Next Article in Journal
Heart Uptake of [18F]Fluoro-4-Thia-Oleate in a Non-Alcoholic Fatty Liver Disease Mouse Model
Next Article in Special Issue
Propolis: A Detailed Insight of Its Anticancer Molecular Mechanisms
Previous Article in Journal
The Hidden Pandemic of COVID-19-Induced Organizing Pneumonia
Previous Article in Special Issue
Components with Anti-Diabetic Activity Isolated from the Leaves and Twigs of Glycosmis pentaphylla Collected in Vietnam
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Marine Sponge Aaptos suberitoides Extract Improves Antiproliferation and Apoptosis of Breast Cancer Cells without Cytotoxicity to Normal Cells In Vitro

1
Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
2
Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
3
School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
4
Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaoshiung Medical University, Kaohsiung 80708, Taiwan
5
Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
6
Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
7
Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Pharmaceuticals 2022, 15(12), 1575; https://doi.org/10.3390/ph15121575
Submission received: 29 November 2022 / Revised: 9 December 2022 / Accepted: 13 December 2022 / Published: 16 December 2022

Abstract

:
The anticancer effects and mechanisms of marine sponge Aaptos suberitoides were rarely assessed, especially for methanol extract of A. suberitoides (MEAS) to breast cancer cells. This study evaluated the differential suppression effects of proliferation by MEAS between breast cancer and normal cells. MEAS demonstrated more antiproliferation impact on breast cancer cells than normal cells, indicating oxidative stress-dependent preferential antiproliferation effects on breast cancer cells but not for normal cells. Several oxidative stress-associated responses were highly induced by MEAS in breast cancer cells but not normal cells, including the generations of cellular and mitochondrial oxidative stress as well as the depletion of mitochondrial membrane potential. MEAS downregulated cellular antioxidants such as glutathione, partly contributing to the upregulation of oxidative stress in breast cancer cells. This preferential oxidative stress generation is accompanied by more DNA damage (γH2AX and 8-hydroxy-2-deoxyguanosine) in breast cancer cells than in normal cells. N-acetylcysteine reverted these MEAS-triggered responses. In conclusion, MEAS is a potential natural product for treating breast cancer cells with the characteristics of preferential antiproliferation function without cytotoxicity to normal cells in vitro.

1. Introduction

Breast cancer accounts for 30% of female cancer and is the leading cause of women’s cancer death [1]. It increases by 0.5% per year. Three major subtypes characterize most breast cancer cells, i.e., estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) [2]. Some 10–15% of breast cancers belong to triple-negative breast cancer (TNBC), i.e., no ER/PR/HER2 [3], which is hard to cure using target therapy. Identifying more anticancer drugs against breast cancer cells is still necessary, particularly for TNBC.
Marine sponges are rich in diverse natural products [4,5,6,7,8] for curing cancer treatments [6]. For example, 70% aqueous ethanol extract of Grayella cyathophora inhibits the proliferation of colon and breast cancer cells [9]. Methanol extract of Crambe displays antiproliferation against pancreatic cancer cells [10]. Marine sponge Lipastrotethya sp. extract suppresses colon cancer cell proliferation [11]. Ethyl acetate extract from the marine sponge Stylissa carteri was reported to inhibit the proliferation of breast cancer cells [12]. Sponge-derived natural products such as hemimycalin C, D, E, and manzamine A show antiproliferation against colon cancer cells [13]. Accordingly, several extracts and bioactive components of marine sponges exhibit anticancer effects.
Aaptos suberitoides (A. suberitoides) is a marine sponge harvesting in Indonesian waters [14], and it was also found on Orchid Island, Taiwan, in the present study. Recently, the ethanol extract of A. suberitoides was reported to inhibit the proliferation and migration of breast cancer cells [15]. However, this study did not examine the detailed anticancer mechanism of ethanol extract of A. suberitoides.
The crude extract of natural products containing several bioactive compounds is expected to exhibit multi-targeting effects against cancer cells with low cytotoxicity to normal cells [16]. Sponges include diverse compounds [17]. Hence, sponge extracts have an improving impact on the antiproliferation of cancer cells. Accordingly, the anticancer effects of A. suberitoides crude extract warrant a detailed investigation of breast cancer cells.
The present investigation assesses the in vitro antiproliferation effects mechanisms of methanol extract of A. suberitoides (MEAS) to breast cancer cells.

2. Results

2.1. HPLC Analysis of MEAS and Aaptamine

In Figure 1A, the HPLC fingerprint profiles of MEAS (red line) and the main product of MEAS (aaptamine) (blue line) at 254 nm were shown. Aaptamine [18] (Figure 1B) was found to appear at 22.730 min, which overlapped the major peak of MEAS. The NMR spectrum of aaptamine is provided in Supplementary Figure S1. The linear equations (Y = 4 × 107 X − 653182, R2 = 0.9997) of aaptamine was deduced by the HPLC peak area in different concentration (Figure 1C). As a result, aaptamine accounts for 15.3% of MEAS.

2.2. Antiproliferation of MEAS-Treated Breast Cancer and Normal Cells

In 24 h MTS viability assay, MEAS reduced the cell viability of breast cancer cells (HCC1937, MDA-MB-231, MDA-MB-468, and MCF7) (Figure 2A). For comparison, MEAS showed high viability of normal cells (H184B5F5/M10; M10) [19,20,21] in tested concentrations of MEAS compared to breast cancer cells. These results revealed the preferential antiproliferation character of MEAS on breast cancer cells showing minor changes to normal cells.
MCF7 and HCC1937 cells with high sensitivity to MEAS were chosen to perform the following experiments. The ROS suppressor NAC mitigated the MEAS-caused inhibitory effects of proliferation against breast cancer cells (Figure 2B). Accordingly, oxidative stress is involved in the antiproliferation of MEAS.

2.3. Cell Cycle Status of MEAS-Treated Breast Cancer and Normal Cells

In 24 h 7-amino actinomycin D (7AAD) assay, the subG1 phase (%) of breast cancer cells (MCF7 and HCC1937) and normal cells (M10) [19,20,21] were deficient (Figure 3A). The G1 phase (%) was decreased, and the G2/M phase (%) was increased in breast cancer cells. In comparison, the changes in G1 and G2/M phase (%) in M10 cells were opposite to breast cancer cells.
Additionally, NAC mitigated the MEAS-caused G1-inducible and G2/M-suppressing effects against breast cancer cells at 12 h treatment for MCF7 cells and at 12 and 24 h for HCC1937 cells (Figure 3B). Accordingly, oxidative stress is involved in the cell cycle disturbance effects of MEAS on breast cancer cells.

2.4. Annexin V Status of MEAS-Treated Breast Cancer and Normal Cells

Annexin V/7ADD assay was adopted for validating apoptosis. MEAS increased the annexin V intensity (+) (%) of breast cancer cells (MCF7 and HCC1937) (Figure 4A). For comparison, MEAS showed high annexin V intensity (+) (%) of breast cancer cells in tested concentrations of MEAS compared to normal cells (M10). These results revealed the preferential apoptosis (annexin V) of MEAS on breast cancer cells showing minor changes to normal cells.
Additionally, NAC mitigated the MEAS-caused annexin V-detected apoptosis against breast cancer cells (Figure 4B). Accordingly, oxidative stress is involved in the apoptosis (annexin V) of MEAS.

