The Fanconi anemia pathway sensitizes to DNA alkylating agents by inducing JNK-p53-dependent mitochondrial apoptosis in breast cancer cells

Retraction in: /10.3892/ijo.2022.5343

  • Authors:
    • Lin Zhao
    • Yanlin Li
    • Miao He
    • Zhiguo Song
    • Shu Lin
    • Zhaojin Yu
    • Xuefeng Bai
    • Enhua Wang
    • Minjie Wei
  • View Affiliations

  • Published online on: April 28, 2014     https://doi.org/10.3892/ijo.2014.2400
  • Pages: 129-138
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The Fanconi anemia/BRCA (FA/BRCA) DNA damage repair pathway plays a pivotal role in the cellular response to DNA alkylating agents and greatly influences drug response in cancer treatment. However, the molecular mechanisms underlying the FA/BRCA pathway reversed resistance have received limited attention. In the present study, we investigated the effect of Fanconi anemia complementation group F protein (FANCF), a critical factor of the FA/BRCA pathway, on cancer cell apoptosis induced by DNA alkylating agents such as mitomycin c (MMC). We found that FANCF shRNA potentiated MMC-induced cytotoxicity and apoptosis in MCF-7 and MDA-MB-231 breast cancer cells. At a mechanistic level, FANCF shRNA downregulated the anti-apoptotic protein Bcl-2 and upregulated the pro-apoptotic protein Bax, accompanied by release of cyt-c and smac into the cytosol in MMC-treated cells. Furthermore, activation of caspase-3 and -9, other than caspase-8, cleavage of poly(ADP ribose) polymerase (PARP), and a decrease of mitochondrial membrane potential (MMP) indicated that involvement of the mitochondrial apoptotic pathway in FANCF silencing of MMC-treated breast cancer cells. A decrease in IAP family proteins XIAP and survivin were also observed following FANCF silencing in MMC-treated breast cancer cells. Notably, FANCF shRNA was able to increase p53 levels through activation of the JNK pathway in MMC-treated breast cancer cells. Furthermore, p53 inhibition using pifithrin-α abolished the induction of caspase-3 and PARP by FANCF shRNA and MMC, indicating that MMC-induced apoptosis is substantially enhanced by FANCF shRNA via p53-dependent mechanisms. To our knowledge, we provide new evidence for the potential application of FANCF as a chemosensitizer in breast cancer therapy.

Introduction

Breast cancer has a high rate of morbidity and mortality, which seriously threaten the health of women (1). To date, chemotherapy has been the most frequently used method for treating breast cancer and other cancers. Drug treatment has significantly prolonged survival time of breast cancer patients, and improved their prognosis. Of the limited number of clinically active anticancer chemotherapeutic compounds, alkylating agents are invaluable drugs which are electrophilic and trigger cell death by covalently binding to cellular nucleophiles such as DNA and proteins. Hundreds of alkylating compounds have been tested for anticancer activity. Despite their frequent use, the therapeutic efficacy of these agents is limited by the development of resistance.

Mitomycin c (MMC), isolated from streptomyces caespitosus, is an alkylating agent that results in damaged DNA cross-links and inhibition of the DNA replication apparatus, leading to cytotoxicity and cell death (2). The therapeutic value of MMC depends on the capability of the cells to repair DNA damage (3). A new and emerging concept designed to sensitize cancer cells to DNA-damaging agents (i.e., chemotherapy and/or radiation) is inhibition of various proteins in the DNA repair pathways. We are focusing on inhibition and manipulation of the Fanconi anemia/breast cancer susceptibility gene (FA/BRCA) pathway in breast cancer.

Fanconi anemia (FA) is an inherited chromosomal instability disorder manifesting a variety of congenital malformations, pancytopenia, and a predisposition to cancer (4). FA protein is a multifunctional protein composed of 15 FA complementation groups (FANC A–C, D1, D2, E, F, G, I, J, L, M, N, O and P) (5), and is involved in cell cycle, DNA damage and repair, apoptosis, gene transcription, and gene stability through common FA/BRCA cellular pathways (6). Following exposure to DNA-damaging agents or during the DNA synthesis (S) phase of the cell cycle, eight of the FA proteins (A, B, C, E, F, G, L and M) assemble into multisubunit nuclear complex that activates the monoubiquitination of the downstream FANCD2 (D2) protein at lysine 561. Monoubiquitination of the D2 protein targets its translocation to BRCA1-, FANCD1/BRCA2- and RAD51-containing nuclear DNA repair foci (7,8). Furthermore, this pathway has been shown to play an important role in the acquisition of drug resistance (911). Disruption of the FA/BRCA pathway results in chromosome instability and hypersensitivity to DNA alkylating agents such as MMC (12,13).

