Abstract Pleurocidin-family cationic antimicrobial peptide NRC-03 exhibits potent and selective cytotoxicity towards cancer cells. However, the anticancer effect of NRC-03 in oral squamous cell carcinoma (OSCC) and the molecular mechanism of NRC-03 induced cancer cell death is still unclear. This study focused to investigate mitochondrial oxidative stress-mediated altered mitochondrial function involved in NRC-03-induced apoptosis of OSCC cells. NRC-03 entered the OSCC cells more easily than that of normal cells and bound to mitochondria as well as the nucleus, causing cell membrane blebbing, mitochondria swelling, and DNA fragmentation. NRC-03 induced high oxygen consumption, reactive oxygen species (ROS) release, mitochondrial dysfunction, and apoptosis in OSCC cells. Non-specific antioxidant N-acetyl-l-cysteine (NAC), or mitochondria-specific antioxidant mitoquinone (MitoQ) alleviated NRC-03-induced apoptosis and mitochondrial dysfunction indicated that NRC-03 exerts a cytotoxic effect in cancer cells via inducing cellular and mitochondrial oxidative stress. Moreover, the expression of cyclophilin D (CypD), the key component of mitochondrial permeability transition pore (mPTP), was upregulated in NRC-03-treated cancer cells. Blockade of CypD by siRNA-mediated depletion or pharmacological inhibitor cyclosporine A (CsA) significantly suppressed NRC-03-induced mitochondrial oxidative stress, mitochondrial dysfunction, and apoptosis. NRC-03 also activated MAPK/ERK and NF-κB pathways. Importantly, intratumoral administration of NRC-03 inhibited the growth of CAL-27 cells-derived tumors on xenografted animal models. Taken together, our study indicates that NRC-03 induces apoptosis in OSCC cells via the CypD-mPTP axis mediated mitochondrial oxidative stress. Keywords: NRC-03, Oral squamous cell carcinoma, Cyclophilin D, Cell apoptosis, Oxidative stress Graphical abstract [41]Image 1 [42]Open in a new tab Highlights * • NRC-03 induces OSCC cell apoptosis and inhibits OSCC ectopic tumor growth in mice. * • NRC-03 binds to the plasma membrane via electrostatic interaction and enters into cells. * • NRC-03 binds to the mitochondrial membrane, enters the nucleus, induces mtROS. * • NRC-03 upregulates CypD expression that triggers mPTP opening and caspase cascade. * • NRC-03 causes mitochondrial dysfunction and DNA fragmentation to induce apoptosis. 1. Introduction Oral squamous cell carcinoma (OSCC) is the most common oral malignancy and represents around 90% of all cancers in the oral cavity [[43]1], causing approximately 300,000 cases and 145,000 deaths worldwide annually [[44]2,[45]3]. OSCC in the initial stages shows an asymptomatic erytholeukoplastic lesion mainly on the tongue, mouth floor, buccal mucosa, gingiva, and lips [[46]4]. In advanced stages of OSCC, the lesions may develop into ulcers and lumps with irregular and poorly defined margins, causing a series of severe symptoms, such as severe pain, bleeding, problems in breathing, and difficulty in speech [[47][5], [48][6], [49][7]]. Because early carcinomas are asymptomatic, most OSCC cases are usually diagnosed in advanced stages [[50]8]. Moreover, OSCC exhibits high aggression, rapid progression, and early relapse, thus causing high mortality with a five-year survival rate of less than 50% [[51]9]. Chemotherapy is a critical treatment option in a multimodality therapeutic approach to treat locally advanced tumors, nonresectable tumors, metastatic tumors, and palliative chemotherapy [[52][10], [53][11], [54][12]]. Cisplatin, carboplatin, 5-fluorouracil, paclitaxel, and docetaxel are the commonly used first-line chemotherapeutic drugs to treat OSCC with a mechanism of damaging cell replication machinery of rapidly