2.5. Caspase Status of MEAS-Treated Breast Cancer and Normal Cells

Caspase signaling activations such as Caspases 3, 8, and 9 were used to assess apoptosis. MEAS increased the Caspases 3, 8, and 9 intensities (+) (%) of breast cancer cells (MCF7 and HCC1937) (Figure 5A,C,E). For comparison, MEAS showed high Cas 3, 8, and 9 intensities (+) (%) of breast cancer cells in tested concentrations of MEAS compared to normal cells. These results revealed the preferential apoptosis (Caspases 3, 8, and 9) of MEAS on breast cancer cells showing minor changes to normal cells (M10).
Additionally, NAC mitigated the MEAS-caused Caspases 3, 8, and 9-detected apoptosis against breast cancer cells (Figure 5B,D,F). Accordingly, oxidative stress is involved in the apoptosis (Caspases 3, 8, and 9) of MEAS.

2.6. Reactive Oxygen Species (ROS) and Mitochondrial Superoxide (MitoSOX) Status of MEAS-Treated Breast Cancer and Normal Cells

MEAS increased oxidative stress, such as ROS and MitoSOX intensities (+) (%) of breast cancer cells (MCF7 and HCC1937) (Figure 6A,C). For comparison, MEAS showed high ROS and MitoSOX intensities (+) (%) of breast cancer cells in tested concentrations of MEAS compared to normal cells. These results revealed the preferential oxidative stress (ROS and MitoSOX) of MEAS on breast cancer cells showing minor changes to normal cells (M10).
Additionally, NAC mitigated the MEAS-caused ROS and MitoSOX against breast cancer cells (Figure 6B,D). Accordingly, oxidative stress is involved in the oxidative stress (ROS and MitoSOX) of MEAS.

2.7. Mitochondrial Membrane Potential (MMP) Status of MEAS-Treated Breast Cancer and Normal Cells

MEAS increased the oxidative stress, such as MMP intensity (−) (%) of breast cancer cells (MCF7 and HCC1937) (Figure 7A). For comparison, MEAS showed high MMP intensity (−) (%) of breast cancer cells in tested concentrations of MEAS compared to normal cells. These results revealed the preferential oxidative stress (MMP depletion) of MEAS on breast cancer cells showing minor changes to normal cells (M10).
Additionally, NAC mitigated the MEAS-caused MMP depletion against breast cancer cells (Figure 7B). Accordingly, oxidative stress is involved in the oxidative stress (MMP depletion) of MEAS.

2.8. Glutathione (GSH) Status of MEAS-Treated Breast Cancer and Normal Cells

The contribution of cellular antioxidants such as GSH in elevating oxidative stress of breast cancer cells treated with MEAS was evaluated. MEAS increased the oxidative stress, such as GSH intensity (−) (%) of breast cancer cells (MCF7 and HCC1937) (Figure 8A). For comparison, MEAS showed high GSH intensity (−) (%) of breast cancer cells in tested concentrations of MEAS compared to normal cells. These results revealed the preferential oxidative stress (GSH depletion) of MEAS on breast cancer cells showing minor changes to normal cells (M10).
Additionally, NAC mitigated the MEAS-caused GSH depletion against breast cancer cells (Figure 8B). Accordingly, oxidative stress is involved in the oxidative stress (GSH depletion) of MEAS.

2.9. DNA Damages Status of MEAS-Treated Breast Cancer and Normal Cells

MEAS increased the DNA damage such as γH2AX and 8-hydroxy-2-Deoxyguanosine (8-OHdG) intensities (+) (%) of breast cancer cells (MCF7 and HCC1937) (Figure 9A and Figure 10A). For comparison, MEAS showed high γH2AX and 8-OHdG intensities (+) (%) of breast cancer cells in tested concentrations of MEAS compared to normal cells. These results revealed the preferential DNA damage (γH2AX and 8-OHdG) of MEAS on breast cancer cells showing minor changes to normal cells (M10).
Additionally, NAC mitigated the MEAS-caused γH2AX and 8-OHdG against breast cancer cells (Figure 9B and Figure 10B). Accordingly, oxidative stress is involved in the damage (γH2AX and 8-OHdG) of MEAS.

3. Discussion

Marine sponges are abundant in natural products for anticancer drug discovery [4,5,6,7,8,10,22,23,24]. The anti-breast cancer effects of ethanol extract of A. suberitoides were reported, such as antiproliferation and antimigration [15], but it did not provide a detailed investigation of the anticancer mechanism. The present study examined the antiproliferation effects and mechanism of methanol extract of A. suberitoides (MEAS) between breast cancer and normal cells.
Several marine sponge extracts showed anticancer effects. The ethanol extract of Aaptos suberitoides shows an IC50 value of 12.0 μg/mL to breast cancer cells (HCC-1954) at 72 h MTT assay [15]. 70% aqueous ethanol extracts of Grayella cyathophora and Negombata magnifica show IC50 values of 2.14 and 1.09 μg/mL to colon cancer cells (Coca-2) at 72 h MTT assay [9]. A 70% ethanol extract of Dysidea avara shows IC50 values of 11.51, 5.11, and 17.54 μg/mL to the breast (MCF7), cervical (HeLa), and colon (HCT116) cancer cells at 48 h MTT assay [25]. Notably, these studies did not consider the drug’s safety for normal cells. They did not investigate the cytotoxicity of normal cells.
In contrast, IC50 values of MEAS at 24 h MTS assay are 17.81, 19.19, 24.41, and 14.23 μg/mL in breast cancer cells (MCF7, HCC1937, MDA-MB-231, and MDA-MB-468, respectively). Both TNBC and non-TNBC cells were sensitive to MEAS. Moreover, the viability of normal cells (M10) [19,20,21] is higher than breast cancer cells following MEAS treatment, suggesting the preferential antiproliferation effects on breast cancer cells. This antiproliferation is modulated by oxidative stress as validated by NAC pretreatment (Figure 2B).
The oxidative stress involvement in MEAS treatment of breast cancer cells was validated by ROS and MitoSOX generation and MMP depletion (Figure 6 and Figure 7). These oxidative stresses were higher in breast cancer cells than in normal cells, indicating MEAS triggers the preferential induction of oxidative stress in breast cancer cells.
The redox status is controlled by the balance between cellular antioxidants and prooxidants [26]. When the levels of prooxidants are higher than that of oxidants, cells suffer from oxidative stress. The suppression of cellular antioxidants is one of the reasons for generating oxidative stress [27]. For example, alantolactone, an Inula helenium-derived natural product, causes oxidative stress by depleting GSH levels of glioblastoma cells [28]. The brown algae-derived fucoidan triggers GSH downregulation to induce oxidative stress in oral cancer cells [29]. Similarly, MEAS promoted GSH depletion in breast cancer cells to a greater extent than in normal cells (Figure 8). Moreover, this MEAS-induced GSH depletion was reversed by NAC pretreatment, a precursor for GSH biosynthesis. Consequently, MEAS stimulates the preferential induction of oxidative stress against breast cancer cells.
Moreover, several marine sponge extracts showed apoptosis-inducible function in cancer cells [25,30,31,32]. For example, ethanol extract of Dysidea avara shows apoptosis of cervical and leukemia cancer cells by annexin V detection [25]. The N-hexane extract of Hyrtios erectus causes subG1 accumulation and activates caspase 3 and caspase 9 in breast cancer cells [33]. For comparison, the subG1 population is few after MEAS treatments for breast cancer and normal cells (Figure 3). However, the subG1 accumulation is not an essential apoptotic response [34]. In some cases, no prominent subG1 peaks are observed in drug-induced apoptosis, depending on the exposure time of drug treatment. For the example of (-)-anonaine treatment to lung cancer cells, the subG1 peaks are very low at 24 and 48 h but high at 72 h [35]. Notably, MEAS triggered apoptosis (annexin V and caspase 3) (Figure 4 and Figure 5) and turned on both extrinsic and intrinsic caspases (caspases 8 and 9) (Figure 5). MEAS promoted more apoptosis in breast cancer cells than in normal cells. This character of preferential apoptosis may attribute to its impact on preferential oxidative stress. Additionally, the MEAS-induced oxidative stress in breast cancer cells promotes a greater extent of DNA damage, such as γH2AX and 8-OHdG, than in normal cells (Figure 9 and Figure 10).
Finally, the impacts of oxidative stress acting on the MEAS-induced cell cycle, oxidative stress, and DNA damage were validated by NAC pretreatment. Therefore, MEAS exerts an oxidative stress-associated mechanism for preferential antiproliferation against breast cancer cells in vitro.