In particular, the FA complex plays a critical role in cell response to chemotherapy-induced DNA damage. As an adaptor protein, FANCF interacts with the FANCC/FANCE subunit through its N-terminal, and with the FANCA/FANCG subunit through its C-terminal. Thus, the FANCF subunit functions as the stabilizing component of the larger FA complex and maintains the biological functions of the FA/BRCA pathway (14). FANCF inhibition mediated by gene promoter methylation and small interfering (si) RNAs, which can promote drug sensitivity of tumor cells, such as ovarian cancer (15), multiple myeloma (9), cervical cancer (16) and glioma (10). More recently, we reported that gene silencing of FANCF induced dysfunction of FA/BRCA pathway and potentiated the sensitivity to mitoxantrone and MMC in breast cancer cells (17,18). However, despite major advances in the understanding of the biochemistry of FA/BRCA pathway, little is known about the mechanisms through which pathway lead to the increased sensitivity of alkylating agents including MMC when FA/BRCA pathway was disrupted. Recent studies have shown that MMC induces apoptosis by both activating caspase-3 and decreasing bcl-2 level (19,20). On the other hand, we have previously shown that disruption of FA/BRCA pathway mediated by FANCF-silencing induced apoptosis via the activation of the mitochondrial apoptosis pathway in breast cancer cells (17). Therefore, we hypothesize that apoptosis-related proteins might play a crucial role in FANCF-sensitizing the MMC in breast cancer cells.

In this context, we investigated the effect of FA/BRCA pathway dysfunction mediated by FANCF inhibition on MMC sensitivity and its underlying mechanisms. In this report, we demonstrate that specific short hairpin RNA (shRNA) decreases the levels of FANCF, mediates FA/ BRCA pathway dysfunction, and potentiates the sensitivity of breast cancer to the alkylating agent MMC. This may be due to the p53-dependent mitochondria-regulated intrinsic death-signaling pathway. We also demonstrated that FANCF inhibition enhanced chemotherapeutic-induced apoptosis in human breast cancer cells through JNK MAPK signaling pathway. To our knowledge, our results add new evidence for the potential application of FANCF as a chemosensitizer in breast cancer therapy.

Materials and methods

Cell culture

Estrogen receptor α (ERα)-positive human breast cancer cell lines MCF-7 and ERα-negative MDA-MB-231 cells were obtained from the American Type Culture Collection (ATCC). Adherent cells were maintained in Dulbecco’s modified Eagle’s medium (DMEM) containing 10% fetal bovine serum, 100 U/ml penicillin, and 100 mg/ml streptomycin in a humidified atmosphere with 5% CO2 at 37°C

Antibodies and reagents

Antibodies against p53, phospho-p53, Bcl-2, Bax, survivin, X-linked inhibitor of apoptosis protein (XIAP), cytochrome c (cyt-c), second mitochondria-derived activator of caspases (smac) and β-actin were from Santa Cruz Biotechnology (Santa Cruz, CA, USA). Antibodies against FANCF, FANCD2, cleaved-caspase-6, cleaved-caspase-8, cleaved-caspase-9, poly(ADP ribose) polymerase (PARP) were from Abcam Inc. (Cambridge, MA, USA). MMC and p53 inhibitor pifithrin-α was purchased from Sigma Chemical Co. (St. Louis, MO, USA).

Construction of the FANCF shRNA expression vector

The FANCF shRNA expression vector was used to achieve specific downregulation of FANCF, as previously described (17). The sequences of the oligonucleotides to construct FANCF shRNA expressing vector was designed as follows: sense, 5′GATCCGCTTCCTGAAGGTGATAGCGTTCAAGAGAC GCTATCACCTTCAGGAAGTTTTTTGGAAA-3′ and anti-sense, 5′-AGCTTTTCCAAAAAACTTCCTGAAGGTGAT AGCGTCTCTTGAACGCTATCACCTTCAGGAAGCG-3′. A scrambled shRNA with no significant homology to human gene sequences was used as a negative control to detect non-specific effects.

FANCF shRNA transfection

Cells were seeded into 6-well plates (3×105 cells/well) or 100-mm dishes (2×106 cells) and were allowed to adhere for 24 h, and after 24 h, cells were transfected with the pSilencer™ 4.1-CMV Control shRNA vector (control shRNA) or pSilencer 4.1-CMV FANCF shRNA vector (FANCF shRNA) using Lipofectamine 2000 (Invitrogen, Carlsbad, CA, USA) according to the manufacturer’s instructions. After 4 h, the culture medium was replaced with fresh media supplemented with 10% FBS, and the cells were harvested at 24 and 48 h after transfection.