dividing cells [[55]6,[56]13]. However, apart from cancer cells, these drugs also damage the healthy cells with a high division rate from bone marrow, hair follicles, and gastrointestinal tract, which leads to a series of severe side effects, such as myelosuppression, alopecia, rashes, and vomiting [[57]14,[58]15]. To approach this problem, continuous efforts have been taken to develop cancer-targeting chemotherapeutic drugs [[59]16]. NRC-03, a 26-residue pleurocidin-like cationic antimicrobial peptides (CAPs), is derived from skin mucous secretions of winter flounder, and has promising cancer-targeting potential [[60]17]. The cationic property allows NRC-03 to specifically target negatively charged cancer cells, thereby causing membrane disturbance and cell death. Recent studies have shown that NRC-03 selectively kills human breast cancer, multiple myeloma, and leukemia cells, but is less toxic to normal cells, such as human umbilical vein endothelial cells, dermal fibroblasts, and erythrocytes [[61][18], [62][19], [63][20], [64][21]]. However, the anticancer potential of NRC-03 towards OSCC has not been investigated yet. Furthermore, the mode and molecular mechanisms of NRC-03-induced cancer cell death remain largely unknown. One major subcellular target of CAPs is mitochondria which play an important role in tumor initiation and progression through ATP production, catabolic and anabolic metabolism, generation of ROS, and apoptosis [[65]22]. Certain CAPs can interact with mitochondria directly or indirectly to trigger ROS generation, causing mitochondrial dysfunction and cancer cell death [[66][23], [67][24], [68][25], [69][26]]. In breast cancer cells, NRC-03 has been shown to co-localize with mitochondria, decrease mitochondrial membrane potential and cause the release of cytochrome c [[70]18]. However, the underlying molecular mechanisms remain unclear. mPTP is a reversible mitochondrial channel, which plays an important physiological role in maintaining mitochondrial homeostasis by the timely discharge of ROS and Ca^2+ from mitochondria [[71]27]. In pathological conditions, such as oxidative stress, continuous mPTP openings cause a burst release of ROS, resulting in mitochondrial dysfunction [[72]28,[73]29], which forms a vicious circle leading to cell death [[74]30]. CypD is a critical regulator for mPTP opening [[75]31]. CypD-dependent mPTP opening has been shown to play a key role in ROS-induced mitochondrial dysfunction and cell death [[76]31,[77]32]. However, the role of CypD-dependent mitochondrial dysfunction in NRC-03-induced cancer cell death is still unclear. In this study, we aimed to investigate the anti-OSCC efficacy of NRC-03 and its underlying mechanisms. We first assessed the cytotoxicity, apoptosis, and DNA damage potential of NRC-03 in two OSCC cell lines (CAL-27 and SCC-9) and normal human oral keratinocytes (HOK). We further investigated the OSCC growth inhibition potential of NRC-03 in nude mice ectopic tumor model. Thereafter, we monitored the interaction between NRC-03 and OSCC cells and evaluated the role of mitochondrial dysfunctions in NRC-03-induced apoptosis of OSCC cells. RNA sequencing was adopted to sort out the involved signaling pathways. Finally, we proved the critical role of CypD-mPTP openings in the anti-OSCC effect of NRC-03. 2. Materials and methods 2.1. Peptide synthesis NRC-03 (GRRKRKWLRRIGKGVKIIGGAALDHL-NH[2]) and tetramethylrhodamine (TRITC)-labeled NRC-03 were synthesized by Top-peptide Co., Ltd. (Shanghai, China) via Fmoc solid-phase peptide synthesis. The purity of the peptide was over 95%. Molecular weight (MW): 2953.4, net charge: +9.5, and isoelectric point (pI):12.67 were the basic biochemical properties of NRC-03 ^19. Lyophilized peptides were reconstituted in serum-free Dulbecco's Modified Eagle Medium/Nutrient Mixture F-12 (DMEM/F-12). All experiments were conducted in a medium containing 2.5% FBS to limit peptide degradation by serum proteases. 2.2. Reagents Cell culture medium and supplements were purchased from GIBCO BRL (Gaithersburg, MD, USA). Anti-β-actin antibody (#4970) and anti-rabbit secondary antibody (#7074) were obtained from Cell Signaling Technology (MA, USA). Anti-CypD antibody (ab110324) and Hydrogen peroxide assay kit - (Fluorometric-Near Infrared) (ab138886) were from Abcam (MA, USA). Anti-mouse secondary antibody (sc-516102) was purchased from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA). MitoSOX Red Mitochondrial Superoxide Indicator, MitoTracker Green FM, and LysoTracker Green DND-26 were from Yeasen biotechnologies Co., Ltd. (Shanghai, China). Terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) kit, Caspase-3 activity assay kit, Caspase-8 activity assay kit, Bradford protein assay kit, Adenosine triphosphate (ATP) assay kit, Hoechst 33342 staining kit, NAC, phenylmethanesulfonyl fluoride (PMSF), and RIPA buffer were from Beyotime Institute of Biotechnology (Shanghai, China). Z-VAD-FMK and cyclosporine A (CsA) were purchased from MedChemExpress (Princeton, NJ, United States). Mitoquinone (MitoQ) was from Cayman Chemical Company (Ann Arbor, MI, USA). Oxygen consumption rate (OCR) assay kit, 2′,7′-dichlorodihydrofluorescein diacetate (DCFH-DA), annexin V-FITC/propidium iodide (Annexin V-PI) staining kit, mitochondrial membrane potential assay kit with JC-1, and BCA protein assay kit were purchased from BestBio biotechnologies Co., Ltd. (Shanghai, China). Lipofectamine 3000 transfection reagents were from Invitrogen (Carlsbad, CA, USA). 2.3. Cell lines and cell culture OSCC cell lines CAL-27 and SCC-9 were purchased from ATCC and cultured in DMEM/F-12. HOK were obtained from ScienCell and cultured in Oral Keratinocyte Medium. The medium was supplemented with 10% fetal bovine serum (FBS) and 1% penicillin-streptomycin in 5% CO[2] at 37 °C in a humidified incubator. 2.4. Cell treatment Test compounds were prepared as stock solutions and diluted to the desired final concentrations immediately before use. The final concentrations of the compounds were as follows: NRC-03 (45 μg/ml), Z-VAD-FMK (50 μM), NAC (5 mM), MitoQ (1 μM), and CsA (2 μM). Cells were treated with or without NRC-03 and the indicated test compounds according to the experiment protocol. 2.5. Cell viability assay In vitro cytotoxicity was determined by the Cell Counting Kit-8 (Dojindo Corp.) assay. Cells were plated in 96-well plates (1 × 10^4 cells/well) and exposed to NRC-03 in the absence or presence of other test compounds. The cells were subsequently incubated for 3 h at 37 °C and the absorbance was measured at 450 nm using Multiskan™ FC Microplate Photometer (Thermo Scientific). 2.6. Colocalization analysis A confocal laser scanning microscopy (CLSM, Leica TCS SP8) was performed to observe the subcellular localization of NRC-03. CAL-27 cells were seeded confocal dishes at a density of 2 × 10^5 cells/well. After incubation for 24 h, the cells were treated with TRITC-labeled NRC-03 at predetermined intervals, cells were washed twice with cold PBS and then incubated with Mitotracker green (excitation: 488 nm, emission: 530 nm), LysoTracker Green (excitation: 504 nm, emission: 511 nm) or Hoechst 33342 (excitation: 350 nm, emission: 405 nm) according to the manufacturer's instructions, respectively. 