Open Access

Targeting transient receptor potential canonical 1 reduces non‑small cell lung cancer chemoresistance and stemness via inhibition of PI3K/AKT signaling

  • Authors:
    • Jiahui Jin
    • Xinyu Yan
    • Yaru Zhao
    • Haojie Zhang
    • Kai Zhuang
    • Yating Wen
    • Jingjing He
    • Junzhen Gao
  • View Affiliations

  • Published online on: April 13, 2023     https://doi.org/10.3892/ol.2023.13810
  • Article Number: 224
  • Copyright: © Jin 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

TRPC1 enhances cell proliferation and migration in non‑small cell lung cancer (NSCLC); however, its effect on NSCLC chemoresistance and stemness remains to be determined. The aim of the current study was to investigate the effect of TRPC1 on NSCLC chemoresistance and stemness and to determine the underlying mechanism of action. Cisplatin‑resistant A549 (A549/CDDP) and H460 (H460/CDDP) cells were first established and were then transfected with negative control small interfering (si)RNA (si‑NC) or TRPC1 siRNA (si‑TRPC1). Cells were then treated with 740 Y‑P, a PI3K/Akt agonist. Subsequently, the sensitivity of A549/CDDP and H460/CDDP cells to CDDP was evaluated. Furthermore, the expression levels of CD133 and CD44, and sphere formation ability were also determined. The results showed that the half‑maximal inhibitory concentration (IC50) of CDDP was significantly higher in A549/CDDP cells compared with A549 cells and in H460/CDDP cells compared with H460 cells. TRPC1 silencing decreased the IC50 value of CDDP compared with the si‑NC group in A549/CDDP (11.78 vs. 21.58 µM; P<0.01) and H460/CDDP (23.76 vs. 43.11 µM; P<0.05) cells. Additionally, TRPC1 knockdown in both cell lines decreased the number of spheres formed compared with the si‑NC group. Furthermore, compared with the si‑NC group, A549/CDDP cells transfected with si‑TRPC1 exhibited decreased levels of both CD133 (P<0.01) and CD44 (P<0.05). However, only CD133 (P<0.05) was downregulated in TRPC1‑depleted H460/CDDP cells compared with the si‑NC group. In addition, TRPC1 knockdown repressed PI3K/AKT signaling compared with the si‑NC group in both A549/CDDP and H460/CDDP cells (all P<0.05). Finally, cell treatment with 740 Y‑P reversed the effect of TRPC1 knockdown on PI3K/AKT signaling, chemoresistance, and cancer stemness in A549/CDDP and H460/CDDP cells (all P<0.05). In conclusion, the results of the current study suggested that targeting TRPC1 could attenuate cancer stemness and chemoresistance via suppression of PI3K/AKT signaling in NSCLC.

Introduction

Lung cancer is one of the most fatal and common types of cancer globally, accounting for ~11.4% of new cancer cases and 18.0% of cancer-related deaths in 2020. Notably, non-small cell lung cancer (NSCLC) accounts for the majority of lung cancer cases (13). To date, chemotherapy remains the most common therapeutic approach for patients with NSCLC. However, a proportion of patients with NSCLC develop chemoresistance during treatment, resulting in a less favorable survival profile (47). Cancer stemness is considered the most crucial factor contributing to chemoresistance (8). Therefore, exploring the mechanism underlying chemoresistance and stemness in NSCLC is of great importance.

Transient receptor potential canonical 1 (TRPC1) mediates the influx of extracellular Ca2+ and plays a critical role in cell proliferation, differentiation, apoptosis, and migration (9,10). The regulatory role of TRPC1 in chemoresistance in solid carcinomas has been well established (1113). A previous study showed that TRPC1 could promote hypoxia-associated epithelial-mesenchymal transition (EMT), subsequently contributing to chemoresistance in endometrial carcinoma (11,13). Another study demonstrated that TRPC1 attenuated the sensitivity of breast cancer cells to chemotherapy (12). However, the role of TRPC1 in NSCLC chemoresistance to cisplatin (CDDP, one of the most widely used and effective compounds in cancer treatment, which functions by binding to the DNA bases in the nucleus and inhibits DNA replication and transcription to exhibit its anti-tumor efficacy) and stemness remains unclear. Therefore, the current study aimed to evaluate the regulatory effect of TRPC1 on NSCLC chemoresistance and stemness, as well as its underlying mechanism of action.