Western blot analysis

Western blot analysis for the presence of specific proteins or for phosphorylated forms of proteins was performed on whole-cell sonicates and lysates from MCF-7 and MDA-MB-231 cells. Protein (30–50 μg) was mixed 4:1 with 5X sample buffer (20% glycerol, 4% sodium dodecyl sulfate, 10% β-mercaptoethanol, 0.05% bromophenol blue and 1.25 M Tris-HCl, pH 6.8; all from Sigma). Equal amount of proteins was loaded onto a 10% sodium dodecyl sulfatepolyacrylamide gel. Cell proteins were transferred to PVDF membranes. The PVDF membranes were then blocked with 5% milk in Tris-buffered saline with 0.1% Tween-20 and then incubated with an appropriate dilution of antibodies (1:1,000 to 1:2,000) overnight at 4°C. The blots were washed and incubated for 1 h with horseradish peroxidase-conjugated anti-IgG antibody (Santa Cruz). Immunocomplexes were visualized by chemiluminescence using ECL (Santa Cruz).

Cell viability assay

The cell viability was assessed using Cell count kit-8 (Dojindo Molecular Technologies, Inc., Gaithersburg, MD, USA). Cells were seeded at 5×103 cells/well in 96-well plates and allowed to grow in the growth medium for 24 h. Cells were transfected with control or FANCF shRNA for 48 h, and then treated with MMC at different concentration (0. l, 0.3, 1, 3, 10, 30 and 100 μM of MMC, respectively) for 24 h. CCK-8 solution (10 μl) was added to 100 μl of media in each well and absorbance was determined at 450 nm after 1 h of incubation at 37°C.

Flow cytometry

Flow cytometry analysis was performed on a FACSCalibur (Becton-Dickinson). For determination of the cell cycle by exclusion of propidium iodide (PI), 500 μl of cell culture were incubated with 30 μg/ml PI for 1 h at room temperature prior to analysis. The cationic fluorescent carbocyanine dye, 5, 5’, 6, 6’-tetrachloro-1,1’,3,3’-tetraethylbenzimidazolyl carbocyanine iodide (JC-1) was used to assess changes in the mitochondrial membrane potential (ΔΨm) observed in apoptotic cells. Cells were incubated for 15 min at 37°C with 15 μg/ml JC-1 before analysis. For determination of apoptotic cells, cells were harvested, washed twice with phosphate-buffered saline (PBS), then incubated for 15 min at room temperature with a solution of fluorescence isothiocyanate (FITC) conjugated Annexin V (2.5 μg/ml) and PI (5 μg/ml) (all from Sigma), and analyzed for apoptosis.

Statistical analysis

Data are presented as the mean ± SD. The data are representative of the averages of at least three independent experiments. Data were analyzed using the one-way ANOVA with post-hoc analysis. P<0.05 was considered statistically significant.

Results

FANCF suppression by shRNA sensitizes breast cancer cells to the alkylating agent MMC

Firstly, we examined the effects of the alkylating agent MMC on the growth of MCF-7 and MDA-MB-231 breast cancer cells. Cellular growth, as determined by MTT assays, MMC dose-dependently inhibited the survival of MCF-7 and MDA-MB-231 with the IC50 of 8.67 and 11.73 μM, respectively (Fig. 1A).

To identify the effect of FANCF expression on MMC-mediated cell proliferation. ShRNA was used to knock down FANCF expression in MCF-7 and MDA-MB-231 breast cancer cells. To verify the results of gene silencing, FANCF expression was detected by western blotting at 24 and 48 h post-transfection. We found that expression of FANCF in the two cell lines (MCF-7 and MDA-MB-231) was inhibited in a time-dependent manner, as compared with the control (cells treated with scrambled shRNA). The results confirmed that FANCF expression was inhibited by transfection with shRNA targeting FANCF (Fig. 1B). Then, the antiproliferative actions of MMC were evaluated in FANCF-silenced MCF-7 and MDA-MB-231 cells. Co-treatment of FANCF shRNA with MMC caused a much greater decrease in viability than MMC alone, the IC50 of MMC in FANCF-silenced MCF-7 and MDA-MB-231 cells reduced to 1.21 and 2.29 μM, respectively (Fig. 1C). These results suggested that FANCF silencing potentiated the cytotoxic effects of MMC on breast cancer cells.

Silencing of FANCF potentiates MMC-induced cell apoptosis

To determine whether increased cytotoxity of MMC in FANCF-silenced cells involved apoptosis, the percentage of apoptotic cells was assessed by Annexin V-FITC and PI double staining, followed by flow cytometric analysis. Apoptosis was detected using flow cytometry in both MCF-7 and MDA-MB-231 cell lines. It was observed that MMC increased the percentage of cells undergoing apoptosis. After FANCF shRNA/MMC combination, a significant increase in the population of cells undergoing apoptosis was recorded compared to the MMC-treated cells (P<0.05) (Fig. 2). The results indicated that FANCF shRNA potentiated MMC-induced cytoxicity in MCF-7 and MDA-MB-231 breast cancer cells by inducing apoptosis.