4. Materials and Methods

4.1. Sample Collection and Identification

The animal material of Aaptos suberitoides was obtained from Orchid Island, Taitung County, Taiwan, in April 2011. The voucher specimen (OISP-4) was given, and the specimen was stored at the Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan. The sponge was identified by co-author Dr. Jui-Hsin Su using the DNA sequence and the sponge spicule morphology.

4.2. Sample Preparation

The animal material (2.3 kg) was extracted by ethanol three times, and the crude extract was partitioned by EtOAc and H2O to give an EtOAc soluble portion. This portion was further partitioned by H2O/methanol/hexanes (1:3:4). After removing the solvents, the methanol extract portion (5.8 g) of A. suberitoides (MEAS) was used for further chromatographic analysis and pharmacological experiments.

4.3. Isolation of Aaptamine

The MEAS was subjected to vacuum liquid chromatography (VLC) eluting with hexanes/CH2Cl2/MeOH (increasing polarity), and ten fractions (Fr. 1 to Fr. 10) were obtained. Fr. 5 (610.6 mg) was purified by a silica gel open column eluting with CH2Cl2/MeOH (10:1 to 0:1) to afford three subfractions (Fr. 5-1 to Fr. 5-3). Fr. 5-2 (577.2 mg) was further purified by CC on LH-20 eluting with 100% MeOH to give aaptamine (492.3 mg) [36].

4.4. HPLC Analysis of A. suberitoides

Separation by high-pressure liquid chromatography (HPLC) was accomplished on a Shimadzu LC-40D solvent delivery module equipped with the Phenomenex Luna 5 µ C18(2) 100A analytical column. The Shimadzu SPD-M40A photodiode array detector and CTO-40S column oven were selected for analysis. The chromatography methods were listed below: Solution A: 0.1% trifluoroacetic acid(aq); solution B: MeCN; flow rate: 1.0 mL/min; 0 min: 1% solution B, 0–35 min: 1% to 40% solution B, 35–40 min: 40%-100% solution B.

4.5. Cell Cultures and Inhibitors

ATCC breast cancer cell lines, such as MCF7 (Luminal A type), and several TNBC cell lines, such as MDA-MB-468 [37], MDA-MB-231, and HCC1937 [38], were included and maintained in DMEM/F12 (3:2) (Gibco, Grand Island, NY, USA) with 10% fetal bovine serum and P/S antibiotics. A non-malignant normal breast epithelial cell line (H184B5F5/M10) [19,20,21] was used as control, which was purchased from Bioresource Collection and Research Center (Hsinchu, Taiwan) and maintained in alpha medium with 10% bovine serum (Gibco, Grand Island, NY) and P/S antibiotics. N-acetylcysteine (NAC) (Sigma-Aldrich, St. Louis, MO, USA) [39,40,41,42] is a glutathione precursor that was commonly used as ROS inhibitor. Under 10 mM for 1 h pretreatment, cells were post-treated with MEAS as indicated in figure legends.

4.6. Cell Viability

Cell viability was detected by Promega’s MTS kit (Madison, WI, USA). Cells were seeded and incubated overnight. Subsequently, cells were used for drug treatment. Finally, MTS reagents were mixed with medium for a 1 h reaction and read by an ELISA reader at 490 nm [29].

4.7. Cell Cycle

Fixed cells were incubated with 1 μg/mL of 7AAD (Biotium; Hayward, CA, USA) [43,44]. The DNA content of 7AAD-positive cells was then detected by the Accuri C6 flow cytometer (BD Biosciences, Franklin Lakes, NJ, USA).

4.8. Apoptosis

Annexin V/7AAD double-staining [45,46] and caspases 3, 8, and 9 activation assays [47] were used to detect apoptosis status. Annexin V-FITC/7AAD (1:1000/1 μg/mL) [48] (Strong Biotech; Taipei, Taiwan) was added to cells for 1 h incubation, and their fluorescence intensities were detected by Accuri C6 flow cytometer. The cell spots for annexin V (+)/7AAD (+ or −) intensity were counted as apoptosis (+) cells.
Moreover, the activities of caspases 3, 8, and 9 were detected by OncoImmunin’s specific peptides (Gaithersburg, MD, USA) according to the manufacturer’s instructions [47,49]. When caspases 3, 8, and 9 are activated, these peptides generate fluorescence and are detected by the Accuri C6 flow cytometer.

4.9. Oxidative Stress

Several oxidative stress-related indicators, such as ROS, MitoSOX, MMP, and GSH, were tested. They were respectively detected by 2′,7′-dichlorodihydrofluorescein diacetate (H2DCFDA) (Sigma-Aldrich) [41,50], MitoSOX™ Red [51] (Thermo Fisher Scientific, Carlsbad, CA), DiOC2(3) (Invitrogen, San Diego, CA, USA) [52], and 5-chloromethylfluorescein diacetate (CMF-DA) (Thermo Fisher Scientific, Carlsbad, CA, USA) (5 μM, 20 min) [29], according to manufacturer’s instructions. In response to these oxidative stresses, the Accuri C6 cytometer detected the generating fluorescence of ROS, MitoSOX, and MMP, while the Guava easyCyte flow cytometer (Luminex, TX, USA) detected the generating fluorescence of GSH.

