findings, suggest that GLO 1 and PKCλ may be cooperatively involved in certain cases of breast cancer.

High GLO 1 and PKCλ expression is correlated with a poorer prognosis in patients with stage III–IV tumors

Next, we examined weather high GLO 1 and PKCλ expression is correlated with prognosis (Figs. 2B-D and S2; Tables I and SIV). Examination of the prognosis of patients with GLO 1high and PKCλhigh breast cancer revealed that GLO 1high was not associated with a poorer OS amongst all patients (Fig. 2B) (21). It was previously shown that overexpression of PKCλ and its signaling promotes TNBC growth and metastasis (30). Consistent with this finding, patients classed as PKCλhigh had worse OS (Fig. 2B; P<0.01) (42). In our previous study, it was shown that patients with stage III or IV cervical cancer with high PKCλ expression had a worse clinical outcome (32). Thus, tumors were classified into early-stage (stage 0-II) and late-stage (stage III–IV) tumors, and the difference in survival was assessed using Kaplan-Meier analysis. Patients with early-stage tumors (GLO 1high, PKCλhigh or GLO 1high PKCλhigh) exhibited similar clinical outcomes to those expressing lower levels of these genes (Fig. 2C), as did those with late-stage tumors (GLO 1high; Fig. 2D). However, patients with late-stage tumors (PKCλhigh) had a worse OS (Fig. 2D) (42). Furthermore, patients with GLO 1high PKCλhigh tumors had poorer prognoses than patients with GLO 1high PKCλlow (adjusted by Holm's method; P=0.015) and GLO 1low PKCλlow (P=0.040), but not GLO 1low PKCλhigh (P=0.83) tumors. Multivariable Cox regression analysis revealed that GLO 1high PKCλhigh was associated with poorer clinical outcomes than GLO 1low PKCλlow (Table I; HR=2.36; 95% CI, 1.08–5.16; P=0.03) and GLO 1high PKCλlow (Table I; HR=3.25, 95% CI, 1.26–8.35; P=0.01) in those with late-stage tumors. Furthermore, normal-like breast cancer patients classified as GLO 1high PKCλhigh also exhibited a worse prognosis (Fig. S2; adjusted by Holm's method; P=0.016 and Table SIV; HR=9.11, 95% CI, 2.07–40.15; P<0.01). Thus, high PKCλ expression, regardless of GLO 1 expression level, contributes to poor clinical outcome. Of note, GLO 1 protein (Fig. 1) and mRNA (Fig. S1) expression was considerably higher in breast cancer, reflecting enhanced glycolysis. Therefore, GLO 1 and PKCλ may function cooperatively to promote cancer progression, and contribute to worse clinical outcomes in patients with late-stage breast cancer.

Table I.

Multivariable Cox regression analysis of the association between GLO 1 and PKCλ expression and breast cancer in all patients, and in patients stratified by stage (0-II and III–IV).

Table I.

Multivariable Cox regression analysis of the association between GLO 1 and PKCλ expression and breast cancer in all patients, and in patients stratified by stage (0-II and III–IV).

ComparisonHazard ratioa (95% confidence interval)P-value
All
  GLO 1high vs. GLO 1low1.06 (0.93–1.22)0.39
  PKCλhigh vs. PKCλlow1.20 (1.05–1.38)<0.01
  GLO 1high PKCλhigh vs. GLO 1low PKCλlow1.18 (0.94–1.49)0.15
  GLO 1high PKCλhigh vs. GLO 1low PKCλhigh0.95 (0.74–1.22)0.69
  GLO 1high PKCλhigh vs. GLO 1high PKCλlow1.08 (0.84–1.40)0.54
Stage 0-II
  GLO 1high vs. GLO 1low0.99 (0.83–1.18)0.92
  PKCλhigh vs. PKCλlow1.14 (0.97–1.35)0.12
  GLO 1high PKCλhigh vs. GLO 1low PKCλlow1.03 (0.77–1.37)0.86
  GLO 1high PKCλhigh vs. GLO 1low PKCλhigh0.85 (0.62–1.17)0.32
  GLO 1high PKCλhigh vs. GLO 1high PKCλlow0.98 (0.71–1.36)0.91
Stage III–IV
  GLO 1high vs. GLO 1low0.89 (0.57–1.39)0.62
  PKCλhigh vs. PKCλlow2.23 (1.41–3.54)<0.01
  GLO 1high PKCλhigh vs. GLO 1low PKCλlow2.36 (1.08–5.16)0.03
  GLO 1high PKCλhigh vs. GLO 1low PKCλhigh1.04 (0.44–2.43)0.93
  GLO 1high PKCλhigh vs. GLO 1high PKCλlow3.25 (1.26–8.35)0.01

a Hazard ratio adjusted by age estimated using Cox proportional hazard model. GLO 1, glyoxalase 1; PKCλ, protein kinase Cλ.