The activation of caspase-3 and -9, but not caspase-8 following MMC treatment was substantially enhanced by FANCF shRNA. In order to study the mechanism of apoptosis involved in FANCF silencing-potentiated MMC sensitivity, we evaluated the effects of FANCF knockdown on several molecules involved in apoptosis in MMC-treated cells. In immunoblot experiments, MMC elicited caspase-3, -9 activation, and cleavage of PARP (a substrate of caspase-3), which were enhanced after FANCF silencing (Fig. 3). Although MCF-7 cells are deficient in caspase-3, they remain susceptible to cell death induced by several stimuli (21). Consistent with a report that other executive caspases, such as caspase-6 and -7, are able to substitute for caspase-3 during apoptosis, the co-treatment with MMC and FANCF shRNA elicited caspase-6 activation in MCF-7 cells. These findings suggest that FANCF shRNA decreases the viability of MMC-treated MCF-7 breast cancer cells by inducing apoptosis through a mechanism mediated by caspase-3, -6, and -9 and involving the intrinsic mitochondrial apoptosis pathway.

Mitochondrial release of cyt-c and smac with loss of mitochondrial membrane potential by MMC treatment are increased by FANCF shRNA

Mitochondrial dysfunction induced apoptosis which is often the consequence of a decrease of ΔΨm. To assess whether FANCF silencing affects the function of mitochondria, ΔΨm changes were measured by employing the mitochondrial fluorescent dye JC-1. As shown in Fig. 4A and B, FANCF silencing in MMC-treated MCF-7 and MDA-MB-231 cells resulted in a decrease of ΔΨm, compared to controls or MMC-treated alone.

We investigated the changes in expression of downstream molecules that occurred after the ΔΨm decreased. We analyzed the release of the proapoptotic mitochondrial intermembrane space proteins cyt-c and smac. Cyt-c was not expressed in the cytoplasm of FANCF-silenced MCF-7 or MDA-MB-231 cells. MMC treatment led to cyt-c and smac release (Fig. 4C and D). Compared with MMC treatment alone, FANCF shRNA and MMC co-treatment increased cyt-c and smac expression of MCF-7 and MDA-MB-231 cells. These findings indicated that the amount of cyt-c and smac in cytoplasm increased as a result of mitochondrial release in FANCF-silenced cells treated with MMC.

The changes in BCL-2 family by MMC treatment are potentiated by FANCF shRNA

To further characterize the molecular mechanisms of the induction of apoptosis by FANCF knockdown, the expression of the pro-apoptotic protein (Bax) and the anti-apoptotic proteins (XIAP, Bcl-2, survivin) of the Bcl-2 family were detected in MCF-7 and MDA-MB-231 cells treated with FANCF shRNA and/or MMC. We found that MMC decreased the expression of XIAP, survivin and Bcl-2, increased Bax expression (Fig. 3A), whereas co-treatment of FANCF shRNA with MMC resulted in enhanced inhibition of XIAP, survivin and Bcl-2. Similarly, MMC in combination with FANCF shRNA induced significant increases in Bax expression when compared with MMC or FANCF shRNA treatment alone (Fig. 5).

p53 is required for FANCF shRNA -induced apoptosis in MMC-treated breast cancer cells

p53 mediates apoptosis in cells suffering from serious DNA damage. Previously, it was reported that MMC causes an increase in the cellular p53 level (22,23). Therefore, we further examined the role of p53 in FANCF shRNA and MMC induced apoptosis. Based on western blotting, an increased expression of p53 protein was seen in MMC-treated cells, and these effects were enhanced by FANCF shRNA to a large extent (Fig. 6A and B). To further confirm the role of p53 in mediating FANCF shRNA-induced apoptosis, we used a p53 inhibitor (pifithrin-α) and tested whether it would prevent FANCF shRNA-induced apoptosis. As shown in Fig. 6C and D, pifithrin α prevents FANCF-shRNA induced activation of caspase-3 and PARP cleavage. These results indicate that p53 is required for FANCF shRNA-induced apoptosis in MMC-treated breast cancer cells.

Inhibitor of JNK protects MMC-treated cells from FANCF shRNA-induced p53 activation in MMC-treated cells

It was shown that MAPK signaling is involved in several events of cellular stress and stimuli-induced cell apoptosis (24). Silencing of the FANCF can cause activation of the MAPK signaling pathway, as reported by us previously (17). Therefore, we examined changes in the expression of proteins association with the MAPK pathway, including JNK, ERK, and p38 in breast cancer cells following FANCF shRNA and MMC treatment. The phosphorylation levels of p38 and JNK were apparently increased in response to the FANCF shRNA and MMC treatment; however, no significant changes of phosphorylation levels of ERK were observed (Fig. 7A and B). These results suggested that sustained activation of the p38 and JNK is involved in FANCF shRNA-induced apoptosis in MCF-7 and MDA-MB-231 cells.