4.10. DNA Damages

Several DNA damage indicators, such as γH2AX and 8-OHdG, were detected. Cells need to process with 75% ethanol fixation before antibody reactions. Except for the extra step for 7AAD (5 μg/mL, 30 min) incubation, γH2AX and 8-OHdG [29] were detected by specific antibodies such as γH2AX [53,54] (Santa Cruz Biotechnology; Santa Cruz, CA, USA)/Alexa Fluor 488-secondary antibody (Cell Signaling Technology, Danvers, MA, USA) and FITC-8-OHdG antibody (Santa Cruz Biotechnology), respectively. Finally, these fluorescence intensities were detected by the Accuri C6 flow cytometer.

4.11. Statistical Analysis

JMP software (SAS Institute Inc., Cary, NC, USA) was used to determine significance. It provides connecting letters for each treatment. When the connecting letters were not overlapped, treatments differed significantly. Examples for determining significance were given in the figure legend.

5. Conclusions

The present study validated the oxidative stress-dependent antiproliferation effects of MEAS in breast cancer cells. MEAS promotes more antiproliferation of breast cancer cells than normal cells. The anti-breast cancer effects of MEAS exert dysregulation of the cell cycle and oxidative stress in breast cancer cells. This abnormal oxidative stress sequentially induces higher apoptosis and DNA damage in breast cancer cells than in normal cells. Therefore, MEAS is a potential marine natural product for breast cancer treatment.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ph15121575/s1, Figure S1: 1H and 13C NMR spectra of aaptamine.

Author Contributions

Conceptualization, J.-P.S., M.-Y.L., Y.-B.C. and H.-W.C.; data curation, M.-Y.L.; formal analysis, M.-Y.L.; methodology, J.-P.S., J.-Y.T., H.H., Z.-Y.L., J.-H.S. and M.-F.H.; supervision, Y.-B.C. and H.-W.C.; writing—original draft, J.-P.S. and H.-W.C.; writing—review and editing, Y.-B.C. and H.-W.C. All authors have read and agreed to the published version of the manuscript.