2.7. Transmission electron microscope (TEM) analysis CAL-27 cells were fixed overnight in 2.5% glutaraldehyde in phosphate buffer (0.1 M, pH7.0). After post-fixation with OsO[4] in phosphate buffer (0.1 M, pH7.0), cells were dehydrated with a graded series of ethanol and embedded in resin. Ultra-thin (70–90 nm) sections were cut, stained by uranyl acetate and alkaline lead citrate, and observed in TEM (Hitachi H-7650). 2.8. Small interfering RNA (siRNA) transfection CypD siRNA targeting human peptidylprolyl isomerase F (PPIF) and negative control (NC) were transfected with Lipofectamine 3000 in CAL-27 cells according to the manufacturer's instructions. The sequence of siRNA-CypD is GACGAGAACTTTACACTGA. 2.9. Measurement of apoptosis by flow cytometry and TUNEL assay CAL-27 cells were cultured in 60-mm dishes and treated with or without other test compounds for 1 h prior to NRC-03 treatment for 4 h, then trypsinized, washed with PBS, and centrifuged at 1000 rpm for 5 min. Then, cells were resuspended in 500 μl binding buffer and stained with Annexin V-FITC and PI according to the protocol. The cells were incubated in the dark at room temperature for 15 min. Finally, the percentage of apoptotic cells and necrotic cells were assessed by flow cytometer analysis (FACS Aria III Cell Sorter, BD, United States). TUNEL staining was carried out to identify the rate of apoptotic cells. For the assay, cells inoculated on confocal dishes were fixed in 4% paraformaldehyde in PBS and permeabilized with 0.2% Triton X-100 in citrate buffer. Samples were incubated with TUNEL reaction mixture at 37 °C for 1 h, counterstained with Hoechst 33342, and observed with CLSM. The percentage of apoptotic cells was estimated by counting a total of 300 cells from random fields. 2.10. Caspase activity assay CAL-27 cells were pre-treated with or without Z-VAD-FMK for 1 h and then co-cultured with NRC-03 for 4 h. Caspase-3 and caspase-8 activity were evaluated according to the manufacturer's instructions. Briefly, the cell lysate was added to the 96-well plates and incubated with 2 mM of the Ac-DEVD-pNA (caspase-3 substrate) or Ac-IETD-pNA (caspase-8 substrate) at 37 °C overnight. The absorbance was read at 405 nm in Multiskan™ FC Microplate Photometer (Thermo Scientific). Protein levels in the cell lysate were measured using a Bradford protein assay kit. The results were expressed as active units of caspase/μg protein. Relative caspase activity was expressed as a fold increase over the control. 2.11. Oxygen consumption rate evaluation The oxygen consumption rate (OCR) was evaluated using an oxygen consumption rate assay kit. Briefly, CAL-27 cells were cultured in a 96-well plate (8 × 10^4 cells/well) with a clear bottom and black sides for 24 h. Next, 100 μl of medium mixed with different concentrations of NRC-03 and 5 μl of oxygen fluorescent probe was added to each well. Thereafter, blocking buffer (2 drops/well) was immediately added to each well to prevent external oxygen generation. After that, the plate was read with a fluorescent microplate reader (Model Infinite 200 Pro, Tecan) at 37 °C (1 read per 3 min, Ex 455/Em 603). Since the fluorescence of this oxygen probe can be quenched by O[2], the value of the fluorescence signal was inversely proportional to the amount of O[2]in each well. OCR was calculated based on the changes of fluorescence signal over 2 h as follows: OCR (%) = (final fluorescence in NRC-03-treated cells−initial fluorescence in NRC-03-treated cells)/(final fluorescence in control cells−initial fluorescence in control cells) × 100%. 