It is clear that activation of the MAPK pathway can induce p53-mediated apoptosis (25). To further test whether activation of JNK and p38 pathways was involved in the FANCF silencing-induced activation of p53 expression, MCF-7 and MDA-MB-231 cells were treated with the p38 inhibitor, SB203580, and the JNK inhibitor, SP600125. After treatment with SB203580, FANCF shRNA had no detectable inhibitory effect on p53 expression. However, p53 expression was still activated by FANCF shRNA in SP600125 treated cells (Fig. 7C and D). These results demonstrated that FANCF gene silencing increased the expression of p53 through activation of JNK pathway.

All the above strongly suggest that mitochondrial apoptosis pathway participates in the FA pathway sensitizing to DNA alkylating agents in human breast carcinoma cells. We characterize the probable mechanisms of FANCF silencing on MMC-induced apoptosis in Fig. 8.

Discussion

The FA/BRCA pathway may display promising chemosensitive potential in cancers (26). Some tumor cells depend on this pathway for the maintenance of chromosome integrity and for their resistance to cytotoxic agents (9). Inhibition of the DNA repair pathway would confer enhanced drug sensitivity to tumor cells and may be useful in conjunction with more traditional cytotoxic treatment, such as alkylating agents (10). In spite of its emerging significance as a new target for preventing acquired drug resistance, the molecular mechanisms underlying the FA/BRCA pathway reversed resistance have received limited attention.

In this study, we therefore evaluated the mechanisms of RNAi-mediated FANCF (a critical factor of FA/BRCA pathway) gene silencing using the sensitizing DNA alkylating agent MMC cytoxicity in vitro in MCF-7 and MDA-MB-231 human breast cancer cells. The present study shows that RNAi suppression of FANCF sensitizes to the DNA alkylating agent by inducing JNK-p53-dependent mitochondrial apoptosis in breast cancer cells.

Reversing cancer cell resistance to apoptosis may be an effective way to suppress cancer malignancies (27). Our previous studies demonstrated that FANCF inhibition induced apoptosis of cancer cells such as ovarian (28) and breast cancer cells (17). In this study, we found that FANCF silencing potentiated the sensitivity of MMC, the combination treatment inhibited cell proliferation, increased apoptosis compared with MMC treatment alone (Figs. 1 and 2). Our results are in agreement with previous findings that FA cells are hypersensitive towards DNA crosslinking agents (29). Thus, the current results warrant further evaluation of the MMC and FANCF shRNA combination as a potential therapeutic regimen against human breast cancer.

The progression of apoptosis involves the activation of a cascade of proteases called caspases. Theoretically, the extrinsic pathway is related to the activation of caspase-8 and the intrinsic pathway is associated with activation of caspase-9. Both pathways converge to a common pathway involving the activation of caspase-3 (30). In the present study, caspase-3 and -9 is activated in FANCF-silenced MCF-7 and MDA-MB-231 cells treated with MMC, and subsequent cleavage of PARP, but caspase-8 expression is unaffected (Fig. 3). These findings demonstrate that the FANCF silencing-induced apoptosis in MMC-treated cells is mediated by the intrinsic mitochondrial pathway, rather than the extrinsic death receptor pathway. Therefore, enhanced MMC sensitivity in FANCF-silenced MCF-7 and MDA-MB-231 cells may partly be due to the regulation of the mitochondrial apoptosis pathway.

The ratio of anti- and pro-apoptotic protein expression, such as Bcl-2/Bax, is crucial for the induction of apoptosis, and it decides the susceptibility of cells to undergo apoptosis (31). Mitochondria play an important role in the signal transduction of apoptosis (32). The translocalization of apoptotic proteins from the cytosol to the mitochondria leads to the release of cyt-c and smac by a decrease in mitochondrial membrane potential (33). In the present study, we showed that FANCF silencing downregulated the expression of and Bcl-2 and upregulated the expression of Bax, and caused the loss of mitochondrial membrane potential and release of cyt-c and smac in MMC-treated MCF-7 and MDA-MB-231 cells (Fig. 4B), further confirmed that FANCF silencing-induced apoptosis with mitochondria-dependent apoptosis.

The inhibitor of apoptosis (IAP) family members survivin and XIAP are potent inhibitors of caspase-3 and -9 activity and have previously been shown to confer resistance to MMC (34). Our results showed that FANCF shRNA decreases the expression of XIAP and surviving (Fig. 5). The enhanced caspase-3 and -9 activation observed in cells treated with FANCF shRNA plus MMC could also be due to reduced levels of XIAP and survivin. Taken together, our results indicated that FANCF shRNA regulates expression levels of apoptosis-related proteins, causes cyt-c and smac release (Fig. 4B) and triggers caspase-dependent cell apoptotic death.