Funding

This work was partly supported by funds from the Ministry of Science and Technology (MOST 111-2320-B-037-015-MY3; MOST-108-2320-B-110-009-MY3; MOST 110-2314-B-037-074-MY3), the Kaohsiung Medical University (KMU-DK(A)111008), the National Sun Yat-sen University–KMU Joint Research Project (#NSYSUKMU 111-P20), and the Kaohsiung Medical University Research Center (KMU-TC108A04).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer statistics, 2021. CA Cancer J. Clin. 2021, 71, 7–33. [Google Scholar] [CrossRef]
  2. Waks, A.G.; Winer, E.P. Breast cancer treatment: A review. JAMA 2019, 321, 288–300. [Google Scholar] [CrossRef]
  3. McCann, K.E.; Hurvitz, S.A.; McAndrew, N. Advances in targeted therapies for triple-negative breast cancer. Drugs 2019, 79, 1217–1230. [Google Scholar] [CrossRef]
  4. Mehbub, M.F.; Lei, J.; Franco, C.; Zhang, W. Marine sponge derived natural products between 2001 and 2010: Trends and opportunities for discovery of bioactives. Mar. Drugs 2014, 12, 4539–4577. [Google Scholar] [CrossRef] [Green Version]
  5. Mehbub, M.F.; Perkins, M.V.; Zhang, W.; Franco, C.M.M. New marine natural products from sponges (Porifera) of the order Dictyoceratida (2001 to 2012); A promising source for drug discovery, exploration and future prospects. Biotechnol. Adv. 2016, 34, 473–491. [Google Scholar] [CrossRef]
  6. Calcabrini, C.; Catanzaro, E.; Bishayee, A.; Turrini, E.; Fimognari, C. Marine sponge natural products with anticancer potential: An updated review. Mar. Drugs 2017, 15, 310. [Google Scholar] [CrossRef] [Green Version]
  7. Varijakzhan, D.; Loh, J.Y.; Yap, W.S.; Yusoff, K.; Seboussi, R.; Lim, S.E.; Lai, K.S.; Chong, C.M. Bioactive compounds from marine sponges: Fundamentals and applications. Mar. Drugs 2021, 19, 246. [Google Scholar] [CrossRef]
  8. Perdicaris, S.; Vlachogianni, T.; Valavanidis, A. Bioactive natural substances from marine sponges: New developments and prospects for future pharmaceuticals. Nat. Prod. Chem. Res. 2013, 1, 114. [Google Scholar] [CrossRef]
  9. El-Damhougy, K.; El-Naggar, H.A.; Ibrahim, H.; Bashar, M.A.; Abou-Senna, F.M. Biological activities of some marine sponge extracts from Aqaba Gulf, Red Sea, Egypt. Int. J. Fish. Aquat. Stud. 2017, 5, 652–659. [Google Scholar]
  10. Ottinger, S.; Kloppel, A.; Rausch, V.; Liu, L.; Kallifatidis, G.; Gross, W.; Gebhard, M.M.; Brummer, F.; Herr, I. Targeting of pancreatic and prostate cancer stem cell characteristics by Crambe crambe marine sponge extract. Int. J. Cancer 2012, 130, 1671–1681. [Google Scholar] [CrossRef]
  11. Choi, K.; Lim, H.K.; Oh, S.R.; Chung, W.H.; Jung, J. Anticancer effects of the marine sponge Lipastrotethya sp. extract on wild-type and p53 knockout HCT116 cells. Evid. Based Complement. Altern. Med. 2017, 2017, 7174858. [Google Scholar] [CrossRef] [PubMed]
  12. Bashari, M.H.; Fadhil, M.; Aulia, Y.; Sari, A.K.; Putri, T.; Qomarilla, N.; Atmaja, H.; Sudji, I.R.; Ariyanto, E.F.; Indrati, A.R.; et al. The ethyl acetate fraction of marine sponge Stylissa carteri induces breast cancer cell death via upregulation of Mcl-1S: An in vitro study. Asian Pac. J. Cancer Prev. 2022, 23, 1653–1660. [Google Scholar] [CrossRef] [PubMed]
  13. Han, N.; Li, J.; Li, X. Natural marine products: Anti-colorectal cancer in vitro and in vivo. Mar. Drugs 2022, 20, 349. [Google Scholar] [CrossRef] [PubMed]
  14. Yu, H.B.; Yang, F.; Sun, F.; Ma, G.Y.; Gan, J.H.; Hu, W.Z.; Han, B.N.; Jiao, W.H.; Lin, H.W. Cytotoxic aaptamine derivatives from the South China Sea sponge Aaptos aaptos. J. Nat. Prod. 2014, 77, 2124–2129. [Google Scholar] [CrossRef]
  15. Bashari, M.H.; Arsydinilhuda, F.Z.; Ilhamsyah, R.S.; Nugrahani, A.D.; Nurdin, R.A.; Kartika, A.; Huda, F.; Abdurahman, M.; Putri, T.; Qomarilla, N.; et al. The ethanol extract of marine sponge Aaptos suberitoides suppress cell viability, cell proliferation and cell migration in HER2-positive breast cancer cell line. Asian Pac. J. Cancer Prev. 2021, 22, 25–32. [Google Scholar] [CrossRef]
  16. Farooq Khan, M.; Nasr, F.A.; Baabbad, A.A.; Alqahtani, A.S.; Wadaan, M.A. Investigating the anticancer activity and characterization of bioactive constituents of Moricandia sinaica (Boiss.) Boiss through in vitro and in silico approaches in triple-negative breast cancer cell line. Appl. Sci. 2021, 11, 1244. [Google Scholar] [CrossRef]
  17. Mioso, R.; Marante, F.J.; Bezerra, R.S.; Borges, F.V.; Santos, B.V.; Laguna, I.H. Cytotoxic compounds derived from marine sponges. A review (2010–2012). Molecules 2017, 22, 208. [Google Scholar] [CrossRef] [Green Version]
  18. Calcul, L.; Longeon, A.; Al Mourabit, A.; Guyot, M.; Bourguet-Kondracki, M.-L. Novel alkaloids of the aaptamine class from an Indonesian marine sponge of the genus Xestospongia. Tetrahedron 2003, 59, 6539–6544. [Google Scholar] [CrossRef]
  19. Wang, N.; Ren, D.; Deng, S.; Yang, X. Differential effects of baicalein and its sulfated derivatives in inhibiting proliferation of human breast cancer MCF-7 cells. Chem. Biol. Interact. 2014, 221, 99–108. [Google Scholar] [CrossRef]
  20. Yen, M.C.; Chou, S.K.; Kan, J.Y.; Kuo, P.L.; Hou, M.F.; Hsu, Y.L. New insight on solute carrier family 27 member 6 (SLC27A6) in tumoral and non-tumoral breast cells. Int. J. Med. Sci. 2019, 16, 366–375. [Google Scholar] [CrossRef] [Green Version]
  21. Jao, H.Y.; Chang, F.R.; Cheng, C.W.; Liang, H.W.; Wang, C.J.; Lee, H.J. Silybum marianum seed disrupts mitosis by reducing polo-like kinase 1 in breast cancer cells. Phytomed. Plus 2022, 2, 100164. [Google Scholar] [CrossRef]
  22. Kim, G.Y.; Kim, W.J.; Choi, Y.H. Pectenotoxin-2 from marine sponges: A potential anti-cancer agent—A review. Mar. Drugs 2011, 9, 2176–2187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Dyshlovoy, S.A.; Honecker, F. Marine compounds and cancer: Updates 2020. Mar. Drugs 2020, 18, 643. [Google Scholar] [CrossRef]
  24. Kumar, M.S.; Adki, K.M. Marine natural products for multi-targeted cancer treatment: A future insight. Biomed. Pharm. 2018, 105, 233–245. [Google Scholar] [CrossRef] [PubMed]
  25. Ciftci, H.I.; Can, M.; Ellakwa, D.E.; Suner, S.C.; Ibrahim, M.A.; Oral, A.; Sekeroglu, N.; Ozalp, B.; Otsuka, M.; Fujita, M.; et al. Anticancer activity of Turkish marine extracts: A purple sponge extract induces apoptosis with multitarget kinase inhibition activity. Invest. New Drugs 2020, 38, 1326–1333. [Google Scholar] [CrossRef] [PubMed]
  26. He, L.; He, T.; Farrar, S.; Ji, L.; Liu, T.; Ma, X. Antioxidants maintain cellular redox homeostasis by elimination of reactive oxygen species. Cell. Physiol. Biochem. 2017, 44, 532–553. [Google Scholar] [CrossRef] [PubMed]