2.12. Measurement of cellular oxidative stress The determination of intracellular oxidant stress was based on the oxidation of DCFH-DA. Briefly, CAL-27 cells were seeded in 60 mm dishes and treated with NRC-03 in the absence or presence of other test compounds for the indicated time, and then were incubated with redox-sensitive dye DCFH-DA at 37 °C for 30 min and analyzed by a flow cytometer. For determining mitochondrial ROS production, CAL-27 cells were cultured in confocal dishes and treated with or without other test compounds for 1 h prior to NRC-03 for 4 h. The cells were stained with 100 nM Mitotracker green for 30 min, and then with 2 μM MitoSOX Red (excitation: 510 nm, emission: 580 nm) for 15 min at 37 °C and visualized with CLSM. The ratio of fluorescence intensities was determined by ImageJ software. For determining hydrogen peroxide (H[2]O[2]) production, CAL-27 cells were cultured in a 96-well plate (8 × 10^4 cells/well) with a clear bottom and black sides for 24 h. The cells were treated with or without other test compounds for 1 h prior to NRC-03 treatment for 4 h. The quantification of H[2]O[2] production was assessed by AbIR Peroxidase Indicator using a Hydrogen peroxide assay kit – (Fluorometric-Near Infrared) complemented with a fluorescence plate reader (Model Infinite 200 Pro, Tecan) at room temperature (Ex 640/Em 680). 2.13. Mitochondrial membrane potential assay CAL-27 cells were plated in 60 mm dishes and grown overnight. Cells were then treated for 2 h with NRC-03 at the indicated concentrations or vehicle control in the absence or presence of other test compounds. Cells were then stained with 1 μM JC-1 dye for 20 min at 37 °C, washed, and assessed via flow cytometer analysis. 2.14. Measurement of cellular ATP level For the measurement of ATP level, whole-cell extracts were lysed in the lysis buffer provided in the ATP assay kit. After centrifugation at 12,000×g for 5 min at 4 °C, the supernatants were transferred to a new 1.5-ml tube for ATP analysis. The luminescence from a 100 μl sample was assayed in a luminometer (Model Infinite 200 Pro, Tecan) together with 100 μl of ATP detection buffer. A standard curve of ATP concentrations (1 nM–1 μM) was prepared from a known amount. 2.15. Western blot analysis After the indicated treatments, cells were collected and lysed in cell lysis buffer containing a 1% protease inhibitor. Total protein concentrations were measured using a BCA protein assay kit. Proteins were separated by electrophoresis and transferred to a polyvinylidene difluoride (PVDF) membrane. The membranes were blocked with blocking buffer and then incubated overnight at 4 °C with the primary antibody, followed by incubation with the secondary antibody at room temperature. Protein bands were visualized with an enhanced chemiluminescence substrate, detected using the Molecular Imager Gel Doc XR + imaging system (Bio-Rad, United States), and quantified with Quantity One Software. 2.16. RNA sequencing The total RNA of CAL-27 cells treated for 4 h with or without NRC-03 was isolated using an RNeasy mini kit (Qiagen, Germany). Paired-end libraries were synthesized by using the TruSeq RNA Sample Preparation Kit (Illumina, USA) following TruSeq RNA Sample Preparation Guide. Purified libraries were quantified by Qubit 2.0 Fluorometer (Life Technologies, USA) and validated by Agilent 2100 bioanalyzer (Agilent Technologies, USA) to confirm the insert size and calculate the mole concentration. A cluster was generated by cBot with the library diluted to 10 pM and then sequenced on the Illumina NovaSeq 6000 (Illumina, USA) by Promegen Biotechnology Co., Ltd, Guangzhou, China. The reads were aligned with Hisat2 (v 2.1.0) to GRCm38 with default parameters [[78]33]. The output SAM (sequencing alignment/map) files were converted to BAM (binary alignment/map) files and sorted using SAMtools (version 1.3.1) [[79]34]. Gene abundance was expressed as fragments per kilobase of exon per million reads mapped (FPKM). StringTie software was used to count the fragment within each gene, and the TMM algorithm was used for normalization [[80]35]. Differential expression analysis for mRNA was performed using R package edgeR. Differentially expressed RNAs with |log2(FC)| value > 1 and q value < 0.05, considered as significantly modulated, were retained for further analysis. 2.17. RT-qPCR analysis To further investigate the effect of NRC-03 on the apoptosis and mitochondrial oxidative stress-related gene expression of OSCC cells, CAL-27 cells treated for 4 h with or without NRC-03 were collected and total RNAs were extracted for RT-qPCR. Following the manufacturer's instructions, complementary DNA was synthesized from 500 ng of total RNA using a Takara PrimeScript™ RT Master Mix in T100 Thermal Cycler (Bio-Rad, United States). The cDNA was assayed using TaKaRa TB Green™ Premix Ex Taq™ in the CFX96 Real-Time system (Bio-Rad, United States). GADPH was used as a reference gene. The primers used for RT-qPCR are listed in [81]Supplementary Table S1. 2.18. Animal xenograft model Female BALB/c nu/nu mice (5-weeks-old) were purchased from the Center for Experimental Animals, Southern Medical University, Guangzhou, China. All animal studies were conducted in accordance with the guidelines of the National Regulation of China for Care and Use of Laboratory Animals (South China Normal University, Guangzhou, China). Southern Medical University Experimental Animal Ethics Committee approved all animal care and study protocols (L2018153). Mice were subcutaneously injected in the right flank with 4 × 10^6 CAL-27 cells in 0.1 ml sterile PBS. At 2–4 weeks post-inoculation, tumors grew to an average volume of 100 mm^3 and the CAL-27 tumor-bearing mice were randomly distributed into two groups (n = 6 per group) and intratumor injected with 50 μl PBS vehicle or 125 μg NRC-03 in 50 μl PBS. Treatments were repeated every other day for 15 days. Mice were monitored daily for tumor growth (using digital calipers), cachexia, and weight loss. Tumor volumes were calculated using the elliptical formula: 1/2 (length × width^2). Some tumors were frozen in liquid nitrogen for western blot and the other part tumors and the organs including the heart, liver, spleen, lungs, and kidneys were formalin-fixed and processed for histological analysis. Hematoxylin and eosin (H&E) staining, Ki-67 staining, and TUNEL staining were performed to detect proliferating cells and apoptotic cells respectively. 2.19. Statistical analysis All data were representative results from at least three independent experiments and mean ± SD. Statistical analysis was performed using one-way analysis of variance (ANOVA) and unpaired Student's t-test by Graphpad Prism 6.0. p < 0.05 was considered statistically significant. 3. Results 3.1. NRC-03 induced apoptosis in oral squamous cell carcinoma cells CCK-8 assay showed that NRC-03 in the concentration range of 15–75 μg/ml inhibited the viability of CAL-27 and SCC-9 cells in a time- and dose-dependent manner ([82]Fig. 1a). Cytotoxicity of all the tested concentrations of NRC-03 toward HOK cell was minimum compared with CAL-27 or SCC-9 cells ([83]Fig. 1a). Annexin/propidium iodide staining assay is an indicator of alterations in cell membrane permeability and apoptosis. Annexin/propidium iodide staining revealed a dose-dependent increase in apoptosis in NRC-03-treated CAL-27 and SCC-9 cells. NRC-03 (30–60 μg/ml) significantly enhanced the rate of apoptosis in CAL-27 and SCC-9 cells by about 2.8–4.4-fold. In contrast, the enhancement magnitude of HOK apoptosis was much lower (about 1.2–2.3-fold) ([84]Fig. 1b and 1c). The pro-apoptotic effects of NRC-03 were further verified by TUNEL staining as an indicator of DNA damage ([85]Fig. 1d). The percentage of TUNEL positive cells