TLSC702 and ATM suppresses breast cancer cell viability

Amongst patients with basal-like breast tumors, the proportion of patients classed as GLO 1high PKCλhigh was larger than the proportion classed as GLO 1low PKCλlow, irrespective of whether they had early- or late-stage tumors (Fig. S3). All patients with breast cancer classed as GLO 1high PKCλhigh primarily had luminal B type breast cancer (43.1%, 59/137), whereas those with GLO 1low PKCλlow primarily had luminal A type breast cancer (42.3%, 459/1,084) (Fig. S3). Similarly, amongst patients with early-stage lesions, those classed as GLO 1high PKCλhigh primarily had luminal B breast cancer (48.4%, 46/95), and those with GLO 1low PKCλlow primarily had luminal A breast cancer (45.2%, 333/737). However, amongst patients with late-stage lesions, those classed as GLO 1high PKCλhigh exhibited higher incidences of luminal A or basal-like type breast cancer (luminal A, 33.3%, 3/9; basal-like, 33.3%, 3/9) compared with patients classed as GLO 1low PKCλlow (luminal A, 28.6%, 18/63; basal-like, 6.3%, 4/63). As shown in Fig. S4, 38% (76/199) of patients with basal-like type cancer were classed as GLO 1high PKCλhigh. In our previous study, it was shown that GLO 1 expression is upregulated in basal-like breast cancer, and that inhibition of GLO 1 suppresses basal-like breast cancer cell viability (21). To further clarify the effects of inhibiting GLO 1 and PKCλ in basal-like breast cancer cells, MCF-10A human normal-like (non-transformed) mammary epithelial cells were compared with MDA-MB-157 and MDA-MB-468 human basal-like breast cancer cells (Fig. 3) (21). Expression levels of both GLO 1 and PKCλ were higher in MDA-MB-157 and MDA-MB-468 cells than in MCF-10A cells (Fig. 3A). TLSC702 has previously been shown to inhibit GLO 1 activity and induce MG accumulation and apoptosis in cancer cells (21,61). Based on the inhibitory effects of TLSC702 on the viability of MCF-10A, MDA-MB-157 and MDA-MB-468 cells, concentrations of 50, 75 and 100 µM TLSC702 were used in the present study (21). To inhibit PKCλ, ATM was used (5 and 10 µM), which interferes with the PB1-PB1 domain interactions between PKCλ and Par6 and induces apoptosis (62,63). As a result of the inhibitory effect of 0.5, 1, 5 and 10 µM ATM on the colony formation ability of MDA-MB-157 cells, it was found that 5 and 10 µM ATM markedly suppressed colony formation compared with the untreated control group (unpublished data). The inhibitor concentrations used in the present study were based on these findings. WST-8 assays showed that MCF-10A cells were less sensitive to GLO 1 inhibition than the two cancer cell lines, consistent with our previous study (Fig. 3B; TLSC702 50 and 75 µM) (21). By contrast, inhibition of PKCλ using ATM did not significantly affect the cell viability of any of the three cell types assessed (Fig. 3B). In addition, the combination of TLSC702 and ATM decreased the viability of the two cancer cell lines, which was reduced to a greater degree than that of the control cells (TLSC702/ATM, 50/10, 75/5, 75/10, 100/5 and 100/10 µM; Fig. 3B).

TLSC702 and ATM suppresses tumor-sphere formation in basal-like breast cancer cells

In vitro tumor-sphere formation was assessed using MDA-MB-157 and MDA-MB-468 cells to determine the roles of GLO 1 and PKCλ in the tumorigenicity of basal-like breast cancer cells. As shown in Fig. 4A and B, treatment with TLSC702 or ATM reduced the relative numbers of tumor-spheres ≥50 µm in diameter. Furthermore, a combination of TLSC702 and ATM inhibited the formation of tumor-spheres ≥50 µm in diameter (Fig. 4A and B; P<0.05). Cell Titer-Glo® assays also showed that the combination of TLSC702 and ATM suppressed the viability of MDA-MB-157 and MDA-MB-468 cells (Fig. 4C and D; P<0.05). Taken together, these results along with the results of the viability assays suggest that GLO 1 and PKCλ are cooperatively involved in cancer progression and survival of basal-like breast cancer cells.

Discussion

In the present study, it was shown that high GLO 1 expression was correlated with high PKCλ expression at the protein and mRNA levels in breast cancer and that patients with breast cancer classed as GLO 1high PKCλhigh had poorer clinical outcomes for late-stage tumors. GLO 1 and PKCλ exhibit low frequencies of gene amplification and mutations in with breast cancer (21,41). It thus appears that in breast cancer, higher GLO 1 and PKCλ mRNA expression and activation reflect higher transcriptional activity rather than gene amplification or mutations. In stage III–IV patients, the IHC value (based on tissue acquisition) is relatively low, and disease burden is approximated using imaging methods, such as CT or PET. However, the present cohort data lacks CT/PET confirmation. Therefore, GLO 1 and PKCλ may not be suitable prognostic factors in stage III–IV patients. However, late-stage breast cancer cases are often resistant or less responsive to conventional medical approaches (3). In the present study, patients with stage III–IV tumors with high GLO 1 and PKCλ expression exhibited poorer overall survival compared with patients expressing lower levels of these genes. Therefore, GLO 1 and PKCλ may potentially serve as effective therapeutic targets for late-stage human breast cancer. However, there is a lack of in vivo studies of breast cancer using TLSC702 and ATM, thus the use of both inhibitors requires further investigation in vivo.

The prognosis of patients with stage 0-II breast cancer with GLO 1high PKCλhigh did not differ significantly from those with GLO 1low PKCλlow. Notably, amongst patients with stage III–IV tumors, those with GLO 1high PKCλhigh status exhibited poorer prognoses. To support increased glycolysis, glycolytic enzymes are upregulated in cancer cells. In breast cancer, one such enzyme is GLO 1 (1921), and MG, an intermediate metabolite of glycolysis, induces GLO 1 expression (64). In the present study, GLO 1 protein expression was detected in all breast cancer samples. These results are in line with a previous report (20). Fig. S1 also showed that GLO 1 gene expression is enhanced at all tumor stages of breast cancer in comparison with normal tissues. Thus, GLO 1 mRNA and protein expression is considerably high in breast cancer, reflecting an increased level of glycolysis. Therefore, it is considered that there is no difference in prognosis between GLO 1high and GLO 1low even when classified by high and low expression in breast cancers in which GLO 1 expression was essentially high. This is in line with the results that we previously analyzed using same gene expression data set, which the prognosis of patients with GLO 1high was not associated with a poorer OS amongst breast cancer patients (21). Thus, the results of the current study suggest that both GLO 1 and PKCλ may be cooperatively involved in breast cancer progression, and contribute to poor prognosis.