  27. Snezhkina, A.V.; Kudryavtseva, A.V.; Kardymon, O.L.; Savvateeva, M.V.; Melnikova, N.V.; Krasnov, G.S.; Dmitriev, A.A. ROS generation and antioxidant defense systems in normal and malignant cells. Oxid. Med. Cell. Longev. 2019, 2019, 6175804. [Google Scholar] [CrossRef] [Green Version]
  28. Khan, M.; Yi, F.; Rasul, A.; Li, T.; Wang, N.; Gao, H.; Gao, R.; Ma, T. Alantolactone induces apoptosis in glioblastoma cells via GSH depletion, ROS generation, and mitochondrial dysfunction. IUBMB Life 2012, 64, 783–794. [Google Scholar] [CrossRef]
  29. Shiau, J.P.; Chuang, Y.T.; Yang, K.H.; Chang, F.R.; Sheu, J.H.; Hou, M.F.; Jeng, J.H.; Tang, J.Y.; Chang, H.W. Brown algae-derived fucoidan exerts oxidative stress-dependent antiproliferation on oral cancer cells. Antioxidants 2022, 11, 841. [Google Scholar] [CrossRef]
  30. Rady, H.M.; Hassan, A.Z.; Salem, S.M.; Mohamed, T.K.; Esmaiel, N.N.; Ez-El-Arab, M.A.; Ibrahim, M.A.; Fouda, F.K. Induction of apoptosis and cell cycle arrest by Negombata magnifica sponge in hepatocellular carcinoma. Med. Chem. Res. 2016, 25, 456–465. [Google Scholar] [CrossRef]
  31. Bae, W.; Lim, H.K.; Kim, K.M.; Cho, H.; Lee, S.Y.; Jeong, C.S.; Lee, H.S.; Jung, J. Apoptosis-inducing activity of marine sponge Haliclona sp. Extracts collected from kosrae in nonsmall cell lung cancer A549 cells. Evid. Based Complement. Altern. Med. 2015, 2015, 717959. [Google Scholar] [CrossRef] [PubMed]
  32. Annamalai, P.; Thayman, M.; Rajan, S.; Raman, L.S.; Ramasubbu, S.; Perumal, P. Ethyl acetate extract from marine sponge Hyattella cribriformis exhibit potent anticancer activity by promoting tubulin polymerization as evidenced mitotic arrest and induction of apoptosis. Pharmacogn. Mag. 2015, 11, 345. [Google Scholar] [PubMed] [Green Version]
  33. Muthiyan, R.; Nambikkairaj, B.; Mahanta, N.; Immanuel, T.; Mandal, R.S.; Kumaran, K.; De, A.K. Antiproliferative and proapoptotic activities of marine sponge Hyrtios erectus extract on breast carcinoma cell line (MCF-7). Pharmacogn. Mag. 2017, 13, S41–S47. [Google Scholar] [PubMed]
  34. Kajstura, M.; Halicka, H.D.; Pryjma, J.; Darzynkiewicz, Z. Discontinuous fragmentation of nuclear DNA during apoptosis revealed by discrete “sub-G1” peaks on DNA content histograms. Cytom. A 2007, 71, 125–131. [Google Scholar] [CrossRef]
  35. Chen, B.H.; Chang, H.W.; Huang, H.M.; Chong, I.W.; Chen, J.S.; Chen, C.Y.; Wang, H.M. (−)-Anonaine induces DNA damage and inhibits growth and migration of human lung carcinoma h1299 cells. J. Agric. Food Chem. 2011, 59, 2284–2290. [Google Scholar] [CrossRef]
  36. Yang, F.; Gao, Y.; Chang, Y.T.; Zou, Y.K.; Houk, K.N.; Lu, J.R.; He, J.; Tang, W.Z.; Liao, H.Z.; Han, H.; et al. Aromatic ring substituted aaptamine analogues as potential cytotoxic agents against extranodal natural killer/T-cell lymphoma. J. Nat. Prod. 2020, 83, 3758–3763. [Google Scholar] [CrossRef]
  37. Holliday, D.L.; Speirs, V. Choosing the right cell line for breast cancer research. Breast Cancer Res. 2011, 13, 215. [Google Scholar] [CrossRef] [Green Version]
  38. Chavez, K.J.; Garimella, S.V.; Lipkowitz, S. Triple negative breast cancer cell lines: One tool in the search for better treatment of triple negative breast cancer. Breast Dis. 2010, 32, 35–48. [Google Scholar] [CrossRef] [Green Version]
  39. Lee, H.Z.; Liu, W.Z.; Hsieh, W.T.; Tang, F.Y.; Chung, J.G.; Leung, H.W. Oxidative stress involvement in Physalis angulata-induced apoptosis in human oral cancer cells. Food Chem. Toxicol. 2009, 47, 561–570. [Google Scholar] [CrossRef]
  40. Wang, T.S.; Lin, C.P.; Chen, Y.P.; Chao, M.R.; Li, C.C.; Liu, K.L. CYP450-mediated mitochondrial ROS production involved in arecoline N-oxide-induced oxidative damage in liver cell lines. Environ. Toxicol. 2018, 33, 1029–1038. [Google Scholar] [CrossRef]
  41. Liu, Y.C.; Peng, B.R.; Hsu, K.C.; El-Shazly, M.; Shih, S.P.; Lin, T.E.; Kuo, F.W.; Chou, Y.C.; Lin, H.Y.; Lu, M.C. 13-Acetoxysarcocrassolide exhibits cytotoxic activity against oral cancer cells through the interruption of the Keap1/Nrf2/p62/SQSTM1 pathway: The need to move beyond classical concepts. Mar. Drugs 2020, 18, 382. [Google Scholar] [CrossRef] [PubMed]
  42. Chang, C.C.; Kuan, C.P.; Lin, J.Y.; Lai, J.S.; Ho, T.F. Tanshinone IIA facilitates TRAIL sensitization by up-regulating DR5 through the ROS-JNK-CHOP signaling axis in human ovarian carcinoma cell lines. Chem. Res. Toxicol. 2015, 28, 1574–1583. [Google Scholar] [CrossRef] [PubMed]
  43. Vignon, C.; Debeissat, C.; Georget, M.T.; Bouscary, D.; Gyan, E.; Rosset, P.; Herault, O. Flow cytometric quantification of all phases of the cell cycle and apoptosis in a two-color fluorescence plot. PLoS ONE 2013, 8, e68425. [Google Scholar] [CrossRef] [Green Version]
  44. Carbonari, M. New use for an old reagent: Cell cycle analysis of DNA content using flow cytometry in formamide treated cells. Cytom. Part A 2016, 89, 498–503. [Google Scholar] [CrossRef] [Green Version]
  45. Huang, H.W.; Tang, J.Y.; Ou-Yang, F.; Wang, H.R.; Guan, P.Y.; Huang, C.Y.; Chen, C.Y.; Hou, M.F.; Sheu, J.H.; Chang, H.W. Sinularin selectively kills breast cancer cells showing G2/M arrest, apoptosis, and oxidative DNA damage. Molecules 2018, 23, 849. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Wang, M.; Wang, J.; Liu, M.; Chen, G. Fluvastatin protects neuronal cells from hydrogen peroxide-induced toxicity with decreasing oxidative damage and increasing PI3K/Akt/mTOR signalling. J. Pharm. Pharmacol. 2021, 73, 515–521. [Google Scholar] [CrossRef] [PubMed]
  47. Lin, C.H.; Chan, H.S.; Tsay, H.S.; Funayama, S.; Kuo, C.L.; Chung, J.G. Ethyl acetate fraction from methanol extraction of Vitis thunbergii var. taiwaniana induced G0 /G1 phase arrest via inhibition of cyclins D and E and induction of apoptosis through caspase-dependent and -independent pathways in human prostate carcinoma DU145 cells. Environ. Toxicol. 2018, 33, 41–51. [Google Scholar]
  48. Fan, H.C.; Hsieh, Y.C.; Li, L.H.; Chang, C.C.; Janouskova, K.; Ramani, M.V.; Subbaraju, G.V.; Cheng, K.T.; Chang, C.C. Dehydroxyhispolon methyl ether, a hispolon derivative, inhibits WNT/beta-catenin signaling to elicit human colorectal carcinoma cell apoptosis. Int. J. Mol. Sci. 2020, 21, 8839. [Google Scholar] [CrossRef]
  49. Liu, S.L.; Yang, K.H.; Yang, C.W.; Lee, M.Y.; Chuang, Y.T.; Chen, Y.N.; Chang, F.R.; Chen, C.Y.; Chang, H.W. Burmannic acid inhibits proliferation and induces oxidative stress response of oral cancer cells. Antioxidants 2021, 10, 1588. [Google Scholar] [CrossRef]
  50. Wu, C.F.; Lee, M.G.; El-Shazly, M.; Lai, K.H.; Ke, S.C.; Su, C.W.; Shih, S.P.; Sung, P.J.; Hong, M.C.; Wen, Z.H.; et al. Isoaaptamine induces T-47D cells apoptosis and autophagy via oxidative stress. Mar. Drugs 2018, 16, 18. [Google Scholar] [CrossRef] [Green Version]