Materials and methods

Cell culture

A549 and H460 cells were purchased from BeNa Culture Collection. A549 and H460 cells resistant to cis-Diamminedichloroplatinum (cisplatin/CDDP, MilliporeSigma; A549/CDPP and H460/CDDP respectively) were established by gradually exposing parental cells to increasing concentrations of CDDP as described previously (14). All cells were maintained in RPMI-1640 medium (Lonza Pharma & Biotech) supplemented with 10% FBS (Lonza Pharma & Biotech) and 1% penicillin/streptomycin solution (Beijing Solarbio Science & Technology Co., Ltd.) at 37°C in a humidified incubator supplied with 5% CO2. To maintain resistance, A549/CDDP and H460/CDDP cells were cultured in the presence of 2 and 4 µM CDDP, respectively. The mRNA and protein expression levels of TRPC1 in A549, A549/CDDP, H460, and H460/CDDP cells were detected by reverse transcription-quantitative PCR (RT-qPCR) and western blot analysis, respectively.

CDDP sensitivity assay

The sensitivity of A549, A549/CDDP, H460, and H460/CDDP cells to CDDP was assessed using Cell Counting Kit-8 assays (CCK-8; MedChemExpress). Briefly, cells were seeded into 96-well plates at a density of 2×103 cells/well. Subsequently, A549 and H460 cells were treated with 0, 0.25, 0.5, 1, 2, 4 or 8 µM CDDP for 48 h. Additionally, A549/CDDP and H460/CDDP cells were treated with 0, 2, 4, 8, 16, 32, or 64 µM CDDP for 48 h. Following treatment, cells were incubated for 2 h in the presence of CCK-8 reagent. The absorbance values were subsequently detected at a wavelength of 450 nm using a plate reader (Molecular Devices, LLC). The half-maximal inhibitory concentration (IC50) of CDDP was evaluated using a sigmoidal dose-response curve, as previously described (15).

Cell transfection

The small interfering RNA (siRNA) constructs targeting TRPC1 (si-TRPC1-1, si-TRPC1-2, and si-TRPC1-3) and the corresponding negative control (si-NC) were obtained from Shanghai GenePharma Co., Ltd. A549/CDDP and H460/CDDP cells were plated into 6-well plates at a density of 2×106 cells/well and then transfected with the above siRNAs using Lipofectamine 3000 transfection reagent (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's instructions. The knockdown efficiency of siRNAs was detected by RT-qPCR and western blot analysis. Since si-TRPC1-2 exhibited the most potent knockdown effect on TRPC1 expression, this siRNA clone was used for the subsequent interference experiments. Further assays, including CDDP sensitivity, sphere formation assay, and western blot analysis were performed after transfection. The sequences of the siRNAs used were as follows: For siRNA-1 sense, CGAUCAUCAAGACCAACUAUA and antisense, UAGUUGGUCUUGAUGAUCGUU; siRNA-2 sense, GGAAGUCUCUUUAAUGCAAUG and antisense, UUGCAUUAAAGAGACUUCCUA); siRNA-3 sense, GCUUUCAGUUGAUAGCAAAUC and antisense, UUUGCUAUCAACUGAAAGCUU); and si-NC sense, UUCUCCGAACGUGUCACGUTT and antisense, ACGUGACACGUUCGGAGAATT.

Cell treatment with 740 Y-P

A549/CDDP and H460/CDDP cells were seeded into 6-well plates and transfected as described above. Subsequently, to evaluate the regulatory effect of TRPC1 on PI3K/AKT signaling, cells were treated with 25 µg/ml 740 Y-P (a PI3K/AKT activator; MedChemExpress) as described previously (16). Following cell treatment with 740 Y-P for 48 h, western blot analysis was performed. Additionally, CDDP sensitivity and sphere formation assays were also performed in cells treated with 740 Y-P for 48 h.