significantly increased in OSCC groups, i.e., 31.61% ± 7.98% in CAL-27 and 28.25% ± 6.64% in SCC-9 cells, respectively ([86]Fig. 1e), while TUNEL staining positivity rate was only 2.37% ± 1.90% in HOK cells after treatment with 45 μg/ml NRC-03. In contrast, it is important to note that NRC-03 did not substantially affect the viability or induce apoptosis in the normal HOK cells ([87]Fig. 1a, 1d, and 1e). Caspase-3 upregulates during apoptosis-mediated cell death. Caspase-8 is a crucial initiator in the death receptor-mediated apoptotic pathway. Next, we detected the caspase-3 and caspase-8 activity in NRC-03-treated CAL-27 cells. NRC-03 treatment didn't activate the caspase-8 activity but caused a remarkable increase in caspase-3 activity of OSCC cells ([88]Fig. 1f and 1g). The median lethal dose of NRC-03 treatment was 45 μg/ml on 4 h treatment. NRC-03 at a concentration of 45 μg/ml significantly inhibited the cell viability of CAL-27 and SCC-9 cells on 24 h treatment, but the effect on normal HOK viability was not that prominent ([89]Fig. 1a). Based on these findings, 45 μg/ml of NRC-03 was chosen as the optimal dose to treat the cells in subsequent experiments. Fig. 1. [90]Fig. 1 [91]Open in a new tab NRC-03 induced apoptosis in oral squamous cell carcinoma cells. (a) Cell viability of NRC-03-treated OSCC cells CAL-27 and SCC-9, and HOK analyzed by CCK-8 assay (n = 4). NRC-03 treatment for 4 h induced apoptosis in OSCC cells. (b) Detection of phosphatidylserine exposure by Annexin-V FITC/PI staining and analysis by flow cytometry. Cells in Q1, Q2, Q3, and Q4 respectively represent cell debris (Annexin-V^-/PI^+), cells in late apoptosis (Annexin-V^+/PI^+), cells in early apoptosis (Annexin-V^+/PI^−), and healthy cells (Annexin-V^-/PI^−). (c) The quantification of apoptotic cells (Annexin-V^+) (n = 3). (d) Representative immunofluorescence images of TUNEL staining of cells treated with 45 μg/ml NRC-03. (e) Quantification of TUNEL positive cells (n = 6). (f) Caspase-3 activity in NRC-03-treated CAL27 cells (n = 3). (g) Caspase-8 activity in NRC-03-treated CAL-27 cells (n = 3). Data are presented as mean ± SD. Significant difference compared with the control group, *p < 0.05, **p < 0.01, ***p < 0.001, and ****p < 0.0001. To further confirm the NRC-03-mediated apoptotic pathway in OSCC cells, we pretreated CAL-27 cells with Z-VAD-FMK, an irreversible pan-caspase inhibitor, along with NRC-03. Z-VAD-FMK treatment counteracted the NRC-03-induced cytotoxicity ([92]Fig. 2a) and apoptosis ([93]Fig. 2b–e). Furthermore, Z-VAD-FMK nullified the promoting effects of NRC-03 on the caspase-3 activity in CAL-27 cells ([94]Fig. 2f). Collectively, these data verified that NRC-03 induced OSCC cell death mainly via the intrinsic/mitochondria-mediated apoptosis pathway. Fig. 2. [95]Fig. 2 [96]Open in a new tab Caspase inhibition attenuated NRC-03-induced apoptosis in CAL-27 cells. CAL-27 cells were cultured in the presence of 45 μg/ml NRC-03 for 4 h with or without caspase inhibitor Z-VAD-FMK (50 μM). (a) Cell viability determined by CCK-8 (n = 4). (b) Flow cytometry analysis of annexin-V positive cells. (c) Quantification of annexin-V positive cells (n = 3). (d, e) TUNEL immunofluorescence staining and quantification of TUNEL positive cells (n = 5). (f) Caspase-3 activity (n = 3). Data are presented as mean ± SD. Significant difference between the groups, **p < 0.01, ***p < 0.001, and ****p < 0.0001. 3.2. NRC-03 inhibits tumor growth in a xenograft model NRC-03 at a dose of 125 μg/animal inhibited the growth of CAL-27-derived tumors in a subcutaneous ectopic tumor model in nude mice ([97]Fig. 3). Importantly, NRC-03 did not cause observable damage to vital organs ([98]Fig. S1a) or bodyweight ([99]Fig. S1b). By day 9, compared with the vehicle control, NRC-03 treatment induced a significant decrease in tumor growth (p < 0.01), which persisted throughout the 15-day study period ([100]Fig. 3b). The final average tumor volumes in the control and 125 μg NRC-03 treated groups were 287.18 ± 66.73 mm^3 and 103.17 ± 48.16 mm^3, respectively ([101]Fig. 3c). Moreover, tumor tissues from the NRC-03-treated group showed a reduced cellular density and proliferation rate ([102]Fig. 3d and e) and increased apoptosis rate ([103]Fig. 3d and f) compared with the control group. These results in vivo are consistent with the results from the in-vitro studies, which highlighted that NRC-03 could inhibit OSCC growth via inducing apoptosis in cancer cells. Fig. 3. [104]Fig. 3 [105]Open in a new tab NRC-03 inhibited tumor growth in the ectopic tumor model of OSCC. CAL-27-derived xenografts were treated with 125 μg NRC-03 every other day for 15 days. (a) Gross images of representative tumor tissues on day 15. (b) Tumor volume at different time points (n = 6). (c) Final tumor volume on day 15 (n = 6). (d) Representative microscopic images of tumor tissue sections showing tumor morphology (H&E staining), proliferation (Ki-67 immunohistochemistry), and apoptosis (TUNEL staining). Quantification of Ki-67-positive cells (e) (n = 6), and apoptotic cells (f) (n = 6). Data are presented as mean ± SD. Significant difference compared with the respective control group, ***p < 0.001, and ****p < 0.0001. 3.3. NRC-03 damaged the cell membrane and specifically entered the cytoplasm and nucleus of CAL-27 cells To investigate whether NRC-03 entered the cytoplasm of OSCC cells, CAL-27 or HOK cells were treated with TRITC labeled NRC-03 for 1 h and monitored continuously by CLSM. CLSM revealed that TRITC labeled NRC-03 rapidly entered the cytoplasm of CAL-27 cells ([106]Fig. 4a). The dynamic process of NRC-03 entering the cell can be seen in the supplementary information (Supplementary Movie 1 and 2). A remarkably increased accumulation of NRC-03 was observed at 8–9 min in CAL-27 cells, which was 1.56-fold higher than that in HOK cells at the same time. With the prolongation of treatment time, the accumulation of NRC-03 gradually increased and stabilized at 30 min in CAL-27. However, the cellular accumulation of NRC-03 reached its maximum at 17–18 min in HOK and then gradually decreased. Moreover, the amount of NRC-03 in CAL-27 cells was 3.48-fold higher than in HOK cells at 60 min ([107]Fig. 4b). Importantly, NRC-03 localization in the nucleus of CAL-27 cells was higher than that in HOK cells. The cell surface of CAL-27 showed a higher accumulation of NRC-03 forming many membrane blebs as indicated with black arrows ([108]Fig. 4a). Moreover, some CAL-27 cells extruded a peptide-bound substance as indicated with white arrows ([109]Fig. 4a), suggesting that the peptides cause significant membrane damage to CAL-27 cells. Taken together, these results demonstrated that NRC-03 selectively targets OSCC cells, causes membrane blebbing, and localizes in the nucleus. Fig. 4. [110]Fig. 4 [111]Open in a new tab NRC-03 damaged the cell membrane of CAL-27 cells and entered the cytoplasm and nucleus. CLSM images of cells were treated with 45 μg/ml NRC-03 for 1 h and photographed by CLSM every 30 s. (a) Peptide and nucleus were visualized by using TRITC-NRC-03 (Red) and Hoechst 33342 (Blue), respectively. The white arrow indicates extruded peptide-bound substance from CAL-27 cells. The black arrow indicates membrane blebs on the cell surface of CAL-27. (b) Quantification of the mean fluorescence intensity of TRITC-NRC-03 at different time points. (For interpretation of the references to color in this figure legend, the