  51. Yeh, C.C.; Tseng, C.N.; Yang, J.I.; Huang, H.W.; Fang, Y.; Tang, J.Y.; Chang, F.R.; Chang, H.W. Antiproliferation and induction of apoptosis in Ca9-22 oral cancer cells by ethanolic extract of Gracilaria tenuistipitata. Molecules 2012, 17, 10916–10927. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Yang, K.H.; Lin, Y.S.; Wang, S.C.; Lee, M.Y.; Tang, J.Y.; Chang, F.R.; Chuang, Y.T.; Sheu, J.H.; Chang, H.W. Soft coral-derived dihydrosinularin exhibits antiproliferative effects associated with apoptosis and DNA damage in oral cancer cells. Pharmaceuticals 2021, 14, 994. [Google Scholar] [CrossRef] [PubMed]
  53. Tanaka, T.; Halicka, D.; Traganos, F.; Darzynkiewicz, Z. Cytometric analysis of DNA damage: Phosphorylation of histone H2AX as a marker of DNA double-strand breaks (DSBs). Methods Mol. Biol. 2009, 523, 161–168. [Google Scholar] [PubMed] [Green Version]
  54. Shih, S.P.; Lu, M.C.; El-Shazly, M.; Lin, Y.H.; Chen, C.L.; Yu, S.S.F.; Liu, Y.C. The antileukemic and anti-prostatic effect of aeroplysinin-1 is mediated through ROS-induced apoptosis via NOX activation and inhibition of HIF-1a activity. Life 2022, 12, 687. [Google Scholar] [CrossRef] [PubMed]
Figure 1. (A) The HPLC fingerprint profiles of MEAS and the main product of MEAS (aaptamine) were shown in red and blue, respectively. (B) Structure of aaptamine. (C) The calibration curve of aaptamine (n = 3).
Figure 1. (A) The HPLC fingerprint profiles of MEAS and the main product of MEAS (aaptamine) were shown in red and blue, respectively. (B) Structure of aaptamine. (C) The calibration curve of aaptamine (n = 3).
Pharmaceuticals 15 01575 g001
Figure 2. MEAS decreased the viability of several kinds of breast cancer cells. (A) Cell viability of MEAS at 24 h MTS assay. Except for normal breast cells (M10), others were TNBC and non-TNBC cells. (B) Cell viability of NAC/MEAS at 24 h MTS assay. NAC/MEAS represents NAC pretreatment (10 mM, 1 h) and MEAS posttreatment for 24 h. Data = mean ± SD (n = 3). Statistical software assigned low-case letters to each treatment. When the letters of different treatments overlapped, the results were significant (p < 0.05). In the example of Figure 2B (MCF7 cells), MEAS treatments at 0, 10, 15, and 25 μg/mL labeled with “a, b, c, and d” differ significantly, determined by non-overlapping conditions. MEAS and NAC/MEAS treatments of MCF7 cells at 10 μg/mL MEAS labeled with “b and ab” show a non-significant difference because they overlapped with “b”, while at 15 μg/mL MEAS labeled with “c and ab” show a significant difference.
Figure 2. MEAS decreased the viability of several kinds of breast cancer cells. (A) Cell viability of MEAS at 24 h MTS assay. Except for normal breast cells (M10), others were TNBC and non-TNBC cells. (B) Cell viability of NAC/MEAS at 24 h MTS assay. NAC/MEAS represents NAC pretreatment (10 mM, 1 h) and MEAS posttreatment for 24 h. Data = mean ± SD (n = 3). Statistical software assigned low-case letters to each treatment. When the letters of different treatments overlapped, the results were significant (p < 0.05). In the example of Figure 2B (MCF7 cells), MEAS treatments at 0, 10, 15, and 25 μg/mL labeled with “a, b, c, and d” differ significantly, determined by non-overlapping conditions. MEAS and NAC/MEAS treatments of MCF7 cells at 10 μg/mL MEAS labeled with “b and ab” show a non-significant difference because they overlapped with “b”, while at 15 μg/mL MEAS labeled with “c and ab” show a significant difference.
Pharmaceuticals 15 01575 g002
Figure 3. MEAS causes cell cycle redistribution of breast cancer cells. (A) Cell cycle assay of MEAS. Except for normal breast cells (M10), others were TNBC and non-TNBC cells. Flow cytometry was performed after 24 h drug treatment. (B) Cell cycle assay of NAC/MEAS. NAC/MEAS represents NAC pretreatment (10 mM, 1 h) and MEAS posttreatment for 0, 12, and 24 h. Data = mean ± SD (n = 3). Statistical software assigned low-case letters to each treatment. When the letters of different treatments overlapped, the results were significant (p < 0.05).
Figure 3. MEAS causes cell cycle redistribution of breast cancer cells. (A) Cell cycle assay of MEAS. Except for normal breast cells (M10), others were TNBC and non-TNBC cells. Flow cytometry was performed after 24 h drug treatment. (B) Cell cycle assay of NAC/MEAS. NAC/MEAS represents NAC pretreatment (10 mM, 1 h) and MEAS posttreatment for 0, 12, and 24 h. Data = mean ± SD (n = 3). Statistical software assigned low-case letters to each treatment. When the letters of different treatments overlapped, the results were significant (p < 0.05).
Pharmaceuticals 15 01575 g003
Figure 4. MEAS increased the annexin V intensity of breast cancer cells. (A) Annexin V-apoptosis assay of MEAS. Except for normal breast cells (M10), others were TNBC and non-TNBC cells. Flow cytometry was performed after 24 h drug treatment. Annexin V (+) (%), such as annexin V (+)/7AAD (+, −), accounts for apoptosis (%). (B) Annexin V-apoptosis assay of NAC/MEAS. NAC/MEAS represents NAC pretreatment (10 mM, 1 h) and MEAS posttreatment for 0, 12, and 24 h. Data = mean ± SD (n = 3). Statistical software assigned low-case letters to each treatment.
Figure 4. MEAS increased the annexin V intensity of breast cancer cells. (A) Annexin V-apoptosis assay of MEAS. Except for normal breast cells (M10), others were TNBC and non-TNBC cells. Flow cytometry was performed after 24 h drug treatment. Annexin V (+) (%), such as annexin V (+)/7AAD (+, −), accounts for apoptosis (%). (B) Annexin V-apoptosis assay of NAC/MEAS. NAC/MEAS represents NAC pretreatment (10 mM, 1 h) and MEAS posttreatment for 0, 12, and 24 h. Data = mean ± SD (n = 3). Statistical software assigned low-case letters to each treatment.
Pharmaceuticals 15 01575 g004
Figure 5. MEAS increased activation of Caspases 3, 8, and 9 of breast cancer cells. (A,C,E) Caspases 3, 8, and 9 activation assays of MEAS. Except for normal breast cells (M10), others were TNBC and non-TNBC cells. Flow cytometry was performed after 24 h drug treatment. (+), inserted within each panel, indicates the caspases 3, 8, and 9 (+) intensity. (B,D,F) Caspases 3, 8, and 9 activation assays of NAC/MEAS. NAC/MEAS represents NAC pretreatment (10 mM, 1 h) and MEAS posttreatment for 0, 12, and 24 h. Data = mean ± SD (n = 3). Statistical software assigned low-case letters to each treatment.
Figure 5. MEAS increased activation of Caspases 3, 8, and 9 of breast cancer cells. (A,C,E) Caspases 3, 8, and 9 activation assays of MEAS. Except for normal breast cells (M10), others were TNBC and non-TNBC cells. Flow cytometry was performed after 24 h drug treatment. (+), inserted within each panel, indicates the caspases 3, 8, and 9 (+) intensity. (B,D,F) Caspases 3, 8, and 9 activation assays of NAC/MEAS. NAC/MEAS represents NAC pretreatment (10 mM, 1 h) and MEAS posttreatment for 0, 12, and 24 h. Data = mean ± SD (n = 3). Statistical software assigned low-case letters to each treatment.