The tumor suppressor p53 has been implicated in many important cellular processes, including regulation of apoptotic cell death. p53 activates several important genes that are crucial for the execution of the intrinsic pathway of apoptosis including pro-apoptotic genes such as Bax (35), Noxa (36) and Puma (37). When p53-dependent apoptosis is employed in cells, these cells typically undergo the intrinsic cell death pathway. So, we studied the involvement of p53 in the apoptosis induced by FANCF knockdown. Our study found that FANCF shRNA activated p53 and that this activation is indispensable for FANCF shRNA-induced apoptosis (Fig. 6), indicating that FANCF shRNA induces apoptosis via a p53-dependent mechanism in MMC-treated cells. Consistent with these findings, Lui et al demonstrated that a defect in Fancd2 in developing zebrafish tissue induced inappropriate activation of p53-mediated programmed cell death (38), highlighting the relationship between p53 and the FA pathway.

p53 induction by MMC was reported previously by other groups (39). However, how MMC upregulates p53 has not been fully elucidated. MAPK regulates diverse cellular programs including embryogenesis, proliferation, differentiation and apoptosis (40). The MAPK family proteins are composed of three protein kinases: ERK1/2 (extracellular signal-regulated kinases 1 and 2), JNK (c-Jun N-terminal kinase) and p38. p53 is a downstream factor of the MAPK pathway, and can be activated by JNK (41). In our study, we found that MMC increases the levels of p-JNK and p-38. SP600125, a specific inhibitor of JNK, effectively blocked MMC-induced p53 in FANCF-silenced MCF-7 and MDA-MB-231 cells (Fig. 7), suggesting the pro-apoptotic effects of FANCF and in MCF-7 and MDA-MB-231 cells are mediated by the JNK-medicated upregulation of p53.

Collectively, our results provide the first mechanistic evidence that FANCF shRNA and MMC co-treatment results in JNK-mediated upregulation of p53, regulates expression levels of apoptosis-related proteins, causes cyt-c release and triggers caspase-dependent cell apoptotic death, thus rendering cancer cells more sensitive to the cytotoxic activities of MMC. In addition, our studies also show that the combined treatment with MMC and FANCF shRNA induces apoptosis in breast cancer cells. Thus, these studies suggest that FANCF shRNA can be administered in combination with MMC, especially for those tumors that develop resistance to MMC.

Acknowledgements

This study was supported by grants from National Natural Science Foundation of China (nos. 30873097, 81173092 and 81202551) and Liaoning Province Scientific Research Foundation of China (nos. 2011415052 and 20111107), and by Specialized Research Fund for the Doctoral Program of Higher Education (no. 20122104120031).

References

1. 

Parkin DM, Bray F, Ferlay J and Pisani P: Global cancer statistics, 2002. CA Cancer J Clin. 55:74–108. 2005. View Article : Google Scholar

2. 

Celli CM and Jaiswal AK: Role of GRP58 in mitomycin C-induced DNA cross-linking. Cancer Res. 63:6016–6025. 2003.PubMed/NCBI

3. 

McHugh PJ, Spanswick VJ and Hartley JA: Repair of DNA inter-strand crosslinks: molecular mechanisms and clinical relevance. Lancet Oncol. 2:483–490. 2001. View Article : Google Scholar : PubMed/NCBI

4. 

Bagby GC and Alter BP: Fanconi anemia. Semin Hematol. 43:147–156. 2006. View Article : Google Scholar

5. 

Stoepker C, Hain K, Schuster B, et al: SLX4, a coordinator of structure-specific endonucleases, is mutated in a new Fanconi anemia subtype. Nat Genet. 43:138–141. 2011. View Article : Google Scholar

6. 

Bogliolo M LA, Callén E, Castellà M, Cappelli E, Ramírez MJ, Creus A, Marcos R, Kalb R, Neveling K, Schindler D and Surrallés J: Histone H2AX and Fanconi anemia FANCD2 function in the same pathway to maintain chromosome stability. EMBO J. 26:1340–1351. 2007.PubMed/NCBI

7. 

Taniguchi T, Garcia-Higuera I, Xu B, et al: Convergence of the fanconi anemia and ataxia telangiectasia signaling pathways. Cell. 109:459–472. 2002. View Article : Google Scholar

8. 

Garcia-Higuera I, Taniguchi T, Ganesan S, et al: Interaction of the Fanconi anemia proteins and BRCA1 in a common pathway. Mol Cell. 7:249–262. 2001. View Article : Google Scholar : PubMed/NCBI

9. 

Chen Q, Van der Sluis PC, Boulware D, Hazlehurst LA and Dalton WS: The FA/BRCA pathway is involved in melphalan-induced DNA interstrand cross-link repair and accounts for melphalan resistance in multiple myeloma cells. Blood. 106:698–705. 2005. View Article : Google Scholar : PubMed/NCBI

10. 

Chen CC, Taniguchi T and D’Andrea A: The Fanconi anemia (FA) pathway confers glioma resistance to DNA alkylating agents. J Mol Med. 85:497–509. 2007. View Article : Google Scholar : PubMed/NCBI

11. 

Taniguchi T, Tischkowitz M, Ameziane N, et al: Disruption of the Fanconi anemia-BRCA pathway in cisplatin-sensitive ovarian tumors. Nat Med. 9:568–574. 2003. View Article : Google Scholar : PubMed/NCBI

12. 

D’Andrea AD and Grompe M: The Fanconi anaemia/BRCA pathway. Nat Rev Cancer. 3:23–34. 2003.

13. 

Auerbach AD: Fanconi anemia and its diagnosis. Mutat Res. 668:4–10. 2009. View Article : Google Scholar : PubMed/NCBI

14. 

Lyakhovich A SJ: New roads to FA/BRCA pathway. Cell Cycle. 6:1019–1023. 2007.PubMed/NCBI

15. 

Olopade OI and Wei M: FANCF methylation contributes to chemoselectivity in ovarian cancer. Cancer Cell. 3:417–420. 2003. View Article : Google Scholar : PubMed/NCBI

16. 

Narayan G A-PH, Nandula SV, Basso K, Sugirtharaj DD, Vargas H, Mansukhani M, Villella J, Meyer L, Schneider A, Gissmann L, Dürst M, Pothuri B and Murty VV: Promoter hypermethylation of FANCF: disruption of Fanconi Anemia-BRCA pathway in cervical cancer. Cancer Res. 64:2994–2997. 2004.PubMed/NCBI

17. 

Li Y, Zhao L, Sun H, et al: Gene silencing of FANCF potentiates the sensitivity to mitoxantrone through activation of JNK and p38 signal pathways in breast cancer cells. PLoS One. 7:e442542012. View Article : Google Scholar : PubMed/NCBI

18. 

Yu J, Zhao L, Li Y, et al: Silencing of Fanconi anemia complementation group F exhibits potent chemosensitization of mitomycin C activity in breast cancer cells. J Breast Cancer. 16:291–299. 2013. View Article : Google Scholar : PubMed/NCBI

19. 

Pirnia F, Schneider E, Betticher DC and Borner MM: Mitomycin C induces apoptosis and caspase-8 and -9 processing through a caspase-3 and Fas-independent pathway. Cell Death Differ. 9:905–914. 2002. View Article : Google Scholar : PubMed/NCBI

20. 

Kim TI, Choi SI, Lee HK, Cho YJ and Kim EK: Mitomycin C induces apoptosis in cultured corneal fibroblasts derived from type II granular corneal dystrophy corneas. Mol Vis. 14:1222–1228. 2008.PubMed/NCBI

21. 

Janicke RU: MCF-7 breast carcinoma cells do not express caspase-3. Breast Cancer Res Treat. 117:219–221. 2009. View Article : Google Scholar : PubMed/NCBI

22. 

Fritsche M, Haessler C and Brandner G: Induction of nuclear accumulation of the tumor-suppressor protein p53 by DNA-damaging agents. Oncogene. 8:307–318. 1993.PubMed/NCBI

23. 

Abbas T, Olivier M, Lopez J, et al: Differential activation of p53 by the various adducts of mitomycin C. J Biol Chem. 277:40513–40519. 2002. View Article : Google Scholar : PubMed/NCBI

24. 

Tournier C, Hess P, Yang DD, et al: Requirement of JNK for stress-induced activation of the cytochrome c-mediated death pathway. Science. 288:870–874. 2000. View Article : Google Scholar : PubMed/NCBI

25. 

Wu GS: The functional interactions between the p53 and MAPK signaling pathways. Cancer Biol Ther. 3:156–161. 2004. View Article : Google Scholar : PubMed/NCBI

26. 

D’Andrea AD: The Fanconi anemia/BRCA signaling pathway: disruption in cisplatin-sensitive ovarian cancers. Cell Cycle. 2:290–292. 2003.PubMed/NCBI

27. 

Wong RS: Apoptosis in cancer: from pathogenesis to treatment. J Exp Clin Cancer Res. 30:872011. View Article : Google Scholar : PubMed/NCBI

28. 

He M, Sun HG, Hao JY, et al: RNA interference-mediated FANCF silencing sensitizes OVCAR3 ovarian cancer cells to adriamycin through increased adriamycin-induced apoptosis dependent on JNK activation. Oncol Rep. 29:1721–1729. 2013.