RNA isolation and RT-qPCR

Total RNA was isolated from A549, A549/CDDP, H460, and H460/CDDP cells using Beyozol (Beyotime Institute of Biotechnology) and subsequently reverse transcribed into cDNA using the QuantiNova RT Kit according to the manufacturer's protocol (Qiagen GmbH). qPCR was performed using the SYBR® Premix DimmerEraser kit (Takara Bio, Inc.). The thermocycling conditions were: 95°C for 30 sec; followed by 40 cycles of 95°C for 5 sec and 61°C for 30 sec. The expression of TRPC1 was assessed using the 2−ΔΔCq method and GAPDH was used as the internal control (17). The sequences of the primers were: TRPC1 forward, 5′-ACCTTCCATTCGTTCATTGG-3′ and reverse, 5′-TGGTGAGGGAATGATGTTGA-3′; and GAPDH forward, 5′-GAGTCCACTGGCGTCTTCAC-3′ and reverse, 5′-ATCTTGAGGCTGTTGTCATACTTCT-3′.

Western blot analysis

Total protein was extracted from cells using RIPA lysis reagent supplemented with 1% PMSF (both from Wuhan Servicebio Technology Co., Ltd.). The protein concentration was measured using a BCA kit according to the manufacturer's protocol (Beyotime Institute of Biotechnology). Subsequently, 40 µg total protein was loaded per a lane on a BeyoGel™ Plus Precast PAGE Gel (Beyotime Institute of Biotechnology), resolved using SDS-PAGE, and then transferred to a nitrocellulose membrane (Wuhan Servicebio Technology Co., Ltd.). Following blocking with 5% nonfat milk (Beyotime Institute of Biotechnology), the membranes were incubated with primary and secondary antibodies for 1 h at 37°C, successively. Signals were visualized using the ECL-PLUS reagent (Beyotime Institute of Biotechnology). The following antibodies were used: Anti-TRPC1 (1:500; cat. no. DF12783; Affinity Biosciences), anti-AKT (dilution, 1:500; cat. no. AF6261; Affbiotech), anti-PI3K (1:500; cat. no. AF6241, Affbiotech), anti-CD133 (1:1,000; cat. no. 51917; Cell Signaling Technology, Inc.), anti-CD44 (1:1,000; cat. no. 37259; Cell Signaling Technology, Inc.), anti-phospho (p)-PI3K (1:2,000; cat. no. ab182651; Abcam), anti-p-AKT (1:1,000; cat. no. ab38449; Abcam), anti-GAPDH (1:5,000; cat. no. GB15004; Wuhan Servicebio Technology Co., Ltd.), and goat anti-rabbit secondary antibody (1:10,000; cat. no. GB23303; Wuhan Servicebio Technology Co., Ltd.).

Sphere formation assay

Sphere formation assays in A549/CDDP and H460/CDDP cells were performed 48 h after transfection. Briefly, cells at a density of 1×103 cells/well were seeded into 6-well ultra-low attachment plates (Corning, Inc.) and were then cultured in spheroid medium, composed of DMEM/Nutrient Mixture F-12 (DMEM-F12; Gibco; Thermo Fisher Scientific, Inc.) containing B-27 Supplement (Thermo Fisher Scientific, Inc.), 20 ng/ml epidermal growth factor (EGF, MedChemExpress), 20 ng/ml basic fibroblast growth factor (MedChemExpress), and 1% penicillin/streptomycin solution (Beijing Solarbio Science & Technology Co., Ltd.). Following 10 days of culture, the number of formed spheres (diameter, >50 µm) was counted, and images were captured under a light microscope with a magnification of ×200.

Statistical analysis

All data were compared using an unpaired Student's t-test or a one-way ANOVA followed by a Tukey's post hoc test in GraphPad Prism 7 (GraphPad Software, Inc.). P<0.05 was considered to indicate a statistically significant difference.