Basal-like breast cancer, the majority of cases of which are TNBC, has the poorest clinical outcomes amongst all breast cancer subtypes (6). Notably, TNBC cells show higher GLO 1 expression levels, higher GLO 1 activity and lower accumulation of a MG-arginine adduct, Arg-pyrimidine (64). Notably, PKCλ is upregulated in patients with TNBC (30), and it was confirmed that the fraction of stage III–IV basal-like breast cancer cases with GLO 1high PKCλhigh were enriched compared with all other breast cancer subtypes. Thirty eight percent of patients (76/199) with basal-like type cancer were classed as GLO 1high PKCλhigh. This result suggests that GLO 1 and PKCλ are cooperatively involved in the progression of basal-like breast cancer. Moreover, the GLO 1 inhibitor TLSC702 and the PKCλ inhibitor ATM suppressed the viability of MDA-MB-157 and MDA-MB-468 basal-like breast cancer cells and tumor-sphere formation using these cells. The Par6-PKCλ complex interacts with epithelial cell transforming sequence 2 to activate Rac1 during cancer cell proliferation (65), and PKCλ activates the Rac1-Pak-Mek1/2-Erk1/2 signaling pathway in lung cancer cell growth and tumorigenicity (47). PKCλ modulates c-Myc via FoxO1 DNA-binding ability and contributes to cell growth of angiosarcoma (48). MG, which induces expression and activity of GLO 1, also induces phosphorylation of Erk1/2 (66). GLO 1 modulates cell viability and tumor formation in ALDH1-positive breast cancer stem cells (CSCs) (21). Moreover, PKCλ also modulates cell viability, Caspase 3-dependent apoptosis and tumor formation in an Akt independent manner in ALDH1-positive breast CSCs (41,42). The results of the present study suggested that GLO 1 is also functionally associated with PKCλ in the progression of ALDH1-positive breast CSCs.

PKCλ is essential for cancer cell survival of ALDH1-positive breast CSCs by maintaining low levels of ROS (41). ALDH1 serves a role in the detoxification of toxic aldehyde intermediaries generated by ROS-induced peroxidation of intracellular lipids (67). Conversely, GLO 1 detoxifies MG, a cytotoxic byproduct of glycolysis that induces apoptosis (68). Inhibition of GLO 1 reduces cell viability and induces apoptosis in ALDH1-positive breast CSCs (21). In the present study, the combination of TLSC702 and ATM suppressed viability of basal-like breast cancer cells. Therefore, GLO 1 and PKCλ may be involved in cell viability by maintaining lower intracellular ROS levels and/or detoxification of MG. However, the detailed relationship between GLO 1 and PKCλ in ALDH1-positive breast CSCs remains to be determined.

GLUT1 facilitates glucose transport, and its expression is increased in several types of cancers, including basal-like breast cancer, where it is involved in cancer progression (49,50). PKCλ is involved in GLUT1 translocation from intracellular vesicles to the plasma membrane in 3T3-L1 adipocytes (51), where it increases glucose accumulation and promotes cell growth via upregulation of GLUT1 (69). Furthermore, PKCλ is involved in insulin-dependent glucose-uptake by GLUT4 translocation to the plasma membrane in 3T3-L1 adipocytes (44,45). In addition, GAPDH is phosphorylated by PKCλ and then interacts directly with the PKCλ regulatory domain to promote microtubule nucleation. (70,71). GLO 1 is also overexpressed in basal-like breast cancer (21). Given the high levels of GLO 1 and PKCλ expression in several basal-like breast cancer types, it may be that PKCλ regulates glucose uptake by GLUT1, leading to increased glycolysis catalyzed in part by GLO 1.

Expression of PKCζ, another aPKC isotype, is also upregulated in breast cancer tissues compared with normal tissues derived from the same patient (41). c-Myc is reportedly phosphorylated by PKCζ in prostate cancer (72), and c-Myc is known to directly transactivate glucose metabolic genes, including GLUT1, PFK-1, GAPDH and enolase, and to increase glucose uptake (73). Thus, c-Myc phosphorylation by PKCζ may contribute to the regulation of glycolysis in breast cancer. In addition, PKCζ phosphorylates Nrf2, which regulates glucose-6-phosphate dehydrogenase (G6PD) gene expression (74). G6PD dehydrogenizes glucose-6-phosphate, an intermediate of glycolysis, and is the rate limiting enzyme in the pentose phosphate pathway (PPP). PPP generates glyceraldehyde 3 phosphate (GAP), an intermediate metabolite of glycolysis, and GAP is converted into MG. Furthermore, despite nutrient stress, PKCζ directly phosphorylates and inhibits the enzymatic activity of PHGDH, which suppresses metabolic reprogramming of glycolytic intermediates (75). This suggests that both PKCλ and PKCζ are associated with glycolysis, directly and indirectly, at different stages in breast cancer development/progression.

Expression levels of GLO 1 and PKCλ mRNA are correlated in ER- and/or PgR-positive and luminal B type breast cancer. Given that luminal B type breast cancer exhibits expression of ER and/or PgR, the correlation between GLO 1 and PKCλ in luminal B tumors may be related to the ER and/or PgR positivity of those tumors. The luminal B subtype is associated with poorer clinical outcomes compared with the luminal A subtype (76). These results therefore suggest that high expression of GLO 1 and PKCλ may be contributed to cancer progression in luminal B.

Kaplan-Meier and multivariable Cox regression analyses showed that normal-like breast cancer patients classified as GLO 1high PKCλhigh also exhibited a worse prognosis. Earlier studies reported that patients with normal-like tumors had a better prognosis compared with patients with other breast cancer subtypes (6). However, unlike other breast cancer subtypes, which have well-described molecular characteristics, the significant features of the normal-like subtype are largely unknown (77), and the roles of GLO 1 and PKCλ remain to be determined.

In conclusion, the levels of GLO 1 and PKCλ expression were shown to be correlated with breast cancer. Patients with late-stage tumors who were classed as GLO 1high PKCλhigh had a poorer prognosis and accounted for a large percentage of cases of basal-like breast cancer. In addition, TLSC702, a GLO 1 inhibitor, and ATM, a PKCλ inhibitor, reduced both cell viability and tumor-sphere formation in basal-like breast cancer cells. It thus appears that GLO 1 and PKCλ are cooperatively involved in cancer progression and contribute to poorer clinical outcomes in late-stage breast cancer patients. It is therefore suggested that GLO 1 and PKCλ are potentially effective therapeutic targets for treatment of late-stage breast cancer.

Supplementary Material

Supporting Data

Acknowledgements

The authors would like to thank Dr Babita Shashni (Department of Materials Science, Graduate School of Pure and Applied Sciences, University of Tsukuba, Ibaraki, Japan) for proofreading the article. The authors would also like to thank Dr Yoshiyasu Nakamura (Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Kanagawa, Japan) for technical support for immunohistochemistry.

Funding

This work was supported by a Grant-in-Aid for Scientific Research (C) of JSPS (grant no. 20K07207), a Grant-in-Aid for JSPS Research Fellows (grant no. 20J11980), JSPS KAKENHI Grant Number JP 16H06277 (CoBiA), the MEXT's Promotion Plan for the Platform of Human Resource Development for Cancer project, 2012–2017, Translational Research Center, Research Institute for Science and Technology, Tokyo University of Science (S1411013) and Nagai Memorial Research Scholarship from the Pharmaceutical Society of Japan.

Availability of data and materials

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

Authors' contributions

HM, AO, ST and YW performed the experiments. HM, AO and KA confirmed the authenticity of all the raw data. HM and AO analyzed the data using IHC Profiler. HM, AO, ST, CO, YNo, YW, YMa, TS, KeS and KYa performed the bioinformatics analysis. RT, KYo, TH, KaS, HI, YMi, YNa, SIT and SO contributed to acquisition of data, and analysis and interpretation of data. HM and KA conceived the study. HM drafted the manuscript. HM, AO, KaS, HI, SO and KA contributed to discussion and review of the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The experimental protocol of the present study was reviewed and approved by the Institutional Review Board of Kanagawa Cancer Center Hospital (approval no. 3-2009; Kanagawa, Japan). Written informed consent for participation was obtained from all patients.

Patient consent for publication

Written informed consent was obtained from all patients for publication.

Competing interests

The authors declare that they have no competing interests.

References

1 

World Cancer Research Fund (WCRF), . Breast cancer statistics. Breast cancer is the most common cancer in women worldwide. WCRF, London. 2018.https://www.wcrf.org/dietandcancer/cancer-trends/breast-cancer-statistics

2 

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

3 

Maughan KL, Lutterbie MA and Ham PS: Treatment of breast cancer. Am Fam Physician. 81:1339–1346. 2010.PubMed/NCBI

4 

van de Vijver MJ, He YD, van't Veer LJ, Dai H, Hart AA, Voskuil DW, Schreiber GJ, Peterse JL, Roberts C, Marton MJ, et al: A gene-expression signature as a predictor of survival in breast cancer. N Engl J Med. 347:1999–2009. 2002. View Article : Google Scholar : PubMed/NCBI

5 

Perou CM, Sørlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, Pollack JR, Ross DT, Johnsen H, Akslen LA, et al: Molecular portraits of human breast tumours. Nature. 406:747–752. 2000. View Article : Google Scholar : PubMed/NCBI

6 

Sørlie T, Perou CM, Tibshirani R, Aas T, Geisler S, Johnsen H, Hastie T, Eisen MB, van de Rijn M, Jeffrey SS, et al: Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci USA. 98:10869–10874. 2001. View Article : Google Scholar

7 

Herschkowitz JI, Simin K, Weigman VJ, Mikaelian I, Usary J, Hu Z, Rasmussen KE, Jones LP, Assefnia S, Chandrasekharan S, et al: Identification of conserved gene expression features between murine mammary carcinoma models and human breast tumors. Genome Biol. 8:R762007. View Article : Google Scholar : PubMed/NCBI

8 

Badve S, Dabbs DJ, Schnitt SJ, Baehner FL, Decker T, Eusebi V, Fox SB, Ichihara S, Jacquemier J, Lakhani SR, et al: Basal-like and triple-negative breast cancers: A critical review with an emphasis on the implications for pathologists and oncologists. Mod Pathol. 24:157–167. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Warburg O, Wind F and Negelein E: The metabolism of tumors in the body. J Gen Physiol. 8:519–530. 1927. View Article : Google Scholar : PubMed/NCBI

10 

Gatenby RA and Gillies RJ: Why do cancers have high aerobic glycolysis? Nat Rev Cancer. 4:891–899. 2004. View Article : Google Scholar : PubMed/NCBI

11 

Sakamoto H, Mashima T, Sato S, Hashimoto Y, Yamori T and Tsuruo T: Selective activation of apoptosis program by S-p-bromobenzylglutathione cyclopentyl diester in glyoxalase I-overexpressing human lung cancer cells. Clin Cancer Res. 7:2513–2518. 2001.PubMed/NCBI

12 

Cheng WL, Tsai MM, Tsai CY, Huang YH, Chen CY, Chi HC, Tseng YH, Chao IW, Lin WC, Wu SM, et al: Glyoxalase-I is a novel prognosis factor associated with gastric cancer progression. PLoS One. 7:e343522012. View Article : Google Scholar : PubMed/NCBI

13 

Ranganathan S and Tew KD: Analysis of glyoxalase-I from normal and tumor tissue from human colon. Biochim Biophys Acta. 1182:311–316. 1993. View Article : Google Scholar : PubMed/NCBI

14 

Zhang S, Liang X, Zheng X, Huang H, Chen X, Wu K, Wang B and Ma S: Glo1 genetic amplification as a potential therapeutic target in hepatocellular carcinoma. Int J Clin Exp Pathol. 7:2079–2090. 2014.PubMed/NCBI

15 

Baunacke M, Horn LC, Trettner S, Engel KM, Hemdan NY, Wiechmann V, Stolzenburg JU, Bigl M and Birkenmeier G: Exploring glyoxalase 1 expression in prostate cancer tissues: Targeting the enzyme by ethyl pyruvate defangs some malignancy-associated properties. Prostate. 74:48–60. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Kreycy N, Gotzian C, Fleming T, Flechtenmacher C, Grabe N, Plinkert P, Hess J and Zaoui K: Glyoxalase 1 expression is associated with an unfavorable prognosis of oropharyngeal squamous cell carcinoma. BMC Cancer. 17:3822017. View Article : Google Scholar : PubMed/NCBI

17 

Zou XY, Ding D, Zhan N, Liu XM, Pan C and Xia YM: Glyoxalase I is differentially expressed in cutaneous neoplasms and contributes to the progression of squamous cell carcinoma. J Invest Dermatol. 135:589–598. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Bair WB III, Cabello CM, Uchida K, Bause AS and Wondrak GT: GLO1 overexpression in human malignant melanoma. Melanoma Res. 20:85–96. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Rulli A, Carli L, Romani R, Baroni T, Giovannini E, Rosi G and Talesa V: Expression of glyoxalase I and II in normal and breast cancer tissues. Breast Cancer Res Treat. 66:67–72. 2001. View Article : Google Scholar : PubMed/NCBI

20 

Peng HT, Chen J, Liu TY, Wu YQ, Lin XH, Lai YH and Huang YF: Up-regulation of the tumor promoter Glyoxalase-1 indicates poor prognosis in breast cancer. Int J Clin Exp Pathol. 10:10852–10862. 2017.PubMed/NCBI

21 

Tamori S, Nozaki Y, Motomura H, Nakane H, Katayama R, Onaga C, Kikuchi E, Shimada N, Suzuki Y, Noike M, et al: Glyoxalase 1 gene is highly expressed in basal-like human breast cancers and contributes to survival of ALDH1-positive breast cancer stem cells. Oncotarget. 9:36515–36529. 2018. View Article : Google Scholar : PubMed/NCBI

22 

Zhang D, Tai LK, Wong LL, Chiu LL, Sethi SK and Koay ES: Proteomic study reveals that proteins involved in metabolic and detoxification pathways are highly expressed in HER-2/neu-positive breast cancer. Mol Cell Proteomics. 4:1686–1696. 2005. View Article : Google Scholar : PubMed/NCBI

23 

Gandalovičová A, Vomastek T, Rosel D and Brábek J: Cell polarity signaling in the plasticity of cancer cell invasiveness. Oncotarget. 7:25022–25049. 2016. View Article : Google Scholar

24 

Akimoto K, Mizuno K, Osada S, Hirai S, Tanuma S, Suzuki K and Ohno S: A new member of the third class in the protein kinase C family, PKC lambda, expressed dominantly in an undifferentiated mouse embryonal carcinoma cell line and also in many tissues and cells. J Biol Chem. 269:12677–12683. 1994. View Article : Google Scholar : PubMed/NCBI

25 

Suzuki A, Akimoto K and Ohno S: Protein kinase C lambda/iota (PKClambda/iota): A PKC isotype essential for the development of multicellular organisms. J Biochem. 133:9–16. 2003. View Article : Google Scholar : PubMed/NCBI

26 

Ohno S: Intercellular junctions and cellular polarity: The PAR-aPKC complex, a conserved core cassette playing fundamental roles in cell polarity. Curr Opin Cell Biol. 13:641–648. 2001. View Article : Google Scholar : PubMed/NCBI

27 

Parker PJ, Justilien V, Riou P, Linch M and Fields AP: Atypical protein kinase Cι as a human oncogene and therapeutic target. Biochem Pharmacol. 88:1–11. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Murray NR, Jamieson L, Yu W, Zhang J, Gökmen-Polar Y, Sier D, Anastasiadis P, Gatalica Z, Thompson EA and Fields AP: Protein kinase Ciota is required for Ras transformation and colon carcinogenesis in vivo. J Cell Biol. 164:797–802. 2004. View Article : Google Scholar : PubMed/NCBI

29 

Kojima Y, Akimoto K, Nagashima Y, Ishiguro H, Shirai S, Chishima T, Ichikawa Y, Ishikawa T, Sasaki T, Kubota Y, et al: The overexpression and altered localization of the atypical protein kinase C lambda/iota in breast cancer correlates with the pathologic type of these tumors. Hum Pathol. 39:824–831. 2008. View Article : Google Scholar : PubMed/NCBI

30 

Paul A, Gunewardena S, Stecklein SR, Saha B, Parelkar N, Danley M, Rajendran G, Home P, Ray S, Jokar I, et al: PKCλ/ι signaling promotes triple-negative breast cancer growth and metastasis. Cell Death Differ. 21:1469–1481. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Mizushima T, Asai-Sato M, Akimoto K, Nagashima Y, Taguri M, Sasaki K, Nakaya MA, Asano R, Tokinaga A, Kiyono T, et al: Aberrant expression of the cell polarity regulator aPKCλ/ι is associated with disease progression in cervical intraepithelial Neoplasia (CIN): A possible marker for predicting CIN prognosis. Int J Gynecol Pathol. 35:106–117. 2016. View Article : Google Scholar : PubMed/NCBI

32 

Tokinaga-Uchiyama A, Mizushima T, Akimoto K, Nagashima Y, Sasaki K, Nakaya MA, Ohashi K, Kubota K, Maruyama Y, Kato H, et al: Aberrant nuclear localization of aPKCλ/ι is associated with poorer prognosis in uterine cervical cancer. Gynecol Pathol. 38:301–309. 2019. View Article : Google Scholar : PubMed/NCBI

33 

Baba J, Kioi M, Akimoto K, Nagashima Y, Taguri M, Inayama Y, Aoki I, Ohno S, Mitsudo K and Tohnai I: Atypical protein kinase Cλ/ι expression is associated with malignancy of oral squamous cell carcinoma. Anticancer Res. 38:6291–6297. 2018. View Article : Google Scholar : PubMed/NCBI

34 

Ishiguro H, Akimoto K, Nagashima Y, Kagawa E, Sasaki T, Sano JY, Takagawa R, Fujinami K, Sasaki K, Aoki I, et al: Coexpression of aPKCλ/ι and IL-6 in prostate cancer tissue correlates with biochemical recurrence. Cancer Sci. 102:1576–1581. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Ishiguro H, Akimoto K, Nagashima Y, Kojima Y, Sasaki T, Ishiguro-Imagawa Y, Nakaigawa N, Ohno S, Kubota Y and Uemura H: aPKClambda/iota promotes growth of prostate cancer cells in an autocrine manner through transcriptional activation of interleukin-6. Proc Natl Acad Sci USA. 106:16369–16374. 2009. View Article : Google Scholar : PubMed/NCBI

36 

Regala RP, Weems C, Jamieson L, Khoor A, Edell ES, Lohse CM and Fields AP: Atypical protein kinase C iota is an oncogene in human non-small cell lung cancer. Cancer Res. 65:8905–8911. 2005. View Article : Google Scholar : PubMed/NCBI

37 

Eder AM, Sui X, Rosen DG, Nolden LK, Cheng KW, Lahad JP, Kango-Singh M, Lu KH, Warneke CL, Atkinson EN, et al: Atypical PKCiota contributes to poor prognosis through loss of apical-basal polarity and cyclin E overexpression in ovarian cancer. Proc Natl Acad Sci USA. 102:12519–12524. 2005. View Article : Google Scholar : PubMed/NCBI

38 

Scotti ML, Bamlet WR, Smyrk TC, Fields AP and Murray NR: Protein kinase Ciota is required for pancreatic cancer cell transformed growth and tumorigenesis. Cancer Res. 70:2064–2074. 2010. View Article : Google Scholar : PubMed/NCBI

39 

Kato S, Akimoto K, Nagashima Y, Ishiguro H, Kubota K, Kobayashi N, Hosono K, Watanabe S, Sekino Y, Sato T, et al: aPKCλ/ι is a beneficial prognostic marker for pancreatic neoplasms. Pancreatology. 13:360–368. 2013. View Article : Google Scholar : PubMed/NCBI

40 

Takagawa R, Akimoto K, Ichikawa Y, Akiyama H, Kojima Y, Ishiguro H, Inayama Y, Aoki I, Kunisaki C, Endo I, et al: High expression of atypical protein kinase C lambda/iota in gastric cancer as a prognostic factor for recurrence. Ann Surg Oncol. 17:81–88. 2010. View Article : Google Scholar : PubMed/NCBI

41 

Nozaki Y, Motomura H, Tamori S, Kimura Y, Onaga C, Kanai S, Ishihara Y, Ozaki A, Hara Y, Harada Y, et al: High PKCλ expression is required for ALDH1-positive cancer stem cell function and indicates a poor clinical outcome in late-stage breast cancer patients. PLoS One. 15:e02357472020. View Article : Google Scholar : PubMed/NCBI

42 

Motomura H, Nozaki Y, Onaga C, Ozaki A, Tamori S, Shiina TA, Kanai S, Ohira C, Hara Y, Harada Y, et al: High expression of c-Met, PKCλ and ALDH1A3 predicts a poor prognosis in late-stage breast cancer. Anticancer Res. 40:35–52. 2020. View Article : Google Scholar : PubMed/NCBI

43 

Akimoto K, Takahashi R, Moriya S, Nishioka N, Takayanagi J, Kimura K, Fukui Y, Osada SI, Mizuno K, Hirai SI, et al: EGF or PDGF receptors activate atypical PKClambda through phosphatidylinositol 3-kinase. EMBO J. 15:788–798. 1996. View Article : Google Scholar : PubMed/NCBI

44 

Kotani K, Ogawa W, Matsumoto M, Kitamura T, Sakaue H, Hino Y, Miyake K, Sano W, Akimoto K, Ohno S and Kasuga M: Requirement of atypical protein kinase clambda for insulin stimulation of glucose uptake but not for Akt activation in 3T3-L1 adipocytes. Mol Cell Biol. 18:6971–6982. 1998. View Article : Google Scholar : PubMed/NCBI

45 

Farese RV: Function and dysfunction of aPKC isoforms for glucose transport in insulin-sensitive and insulin-resistant states. Am J Physiol Endocrinol Metab. 283:E1–E11. 2002. View Article : Google Scholar : PubMed/NCBI

46 

Akimoto K, Nakaya M, Yamanaka T, Tanaka J, Matsuda S, Weng QP, Avruch J and Ohno S: Atypical protein kinase Clambda binds and regulates p70 S6 kinase. Biochem J. 335:417–424. 1998. View Article : Google Scholar : PubMed/NCBI

47 

Regala RP, Weems C, Jamieson L, Copland JA, Thompson EA and Fields AP: Atypical protein kinase Ciota plays a critical role in human lung cancer cell growth and tumorigenicity. J Biol Chem. 280:31109–31115. 2005. View Article : Google Scholar : PubMed/NCBI

48 

Riddell M, Nakayama A, Hikita T, Mirzapourshafiyi F, Kawamura T and Pasha A: aPKC controls endothelial growth by modulating c-Myc via FoxO1 DNA-binding ability. Nat Commun. 9:53572018. View Article : Google Scholar : PubMed/NCBI

49 

Szablewski L: Expression of glucose transporters in cancers. Biochim Biophys Acta. 1835:164–169. 2013.PubMed/NCBI

50 

Hussein YR, Bandyopadhyay S, Semaan A, Ahmed Q, Albashiti B, Jazaerly T, Nahleh Z and Ali-Fehmi R: Glut-1 expression correlates with basal-like breast cancer. Transl Oncol. 4:321–327. 2011. View Article : Google Scholar : PubMed/NCBI

51 

Bosch RR, Bazuine M, Span PN, Willems PH, Olthaar AJ, van Rennes H, Maassen JA, Tack CJ, Hermus AR and Sweep CG: Regulation of GLUT1-mediated glucose uptake by PKClambda-PKCbeta(II) interactions in 3T3-L1 adipocytes. Biochem J. 384:349–355. 2004. View Article : Google Scholar : PubMed/NCBI

52 

Varghese F, Bukhari AB, Malhotra R and De A: IHC Profiler: An open source plugin for the quantitative evaluation and automated scoring of immunohistochemistry images of human tissue samples. PLoS One. 9:e968012014. View Article : Google Scholar : PubMed/NCBI

53 

Nozaki Y, Tamori S, Inada M, Katayama R, Nakane H, Minamishima O, Onodera Y, Abe M, Shiina S, Tamura K, et al: Correlation between c-Met and ALDH1 contributes to the survival and tumor-sphere formation of ALDH1 positive breast cancer stem cells and predicts poor clinical outcome in breast cancer. Genes Cancer. 8:628–639. 2017. View Article : Google Scholar : PubMed/NCBI

54 

Sato K and Akimoto K: Expression levels of KMT2C and SLC20A1 identified by information-theoretical analysis are powerful prognostic biomarkers in estrogen receptor-positive breast cancer. Clin Breast Cancer. 17:e135–e142. 2017. View Article : Google Scholar : PubMed/NCBI

55 

Curtis C, Shah SP, Chin SF, Turashvili G, Rueda OM, Dunning MJ, Speed D, Lynch AG, Samarajiwa S, Yuan Y, et al: The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature. 486:346–352. 2012. View Article : Google Scholar : PubMed/NCBI

56 

Pereira B, Chin SF, Rueda OM, Vollan HK, Provenzano E, Bardwell HA, Pugh M, Jones L, Russell R, Sammut SJ, et al: The somatic mutation profiles of 2,433 breast cancers refine their genomic and transcriptomic landscapes. Nat Commun. 7:114792016. View Article : Google Scholar : PubMed/NCBI

57 

Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, Jacobsen A, Byrne CJ, Heuer ML, Larsson E, et al: The cBio cancer genomics portal: An open platform for exploring multidimensional cancer genomics data. Cancer Dicov. 2:401–404. 2012. View Article : Google Scholar : PubMed/NCBI

58 

Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, Sun Y, Jacobsen A, Sinha R, Larsson E, et al: Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 6:pl12013. View Article : Google Scholar : PubMed/NCBI

59 

Cancer Genome Atlas Network, . Comprehensive molecular portraits of human breast tumours. Nature. 490:61–70. 2012. View Article : Google Scholar : PubMed/NCBI

60 

Rhodes DR, Yu J, Shanker K, Deshpande N, Varambally R, Ghosh D, Barrette T, Pandey A and Chinnaiyan AM: ONCOMINE: A cancer microarray database and integrated data-mining platform. Neoplasia. 6:1–6. 2004. View Article : Google Scholar : PubMed/NCBI

61 

Takasawa R, Tao A, Saeki K, Shionozaki N, Tanaka R, Uchiro H, Takahashi S, Yoshimori A and Tanuma S: Discovery of a new type inhibitor of human glyoxalase I by myricetin-based 4-point pharmacophore. Bioorg. Med Chem Lett. 21:4337–4342. 2011. View Article : Google Scholar : PubMed/NCBI

62 

Stallings-Mann M, Jamieson L, Regala RP, Weems C, Murray NR and Fields AP: A novel small-molecule inhibitor of protein kinase Ciota blocks transformed growth of non-small-cell lung cancer cells. Cancer Res. 66:1767–1774. 2006. View Article : Google Scholar : PubMed/NCBI

63 

Trani M, Sorrentino A, Busch C and Landström M: Pro-apoptotic effect of aurothiomalate in prostate cancer cells. Cell Cycle. 8:306–313. 2009. View Article : Google Scholar : PubMed/NCBI

64 

Chiavarina B, Nokin MJ, Durieux F, Bianchi E, Turtoi A, Peulen O, Peixoto P, Irigaray P, Uchida K, Belpomme D, et al: Triple negative tumors accumulate significantly less methylglyoxal specific adducts than other human breast cancer subtypes. Oncotarget. 5:5472–5482. 2014. View Article : Google Scholar : PubMed/NCBI

65 

Justilien V and Fields AP: Ect2 links the PKCiota-Par6alpha complex to Rac1 activation and cellular transformation. Oncogene. 28:3597–3607. 2009. View Article : Google Scholar : PubMed/NCBI

66 

Guo Y, Zhang Y, Yang X, Lu P, Yan X, Xiao F, Zhou H, Wen C, Shi M, Lu J and Meng QH: Effects of methylglyoxal and glyoxalase I inhibition on breast cancer cells proliferation, invasion, and apoptosis through modulation of MAPKs, MMP9, and Bcl-2. Cancer Biol Ther. 17:169–180. 2016. View Article : Google Scholar : PubMed/NCBI

67 

Singh S, Brocker C, Koppaka V, Chen Y, Jackson BC and Matsumoto A: Aldehyde dehydrogenases in cellular responses to oxidative/electrophilic stress. Free Radic Biol Med. 56:89–101. 2013. View Article : Google Scholar : PubMed/NCBI

68 

Kang Y, Edwards LG and Thornalley PJ: Effect of methylglyoxal on human leukaemia 60 cell growth: Modification of DNA G1 growth arrest and induction of apoptosis. Leuk Res. 20:97–405. 1996. View Article : Google Scholar

69 

Liu L, Lei B, Wang L, Chang C, Yang H, Liu J, Huang G and Xie W: Protein kinase C-iota-mediated glycolysis promotes non-small-cell lung cancer progression. Onco Targets Ther. 12:5835–5848. 2019. View Article : Google Scholar : PubMed/NCBI

70 

Tisdale EJ: Glyceraldehyde-3-phosphate dehydrogenase is phosphorylated by protein kinase Ciota/lambda and plays a role in microtubule dynamics in the early secretory pathway. J Biol Chem. 277:3334–3341. 2002. View Article : Google Scholar : PubMed/NCBI

71 

Tisdale EJ: Rab2 interacts directly with atypical protein kinase C (aPKC) iota/lambda and inhibits aPKCiota/lambda-dependent glyceraldehyde-3-phosphate dehydrogenase phosphorylation. J Biol Chem. 278:52524–52530. 2003. View Article : Google Scholar : PubMed/NCBI

72 

Kim JY, Valencia T, Abu-Baker S, Linares J, Lee SJ, Yajima T, Chen J, Eroshkin A, Castilla EA, Brill LM, et al: c-Myc phosphorylation by PKCζ represses prostate tumorigenesis. Proc Natl Acad Sci USA. 110:6418–6423. 2013. View Article : Google Scholar : PubMed/NCBI

73 

Osthus RC, Shim H, Kim S, Li Q, Reddy R, Mukherjee M, Xu Y, Wonsey D, Lee LA and Dang CV: Deregulation of glucose transporter 1 and glycolytic gene expression by c-Myc. J Biol Chem. 275:21797–21800. 2000. View Article : Google Scholar : PubMed/NCBI

74 

Gjyshi O, Bottero V, Veettil MV, Dutta S, Singh VV and Chikoti L: Kaposi's sarcoma-associated herpesvirus induces Nrf2 during de novo infection of endothelial cells to create a microenvironment conducive to infection. PLoS Pathog. 10:e10044602014. View Article : Google Scholar : PubMed/NCBI

75 

Ma L, Tao Y, Duran A, Llado V, Galvez A, Barger JF, Castilla EA, Chen J, Yajima T, Porollo A, et al: Control of nutrient stress-induced metabolic reprogramming by PKCζ in tumorigenesis. Cell. 152:599–611. 2013. View Article : Google Scholar : PubMed/NCBI

76 

Franchet C, Duprez-Paumier R and Lacroix-Triki M: Molecular taxonomy of luminal breast cancer in 2015. Bull Cancer. 102 (6 Suppl 1):S34–S46. 2015.(In French). View Article : Google Scholar : PubMed/NCBI

77 

Liu Z, Zhang XS and Zhang S: Breast tumor subgroups reveal diverse clinical prognostic power. Sci Rep. 4:40022014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2021
Volume 22 Issue 1

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Motomura H, Ozaki A, Tamori S, Onaga C, Nozaki Y, Waki Y, Takasawa R, Yoshizawa K, Mano Y, Sato T, Sato T, et al: Glyoxalase 1 and protein kinase Cλ as potential therapeutic targets for late‑stage breast cancer. Oncol Lett 22: 547, 2021
APA
Motomura, H., Ozaki, A., Tamori, S., Onaga, C., Nozaki, Y., Waki, Y. ... Akimoto, K. (2021). Glyoxalase 1 and protein kinase Cλ as potential therapeutic targets for late‑stage breast cancer. Oncology Letters, 22, 547. https://doi.org/10.3892/ol.2021.12808
MLA
Motomura, H., Ozaki, A., Tamori, S., Onaga, C., Nozaki, Y., Waki, Y., Takasawa, R., Yoshizawa, K., Mano, Y., Sato, T., Sasaki, K., Ishiguro, H., Miyagi, Y., Nagashima, Y., Yamamoto, K., Sato, K., Hanawa, T., Tanuma, S., Ohno, S., Akimoto, K."Glyoxalase 1 and protein kinase Cλ as potential therapeutic targets for late‑stage breast cancer". Oncology Letters 22.1 (2021): 547.
Chicago
Motomura, H., Ozaki, A., Tamori, S., Onaga, C., Nozaki, Y., Waki, Y., Takasawa, R., Yoshizawa, K., Mano, Y., Sato, T., Sasaki, K., Ishiguro, H., Miyagi, Y., Nagashima, Y., Yamamoto, K., Sato, K., Hanawa, T., Tanuma, S., Ohno, S., Akimoto, K."Glyoxalase 1 and protein kinase Cλ as potential therapeutic targets for late‑stage breast cancer". Oncology Letters 22, no. 1 (2021): 547. https://doi.org/10.3892/ol.2021.12808