29. 

D’Andrea AD and Grompe M: Molecular biology of Fanconi anemia: implications for diagnosis and therapy. Blood. 90:1725–1736. 1997.

30. 

Tang D, Lotze MT, Kang R and Zeh HJ: Apoptosis promotes early tumorigenesis. Oncogene. 30:1851–1854. 2011. View Article : Google Scholar : PubMed/NCBI

31. 

Cory S and Adams JM: The Bcl2 family: regulators of the cellular life-or-death switch. Nat Rev Cancer. 2:647–656. 2002. View Article : Google Scholar : PubMed/NCBI

32. 

Tait SW and Green DR: Mitochondria and cell death: outer membrane permeabilization and beyond. Nat Rev Mol Cell Biol. 11:621–632. 2010. View Article : Google Scholar : PubMed/NCBI

33. 

Shimizu S, Narita M and Tsujimoto Y: Bcl-2 family proteins regulate the release of apoptogenic cytochrome c by the mitochondrial channel VDAC. Nature. 399:483–487. 1999. View Article : Google Scholar : PubMed/NCBI

34. 

Zhou ZX, Zhang JH, Zhang LJ, Huang XH and Liu ZJ: Expression of survivin mRNA and protein in mitomycin-treated hepatoma carcinoma Hepa1-6 cells. Nan Fang Yi Ke Da Xue Xue Bao. 28:230–232. 2008.(In Chinese).

35. 

Miyashita T and Reed JC: Tumor suppressor p53 is a direct transcriptional activator of the human bax gene. Cell. 80:293–299. 1995. View Article : Google Scholar : PubMed/NCBI

36. 

Oda E, Ohki R, Murasawa H, et al: Noxa, a BH3-only member of the Bcl-2 family and candidate mediator of p53-induced apoptosis. Science. 288:1053–1058. 2000. View Article : Google Scholar : PubMed/NCBI

37. 

Nakano K and Vousden KH: PUMA, a novel proapoptotic gene, is induced by p53. Mol Cell. 7:683–694. 2001. View Article : Google Scholar : PubMed/NCBI

38. 

Liu TX, Howlett NG, Deng M, et al: Knockdown of zebrafish Fancd2 causes developmental abnormalities via p53-dependent apoptosis. Dev Cell. 5:903–914. 2003. View Article : Google Scholar : PubMed/NCBI

39. 

Jiang YY, Wang HJ, Wang J, Tashiro S, Onodera S and Ikejima T: The protective effect of silibinin against mitomycin C-induced intrinsic apoptosis in human melanoma A375-S2 cells. J Pharmacol Sci. 111:137–146. 2009. View Article : Google Scholar : PubMed/NCBI

40. 

Raman M, Chen W and Cobb MH: Differential regulation and properties of MAPKs. Oncogene. 26:3100–3112. 2007. View Article : Google Scholar : PubMed/NCBI

41. 

Liu J and Lin A: Role of JNK activation in apoptosis: a double-edged sword. Cell Res. 15:36–42. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2014
Volume 45 Issue 1

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
Zhao L, Li Y, He M, Song Z, Lin S, Yu Z, Bai X, Wang E and Wei M: The Fanconi anemia pathway sensitizes to DNA alkylating agents by inducing JNK-p53-dependent mitochondrial apoptosis in breast cancer cells Retraction in /10.3892/ijo.2022.5343. Int J Oncol 45: 129-138, 2014
APA
Zhao, L., Li, Y., He, M., Song, Z., Lin, S., Yu, Z. ... Wei, M. (2014). The Fanconi anemia pathway sensitizes to DNA alkylating agents by inducing JNK-p53-dependent mitochondrial apoptosis in breast cancer cells Retraction in /10.3892/ijo.2022.5343. International Journal of Oncology, 45, 129-138. https://doi.org/10.3892/ijo.2014.2400
MLA
Zhao, L., Li, Y., He, M., Song, Z., Lin, S., Yu, Z., Bai, X., Wang, E., Wei, M."The Fanconi anemia pathway sensitizes to DNA alkylating agents by inducing JNK-p53-dependent mitochondrial apoptosis in breast cancer cells Retraction in /10.3892/ijo.2022.5343". International Journal of Oncology 45.1 (2014): 129-138.
Chicago
Zhao, L., Li, Y., He, M., Song, Z., Lin, S., Yu, Z., Bai, X., Wang, E., Wei, M."The Fanconi anemia pathway sensitizes to DNA alkylating agents by inducing JNK-p53-dependent mitochondrial apoptosis in breast cancer cells Retraction in /10.3892/ijo.2022.5343". International Journal of Oncology 45, no. 1 (2014): 129-138. https://doi.org/10.3892/ijo.2014.2400