Results

TRPC1 is upregulated in chemoresistant NSCLC cells

The IC50 values of CDDP were 1.96 (Fig. 1A), 20.68 (Fig. 1B), 4.55 (Fig. 1C), and 43.40 µM (Fig. 1D) in parental A549 cells, A549/CDDP cells, parental H460 cells, and H460/CDDP cells, respectively, thus confirming that CDDP-resistant NSCLC cells were successfully established. Subsequently, TRPC1 expression was detected in the above cells and the results showed that the mRNA and protein expression levels of TRPC1 were notably increased in A549/CDDP cells compared with the parental A549 cells. Consistently, the same trend was observed in H460/CDDP cells compared with the parental H460 cells (all P<0.05; Fig. 1E-G).

TRPC1 knockdown attenuates the chemoresistance of NSCLC cells to CDDP

To further investigate the effect of TRPC1 knockdown on NSCLC chemoresistance, A549/CDDP and H460/CDDP cells were transfected with one of three siRNAs targeting TRPC1. RT-qPCR (Fig. 2A and B) and western blot analysis (Fig. 2C-E) revealed that the si-TRPC1-2 construct exhibited the highest TRPC1 knockdown activity in both A549/CDDP and H460/CDDP cells. Therefore, si-TRPC1-2 was chosen for the subsequent experiments. The IC50 value of CDDP was significantly reduced in TRPC1-knockdown A549/CDDP cells compared with the si-NC group (11.78 vs. 21.58 µM; P<0.01); this trend was also observed in H460/CDDP cells (23.76 vs. 43.11 µM; P<0.05; Fig. 3A-D).

TRPC1 knockdown reduces NSCLC stemness

The number of spheres formed was markedly decreased in both the TRPC1-knockdown A549/CDDP (P<0.01; Fig. 4A and B) and H460/CDDP (P<0.05; Fig. 4A and C) cells compared with the respective si-NC group. Furthermore, in A549/CDDP cells, the expression levels of CD133 (P<0.01) and CD44 (P<0.05) were reduced in the si-TRPC1 group compared with the si-NC group (Fig. 4D and E). However, only CD133 expression was significantly downregulated in TRPC1-depleted H460/CDDP cells (the transfection efficiency of si-TRPC1 transfected H460/CDDP cells was not sufficient) (P<0.05; Fig. 4D and F).

TRPC1 knockdown reduces PI3K/AKT signaling

TRPC1 knockdown reduced the p-PI3K/PI3K and p-AKT/AKT ratios in both A549/CDDP (both P<0.05; Fig. 5A and B) and H460/CDDP cells (both P<0.01; Fig. 5A and C) compared with the respective si-NC group.

Cell treatment with 740 Y-P alleviates the effect of TRPC1 knockdown on NSCLC cell chemoresistance and stemness

A549/CDDP (both P<0.01; Fig. 6A and B) and H460/CDDP (both P<0.001; Fig. 6A and C) cell treatment with 740 Y-P increased the p-PI3K/PI3K and p-AKT/AKT ratios, which were previously decreased by TRPC1 knockdown. Additionally, treatment with 740 Y-P increased the IC50 values of CDDP in si-TRPC1-transfected A549/CDDP cells (23.18 vs. 12.96 µM; P<0.05; Fig. 7A). Similar results were observed in the H460/CDDP cells (44.41 vs. 21.60 µM; P<0.01; Fig. 7B). Finally, the number of spheres formed was increased in the TRPC1-knockdown A549/CDDP (P<0.05; Fig. 7C and E) and H460/CDDP cells (P<0.05; Fig. 7D and E) treated with 740 Y-P.

Discussion

TRPC1, a member of the calcium channel family of proteins, modulates several cellular functions including cell proliferation, survival, and migration (18). The role of TRPC1 in cancer has been gradually uncovered (1923). More specifically, a study illustrated that TRPC1 inhibited cell proliferation and invasion in esophageal squamous cell carcinoma (19). Another study revealed that TRPC1 overexpression promoted the migration of human malignant glioma cells (20). Additionally, a previous study suggested that TRPC1 could exacerbate metastasis in gastric cancer via regulation of a circular RNA-7/microRNA-135a-5p axis (21). More importantly, the regulatory role of TRPC1 in NSCLC has also been investigated; TRPC1 was shown to interact with EGFR and correspondingly facilitate the proliferation of NSCLC cells (22). In addition, another study indicated that TRPC1 could promote the proliferation of NSCLC cells (23). However, the effect of TRPC1 on NSCLC chemoresistance and stemness has not been previously explored. Consequently, the present study is the first to explore the regulatory effect of TRPC1 on NSCLC chemoresistance and stemness, as well as to determine the underlying mechanism.

Regarding the role of TRPC1 on chemoresistance, it has been reported that TRPC1 modulates chemoresistance in several types of cancer (24,25). A previous study demonstrated that TRPC1 enhanced store-operated Ca2+ entry and was thus involved in the resistance of breast cancer cells to chemotherapeutic drugs, such as cisplatin, 5-fluorouracil and paclitaxel (24). Another study showed that TRPC1 could interact with stromal interaction molecule 1 and calcium release-activated calcium channel protein 1 to induce chemoresistance in hepatocellular carcinoma (25). However, its effect on chemoresistance in NSCLC cells remains unknown. In the current study, TRPC1 was upregulated in chemoresistant NSCLC cells, while TRPC1 knockdown restored chemosensitivity in CDDP-resistant NSCLC cells. The above effects could be due to the fact that TRPC1 knockdown could prevent autophagy, which in turn could inhibit tumor cell apoptosis mediated by chemotherapeutic drugs. Therefore, TRPC1 silencing may decrease chemoresistance in NSCLC cells (26,27). Secondly, TRPC1 knockdown could inhibit the activity of CDK1 and CyclinB1, which are involved in the G2 to the M phase transition of the cell cycle (28). Additionally, chemotherapeutic drugs, such as CDDP, bind with DNA primarily during the G2/M phase transition to exert a cytotoxic effect (29). Therefore, TRPC1 knockdown may enhance chemosensitivity in chemoresistant NSCLC cells.

Cancer stemness plays a critical role in the pathology of chemoresistance. It has been reported that the microenvironment of cancer stem cells can promote chemoresistance through several factors (30). Regarding the effect of TRPC1 on cancer stemness, a previous study reported that TRPC1 was associated with stemness in dental pulp stem cells (31). Herein, TRPC1 knockdown decreased sphere formation and reduced the expression levels of CD133 and CD44 in chemoresistant NSCLC cells. A possible explanation could be that TRPC1 silencing could attenuate EMT via inhibiting the PI3K/AKT signaling pathway. It has been reported that EMT is associated with cancer stemness in several types of cancer (32,33), suggesting that TRPC1 knockdown could reduce cancer stemness in NSCLC cells. In addition, another interesting finding of the present study was that there was no difference in CD44/GAPDH expression between the si-NC transfected H460/CDDP cells and si-TRPC1 transfected H460/CDDP cells, whereas CD44/GAPDH was lower in the si-TRPC1 transfected A549/CDDP cells compared with si-NC transfected A549/CDDP cells; a possible explanation for this could be that: TRPC1 has a limited effect on CD44 expression in H460 cells compared with A549/CDDP cells. Hence, TRPC1 has a limited effect on CD44 expression in H460/CDDP cells compared with that in A549/CDDP cells; however, this effect requires further validation.

The effect of TRPC1 on PI3K/AKT signaling has been previously investigated. TRPC1 promotes hypoxia-associated EMT via activation of the PI3K/AKT signaling pathway in breast cancer cells (11). Another study showed that TRPC1 could enhance the resistance of colon cancer cells to drugs by regulating PI3K/AKT signaling (29). However, whether these mechanisms occur in NSCLC cells has not been determined. Herein, it was shown that TRPC1 knockdown inhibited PI3K/AKT signaling. However, cell treatment with 740 Y-P promoted PI3K/AKT signaling, chemoresistance, and stemness in TRPC1-depleted chemoresistant NSCLC cells. The above finding could be due to an increase in the influx of Ca2+ from the extracellular environment, which in turn may reverse the TRPC1 knockdown-mediated inhibition of downstream AKT phosphorylation. That is, TRPC1 knockdown attenuated the PI3K/AKT signaling pathway, further enhancing chemosensitivity and attenuating the stemness of chemoresistant NSCLC cells (34).

The current study has some limitations: i) There are no data to show whether this mechanism is observed in vivo; ii) Since TRPC1 expression was relatively high in CDDP-resistant NSCLC cells, a TRPC1 overexpression plasmid may not exert any notable effects on CDDP-resistant NSCLC cells, thus overexpression plasmids were not used in the current study. However, experiments where TRPC1 expression is overexpressed following knockdown may have value to show the necessity and sufficiency of TRPC1 expression; iii) although some previous studies have already investigated the regulatory role of TRPC1 on the proliferation and differentiation of NSCLC cells (22,23), the absence of these experiments to evaluate the effect of TRPC1 on these phenotypes in NSCLC cells is a limitation of the present study; iv) the absence of non-cancerous cell lines as a negative control is a major limitation of the present study.

Collectively, the results of the current study suggested that targeting TRPC1 could attenuate NSCLC stemness and chemoresistance via inactivation of PI3K/AKT signaling; however, further studies are required to determine if this effect is observed in vivo.

Acknowledgments

Not applicable.

Funding

This study was supported by the Beijing Medical Award Foundation of China (grant no. YXJL-2020-0785-0178).

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

JG contributed to the conception and design of the study. JJ and XY performed the experiment. JH was responsible for the interpretation of data for the work. YZ, HZ, KZ and YW contributed to the data acquisition, data analysis and data interpretation. All authors contributed to the drafting/revising of article. JH and JG confirm the authenticity of all the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

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

2 

Thai AA, Solomon BJ, Sequist LV, Gainor JF and Heist RS: Lung cancer. Lancet. 398:535–554. 2021. View Article : Google Scholar : PubMed/NCBI

3 

Rodak O, Peris-Diaz MD, Olbromski M, Podhorska-Okołów M and Dzięgiel P: Current landscape of non-small cell lung cancer: Epidemiology, histological classification, targeted therapies, and immunotherapy. Cancers (Basel). 13:47052021. View Article : Google Scholar : PubMed/NCBI

4 

Shah P, Sands J and Normanno N: The expanding capability and clinical relevance of molecular diagnostic technology to identify and evaluate EGFR mutations in advanced/metastatic NSCLC. Lung Cancer. 160:118–126. 2021. View Article : Google Scholar : PubMed/NCBI

5 

Chaft JE, Rimner A, Weder W, Azzoli CG, Kris MG and Cascone T: Evolution of systemic therapy for stages I–III non-metastatic non-small-cell lung cancer. Nat Rev Clin Oncol. 18:547–557. 2021. View Article : Google Scholar : PubMed/NCBI

6 

Fadejeva I, Olschewski H and Hrzenjak A: MicroRNAs as regulators of cisplatin-resistance in non-small cell lung carcinomas. Oncotarget. 8:115754–115773. 2017. View Article : Google Scholar : PubMed/NCBI

7 

de Graeff A, Slebos RJ and Rodenhuis S: Resistance to cisplatin and analogues: Mechanisms and potential clinical implications. Cancer Chemother Pharmacol. 22:325–332. 1988. View Article : Google Scholar : PubMed/NCBI

8 

Jing N, Gao WQ and Fang YX: Regulation of formation, stemness and therapeutic resistance of cancer stem cells. Front Cell Dev Biol. 9:6414982021. View Article : Google Scholar : PubMed/NCBI

9 

Elzamzamy OM, Penner R and Hazlehurst LA: The role of TRPC1 in modulating cancer progression. Cells. 9:3882020. View Article : Google Scholar : PubMed/NCBI

10 

Baudel MASM, Shi J, Large WA and Albert AP: Insights into activation mechanisms of store-operated TRPC1 channels in vascular smooth muscle. Cells. 9:1792020. View Article : Google Scholar : PubMed/NCBI

11 

Van den Eynde C, De Clercq K, Van Bree R, Luyten K, Annibali D, Amant F, Han S, Van Nieuwenhuysen E, Baert T, Peeraer K, et al: TRP channel expression correlates with the epithelial-mesenchymal transition and high-risk endometrial carcinoma. Cell Mol Life Sci. 79:262021. View Article : Google Scholar : PubMed/NCBI

12 

Tai YK, Chan KKW, Fong CHH, Ramanan S, Yap JLY, Yin JN, Yip YS, Tan WR, Koh APF, Tan NS, et al: Modulated TRPC1 expression predicts sensitivity of breast cancer to doxorubicin and magnetic field therapy: Segue towards a precision medicine approach. Front Oncol. 11:7838032022. View Article : Google Scholar : PubMed/NCBI

13 

De Las Rivas J, Brozovic A, Izraely S, Casas-Pais A, Witz IP and Figueroa A: Cancer drug resistance induced by EMT: Novel therapeutic strategies. Arch Toxicol. 95:2279–2297. 2021. View Article : Google Scholar : PubMed/NCBI

14 

Pan C, Jin L, Wang X, Li Y, Chun J, Boese AC, Li D, Kang HB, Zhang G, Zhou L, et al: Inositol-triphosphate 3-kinase B confers cisplatin resistance by regulating NOX4-dependent redox balance. J Clin Invest. 129:2431–2445. 2019. View Article : Google Scholar : PubMed/NCBI

15 

Tian S, Lou L, Tian M, Lu G, Tian J and Chen X: MAPK4 deletion enhances radiation effects and triggers synergistic lethality with simultaneous PARP1 inhibition in cervical cancer. J Exp Clin Cancer Res. 39:1432020. View Article : Google Scholar : PubMed/NCBI

16 

Wang J, Liang J, Li H, Han J, Jiang J, Li Y, Feng Z, Zhao R and Tian H: Oncogenic role of abnormal spindle-like microcephaly-associated protein in lung adenocarcinoma. Int J Oncol. 58:232021. View Article : Google Scholar : PubMed/NCBI

17 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

18 

Nesin V and Tsiokas L: TRPC1. Handb Exp Pharmacol. 222:15–51. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Zeng YZ, Zhang YQ, Chen JY, Zhang LY, Gao WL, Lin XQ, Huang SM, Zhang F and Wei XL: TRPC1 inhibits cell proliferation/invasion and is predictive of a better prognosis of esophageal squamous cell carcinoma. Front Oncol. 11:6277132021. View Article : Google Scholar : PubMed/NCBI

20 

Bomben VC, Turner KL, Barclay TT and Sontheimer H: Transient receptor potential canonical channels are essential for chemotactic migration of human malignant gliomas. J Cell Physiol. 226:1879–1888. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Zhang Z, Ren L, Zhao Q, Lu G, Ren M, Lu X, Yin Y, He S and Zhu C: TRPC1 exacerbate metastasis in gastric cancer via ciRS-7/miR-135a-5p/TRPC1 axis. Biochem Biophys Res Commun. 529:85–90. 2020. View Article : Google Scholar : PubMed/NCBI

22 

Tajeddine N and Gailly P: TRPC1 protein channel is major regulator of epidermal growth factor receptor signaling. J Biol Chem. 287:16146–16157. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Jiang HN, Zeng B, Zhang Y, Daskoulidou N, Fan H, Qu JM and Xu SZ: Involvement of TRPC channels in lung cancer cell differentiation and the correlation analysis in human non-small cell lung cancer. PLoS One. 8:e676372013. View Article : Google Scholar : PubMed/NCBI

24 

Hasna J, Hague F, Rodat-Despoix L, Geerts D, Leroy C, Tulasne D, Ouadid-Ahidouch H and Kischel P: Orai3 calcium channel and resistance to chemotherapy in breast cancer cells: The p53 connection. Cell Death Differ. 25:693–707. 2018. View Article : Google Scholar : PubMed/NCBI

25 

Tang BD, Xia X, Lv XF, Yu BX, Yuan JN, Mai XY, Shang JY, Zhou JG, Liang SJ and Pang RP: Inhibition of Orai1-mediated Ca2+ entry enhances chemosensitivity of HepG2 hepatocarcinoma cells to 5-fluorouracil. J Cell Mol Med. 21:904–915. 2017. View Article : Google Scholar : PubMed/NCBI

26 

Sukumaran P, Sun Y, Antonson N and Singh BB: Dopaminergic neurotoxins induce cell death by attenuating NF-κB-mediated regulation of TRPC1 expression and autophagy. FASEB J. 32:1640–1652. 2018. View Article : Google Scholar : PubMed/NCBI

27 

Usman RM, Razzaq F, Akbar A, Farooqui AA, Iftikhar A, Latif A, Hassan H, Zhao J, Carew JS, Nawrocki ST and Anwer F: Role and mechanism of autophagy-regulating factors in tumorigenesis and drug resistance. Asia Pac J Clin Oncol. 17:193–208. 2021. View Article : Google Scholar : PubMed/NCBI

28 

Ma S, Kong D, Fu X, Liu L, Liu Y, Xue C, Tian Z, Li L and Liu X: p53-induced autophagy regulates chemotherapy and radiotherapy resistance in multidrug resistance cancer cells. Dose Response. 19:155932582110480462021. View Article : Google Scholar : PubMed/NCBI

29 

Galluzzi L, Senovilla L, Vitale I, Michels J, Martins I, Kepp O, Castedo M and Kroemer G: Molecular mechanisms of cisplatin resistance. Oncogene. 31:1869–1883. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Barbato L, Bocchetti M, Di Biase A and Regad T: Cancer stem cells and targeting strategies. Cells. 8:9262019. View Article : Google Scholar : PubMed/NCBI

31 

Yao W, Wang L, Huang H, Li X, Wang P, Mi K, Cheng J, Liu H, Gu C, Huang L and Huang J: All-trans retinoic acid reduces cancer stem cell-like cell-mediated resistance to gefitinib in NSCLC adenocarcinoma cells. BMC Cancer. 20:3152020. View Article : Google Scholar : PubMed/NCBI

32 

Pan G, Liu Y, Shang L, Zhou F and Yang S: EMT-associated microRNAs and their roles in cancer stemness and drug resistance. Cancer Commun (Lond). 41:199–217. 2021. View Article : Google Scholar : PubMed/NCBI

33 

Xu F, Liu XC, Li L, Ma CN and Zhang YJ: Effects of TRPC1 on epithelial mesenchymal transition in human airway in chronic obstructive pulmonary disease. Medicine (Baltimore). 96:e81662017. View Article : Google Scholar : PubMed/NCBI

34 

Zanou N, Schakman O, Louis P, Ruegg UT, Dietrich A, Birnbaumer L and Gailly P: Trpc1 ion channel modulates phosphatidylinositol 3-kinase/Akt pathway during myoblast differentiation and muscle regeneration. J Biol Chem. 287:14524–14534. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2023
Volume 25 Issue 6

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
Jin J, Yan X, Zhao Y, Zhang H, Zhuang K, Wen Y, He J and Gao J: Targeting transient receptor potential canonical 1 reduces non‑small cell lung cancer chemoresistance and stemness via inhibition of PI3K/AKT signaling. Oncol Lett 25: 224, 2023
APA
Jin, J., Yan, X., Zhao, Y., Zhang, H., Zhuang, K., Wen, Y. ... Gao, J. (2023). Targeting transient receptor potential canonical 1 reduces non‑small cell lung cancer chemoresistance and stemness via inhibition of PI3K/AKT signaling. Oncology Letters, 25, 224. https://doi.org/10.3892/ol.2023.13810
MLA
Jin, J., Yan, X., Zhao, Y., Zhang, H., Zhuang, K., Wen, Y., He, J., Gao, J."Targeting transient receptor potential canonical 1 reduces non‑small cell lung cancer chemoresistance and stemness via inhibition of PI3K/AKT signaling". Oncology Letters 25.6 (2023): 224.
Chicago
Jin, J., Yan, X., Zhao, Y., Zhang, H., Zhuang, K., Wen, Y., He, J., Gao, J."Targeting transient receptor potential canonical 1 reduces non‑small cell lung cancer chemoresistance and stemness via inhibition of PI3K/AKT signaling". Oncology Letters 25, no. 6 (2023): 224. https://doi.org/10.3892/ol.2023.13810