Pharmaceuticals 15 01575 g005aPharmaceuticals 15 01575 g005b
Figure 6. MEAS increased ROS and MitoSOX intensities of breast cancer cells. (A,C) ROS and MitoSOX assays of MEAS. Except for normal breast cells (M10), others were TNBC and non-TNBC cells. Flow cytometry was performed after 24 h drug treatment. (+), inserted within each panel, indicates the ROS and MitoSOX (+) intensity. (B,D) ROS and MitoSOX assays of NAC/MEAS. NAC/MEAS represents NAC pretreatment (10 mM, 1 h) and MEAS posttreatment for 0, 12, and 24 h. Data = mean ± SD (n = 3). Statistical software assigned low-case letters to each treatment.
Figure 6. MEAS increased ROS and MitoSOX intensities of breast cancer cells. (A,C) ROS and MitoSOX assays of MEAS. Except for normal breast cells (M10), others were TNBC and non-TNBC cells. Flow cytometry was performed after 24 h drug treatment. (+), inserted within each panel, indicates the ROS and MitoSOX (+) intensity. (B,D) ROS and MitoSOX assays of NAC/MEAS. NAC/MEAS represents NAC pretreatment (10 mM, 1 h) and MEAS posttreatment for 0, 12, and 24 h. Data = mean ± SD (n = 3). Statistical software assigned low-case letters to each treatment.
Pharmaceuticals 15 01575 g006aPharmaceuticals 15 01575 g006b
Figure 7. MEAS increased MMP (−) intensities of breast cancer cells. (A) MMP assay of MEAS. Except for normal breast cells (M10), others were TNBC and non-TNBC cells. Flow cytometry was performed after 24 h drug treatment. (−), inserted within each panel, indicates the MMP (−) intensity. (B) MMP assay of NAC/MEAS. NAC/MEAS represents NAC pretreatment (10 mM, 1 h) and MEAS posttreatment for 0, 12, and 24 h. Data = mean ± SD (n = 3). Statistical software assigned low-case letters to each treatment.
Figure 7. MEAS increased MMP (−) intensities of breast cancer cells. (A) MMP assay of MEAS. Except for normal breast cells (M10), others were TNBC and non-TNBC cells. Flow cytometry was performed after 24 h drug treatment. (−), inserted within each panel, indicates the MMP (−) intensity. (B) MMP assay of NAC/MEAS. NAC/MEAS represents NAC pretreatment (10 mM, 1 h) and MEAS posttreatment for 0, 12, and 24 h. Data = mean ± SD (n = 3). Statistical software assigned low-case letters to each treatment.
Pharmaceuticals 15 01575 g007
Figure 8. MEAS increased GSH (−) intensities of breast cancer cells. (A) GSH assay of MEAS. Except for normal breast cells (M10), others were TNBC and non-TNBC cells. Flow cytometry was performed after 6 h drug treatment. (−), inserted within each panel, indicates the GSH (−) intensity. (B) GSH assay of NAC/MEAS. NAC/MEAS represents NAC pretreatment (10 mM, 1 h) and MEAS posttreatment for 0, 1, 3, and 6 h. Data = mean ± SD (n = 3). Statistical software assigned low-case letters to each treatment.
Figure 8. MEAS increased GSH (−) intensities of breast cancer cells. (A) GSH assay of MEAS. Except for normal breast cells (M10), others were TNBC and non-TNBC cells. Flow cytometry was performed after 6 h drug treatment. (−), inserted within each panel, indicates the GSH (−) intensity. (B) GSH assay of NAC/MEAS. NAC/MEAS represents NAC pretreatment (10 mM, 1 h) and MEAS posttreatment for 0, 1, 3, and 6 h. Data = mean ± SD (n = 3). Statistical software assigned low-case letters to each treatment.
Pharmaceuticals 15 01575 g008
Figure 9. MEAS increased γH2AX intensity of breast cancer cells. (A) γH2AX assay of MEAS. Except for normal breast cells (M10), others were TNBC and non-TNBC cells. Flow cytometry was performed after 24 h drug treatment. (+), inserted within each panel, indicates the γH2AX (+) intensity. (B) γH2AX assay of NAC/MEAS. NAC/MEAS represents NAC pretreatment (10 mM, 1 h) and MEAS posttreatment for 0, 12, and 24 h. Data = mean ± SD (n = 3). Statistical software assigned low-case letters to each treatment.
Figure 9. MEAS increased γH2AX intensity of breast cancer cells. (A) γH2AX assay of MEAS. Except for normal breast cells (M10), others were TNBC and non-TNBC cells. Flow cytometry was performed after 24 h drug treatment. (+), inserted within each panel, indicates the γH2AX (+) intensity. (B) γH2AX assay of NAC/MEAS. NAC/MEAS represents NAC pretreatment (10 mM, 1 h) and MEAS posttreatment for 0, 12, and 24 h. Data = mean ± SD (n = 3). Statistical software assigned low-case letters to each treatment.
Pharmaceuticals 15 01575 g009
Figure 10. MEAS increased the 8-OHdG intensity of breast cancer cells. (A) 8-OHdG assay of MEAS. Except for normal breast cells (M10), others were TNBC and non-TNBC cells. Flow cytometry was performed after 24 h drug treatment. (+), inserted within each panel, indicates the 8-OHdG (+) intensity. (B) 8-OHdG assay of NAC/MEAS. NAC/MEAS represents NAC pretreatment (10 mM, 1 h) and MEAS posttreatment for 0, 12, and 24 h. Data = mean ± SD (n = 3). Statistical software assigned low-case letters to each treatment.
Figure 10. MEAS increased the 8-OHdG intensity of breast cancer cells. (A) 8-OHdG assay of MEAS. Except for normal breast cells (M10), others were TNBC and non-TNBC cells. Flow cytometry was performed after 24 h drug treatment. (+), inserted within each panel, indicates the 8-OHdG (+) intensity. (B) 8-OHdG assay of NAC/MEAS. NAC/MEAS represents NAC pretreatment (10 mM, 1 h) and MEAS posttreatment for 0, 12, and 24 h. Data = mean ± SD (n = 3). Statistical software assigned low-case letters to each treatment.
Pharmaceuticals 15 01575 g010
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Shiau, J.-P.; Lee, M.-Y.; Tang, J.-Y.; Huang, H.; Lin, Z.-Y.; Su, J.-H.; Hou, M.-F.; Cheng, Y.-B.; Chang, H.-W. Marine Sponge Aaptos suberitoides Extract Improves Antiproliferation and Apoptosis of Breast Cancer Cells without Cytotoxicity to Normal Cells In Vitro. Pharmaceuticals 2022, 15, 1575. https://doi.org/10.3390/ph15121575

AMA Style

Shiau J-P, Lee M-Y, Tang J-Y, Huang H, Lin Z-Y, Su J-H, Hou M-F, Cheng Y-B, Chang H-W. Marine Sponge Aaptos suberitoides Extract Improves Antiproliferation and Apoptosis of Breast Cancer Cells without Cytotoxicity to Normal Cells In Vitro. Pharmaceuticals. 2022; 15(12):1575. https://doi.org/10.3390/ph15121575

Chicago/Turabian Style

Shiau, Jun-Ping, Min-Yu Lee, Jen-Yang Tang, Hsin Huang, Zheng-Yu Lin, Jui-Hsin Su, Ming-Feng Hou, Yuan-Bin Cheng, and Hsueh-Wei Chang. 2022. "Marine Sponge Aaptos suberitoides Extract Improves Antiproliferation and Apoptosis of Breast Cancer Cells without Cytotoxicity to Normal Cells In Vitro" Pharmaceuticals 15, no. 12: 1575. https://doi.org/10.3390/ph15121575

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop