Research Article Comparison of Apoptotic Inducing ...

2 downloads 71 Views 4MB Size Report
4 Faculty of Veterinary Medicine, University of Sulaimani, Sulaymaniyah City, Kurdistan Region, Iraq. 5 DigiCare Behavioral Research, Casa Grande, AZ, USA.
Hindawi Publishing Corporation Journal of Nanomaterials Article ID 742738

Research Article Comparison of Apoptotic Inducing Effect of Zerumbone and Zerumbone-Loaded Nanostructured Lipid Carrier on Human Mammary Adenocarcinoma MDA-MB-231 Cell Line Mahnaz Hosseinpour,1 Ahmad Bustamam Abdul,1,2 Heshu Sulaiman Rahman,1,3,4 Abdullah Rasedee,1,3 Swee Keong Yeap,1 Negin Ahmadi,1 Hemn Hassan Othman,3,4 and Max Stanley Chartrand5 1

Institute of Bioscience, Universiti Putra Malaysia, (UPM) 43400 Serdang, Selangor, Malaysia Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, (UPM) 43400 Serdang, Selangor, Malaysia 3 Faculty of Veterinary Medicine, Universiti Putra Malaysia, (UPM) 43400 Serdang, Selangor, Malaysia 4 Faculty of Veterinary Medicine, University of Sulaimani, Sulaymaniyah City, Kurdistan Region, Iraq 5 DigiCare Behavioral Research, Casa Grande, AZ, USA 2

Correspondence should be addressed to Mahnaz Hosseinpour; mahnaz [email protected] and Ahmad Bustamam Abdul; [email protected] Received 10 August 2014; Revised 30 October 2014; Accepted 30 October 2014 Academic Editor: Hongchen Chen Gu Copyright © Mahnaz Hosseinpour et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. This study investigated the anticancer effect of zerumbone (ZER) and zerumbone-loaded nanostructured lipid carrier (ZER-NLC) on the human mammary gland adenocarcinoma (MDA-MB-231) cell line. The effect of ZER and ZER-NLC on MDA-MB-231 cells was determined via electron and fluorescent microscopy and flow cytometry using the Annexin V, cell cycle, and Tdt-mediated dUTP nick-end labeling assays. We demonstrated that ZER and ZER-NLC significantly suppressed the proliferation of MDA-MB231 cells with an IC50 of 5.96 ± 0.13 and 6.01 ± 0.11 𝜇g/mL, respectively. ZER and ZER-NLC arrested MDA-MB-231 cell cycle at the G2/M phase. The induction of apoptosis by ZER and ZER-NLC was via the intrinsic pathway through the release of cytochrome c and activation of caspase-3 and caspase-9. The treatments also caused the downregulation of antiapoptotic Bcl-2, Bcl-xL proteins, and proliferating cell nuclear protein and upregulation of proapoptotic Bax protein. Therefore, loading of ZER into NLC did not compromise the anticancer effects of ZER on MDA-MB-231 cells. In conclusion, ZER-NLC, which increased the bioavailability of ZER, is an effective agent in the treatment of cancers.

1

2

1. Introduction The main life-threatening and the most prevalent diseases in women are breast cancers [1]. Development of breast cancers is associated with several risk factors such as age, genes, and family history. There are several treatment methods currently being instituted for breast cancers to include chemotherapy, hormonal therapy using tamoxifen and doxorubicin, radiotherapy, and surgery. Most of these treatment methods are not specific for cancer cells and more often also damage normal cells. Current studies are increasingly providing evidences that therapeutic compounds from natural sources are

promising for effective treatment of chronic human diseases including cancers, without significant side-effect [2]. Among these compounds is zerumbone (ZER), extracted from the essential oils of Zingiber zerumbet Smith rhizome. Zerumbone has versatile pharmaceutical properties including anti-inflammatory, antitumor, antioxidant, antimicrobial, and antinociceptive activities [3, 4]. This compound also significantly suppresses activation of nuclear factor 𝜅-light-chain-enhancer in activated B cells (NF-𝜅B) and NF-𝜅B-related gene expression generated by carcinogens in various other cells [5]. Zerumbone has been reported to

2

Journal of Nanomaterials MDA-MB-231

120

100 Cell viability (%)

100 Cell viability (%)

MDA-MB-231

120

80 60 40

80 60 40 20

20

0

0 0

50 100 ZER concentration (𝜇g/mL) 24 h 48 h

0

150

50

100

150

ZER-NLC concentration (𝜇g/mL) 24 h 48 h

72 h

72 h

(a)

(b)

Figure 1: Viability of MDA-MB-231 cells treated with (a) ZER and (b) ZER-NLC after 24, 48, and 72 hours. MCF-10A

120

100 Cell viability (%)

100 Cell viability (%)

MCF-10A

120

80 60 40

80 60 40 20

20

0

0 0

50

100

150

0

50

24 h 48 h

72 h

(a)

100

150

ZER-NLC concentration (𝜇g/mL)

ZER concentration (𝜇g/mL) 24 h 48 h

72 h

(b)

Figure 2: Viability of MCF-10A cells treated with (a) ZER and (b) ZER-NLC after 24, 48, and 72 hours.

inhibit proliferation of cervical, ovarian, colon, breast, and liver cancers [6, 7]. Although zerumbone has pharmaceutical potentials, like many other drugs and synthetic therapeutic compounds, its application in therapy is hindered by poor water solubility that leads to poor bioavailability and delivery [8]. This property retards zerumbone absorption in the gastrointestinal tract making oral applications relatively ineffective. The poor absorption in the gastrointestinal tract not only reduces bioavailability of zerumbone but also causes mucosal toxicity [9, 10]. One approach to overcome this obstacle is to incorporate drugs into lipid nanocarriers, which would improve solubility and delivery of zerumbone by facilitating transportation across several anatomical barriers. Solid lipid nanoparticles (SLN), nanostructured lipid carriers (NLC), and lipid-drug conjugates are examples of lipid-based carrier systems that can be used to incorporate therapeutic compounds. Compound-loaded lipids nanocarriers exhibit

desirable characteristics including low toxicity, controlled release, biodegradable, high drug content, low cost, and easy upscaling [11, 12]. Nanostructured lipid carrier, a second generation lipid nanocarrier produced by high-pressure homogenization, is composed of a solid and liquid lipid matrix with disrupted crystalline nanostructure [13, 14]. This lipid nanocarrier, while overcoming some of potential therapeutic application problems associated with SLN, also possesses high loading capacity, low water content of particle suspension, and avoidance of drug expulsion [15, 16]. The purpose of this study was to determine the effect of ZER and zerumbone-loaded nanostructured lipid carriers (ZER-NLC) on the proliferation of a breast cancer (MDA-MB-231) cell line.

3

Journal of Nanomaterials

3 48 hours

72 hours

(b)

(c)

Control

24 hours

(a)

CS MB

NF

ZER

MB

MB

AB MB NF

(d)

(e)

(f)

MB ZER-NLC

CS

NF MB

(g)

(h)

(i)

Figure 3: Morphology of MDA-MB-231 cells treated with (d, e, f) ZER and (g, h, i) ZER-NLC after 24, 48, and 72 hours. (a), (b), and (c) are untreated cells after 24, 48, and 72 hours, respectively. MB = membrane blebbing, NF = nuclear fragmentation, CS = cell shrinkage, and AB = apoptotic body (×400).

2. Materials and Methods 2.1. Materials. The pure colorless ZER and ZER-NLC were prepared according to the method described by Rahman et al. [10]. MDA-MB-231 cells (human breast adenocarcinoma), MCF-10A (normal breast cell line), RPMI-1640, and Dulbecco’s modified Eagle’s medium (DMEM) were obtained from the American Type Culture Collection (Baltimore, MD, USA). Dimethyl sulfoxide (DMSO), fetal bovine serum (FBS), penicillin-streptomycin, propidium iodide, acridine orange, trypsin-EDTA, and MTT powder were purchased from Sigma Chemical Co. (St. Louis, MO, USA). 2.2. Cytotoxicity Assay. MDA-MB-231 and MCF-10A cells were cultured according to the manufacture’s instruction (ATCC, USA) and each cell line was seeded at a concentration of 2 × 105 cells/mL into a 96-well plate. The cytotoxic effect of 100, 50, 25, 12.5, 6.25, 3.125, 1.562, and 0 𝜇g/mL of ZER and

ZER-NLC on these breast cells was determined using the 3[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide (MTT) dye after 24, 48, and 72 hours of incubation [17]. All assays were done in triplicate and 0.1% DMSO was used as the negative control. 2.3. Morphological Characterization. MDA-MB-231 cells (2 × 106 cells/mL) were then treated with IC50 concentrations of ZER and ZER-NLC for 24, 48, and 72 hours. At end of each treatment period, the morphology of ZER- and ZER-NLCtreated cells was observed under an inverted microscope (Leica, Tokyo, Japan). 2.4. Quantification of Apoptosis. To determine the effect of ZER and ZER-NLC on apoptosis of breast cancer cells, the MDA-MB-231 cells (2 × 106 cells/mL) treated for 24, 48, and 72 hours were double-stained with propidium iodide (PI) and acridine-orange (AO) according to standard procedure

4

Journal of Nanomaterials

Control

VC

50 𝜇m

(a) 24 hr

48 hr

72 hr

ZER

LA MB CF

SN LA

CC

CC

50 𝜇m

50 𝜇m

50 𝜇m

(b1)

(c1)

(d1)

24 hr

48 hr

72 hr

ZER-NLC

SN CC

CF

CC

MB

LA 50 𝜇m

(b2)

50 𝜇m

50 𝜇m

(c2)

(d2)

Figure 4: Fluorescent micrograph of AO/PI-stained cells treated with ZER and ZER-NLC. (a) Untreated cells, (b1 and b2) 24-hour treatment, (c1 and c2) 48-hour treatment, and (d1 and d2) 72-hour treatment. VC = viable cells; CC = chromatin condensation; CF = chromatin fragmentation; MB = membrane blebbing; LA = late apoptosis; SN= secondary necrosis.

and examined under fluorescence microscope (Leica, Tokyo, Japan) [18]. 2.5. Annexin Assay. Induction of apoptosis in the treated MDA-MB-231 cells was determined using the Annexin

V-fluorescein isothiocyanate (FITC) kit (Sigma-Aldrich) according to manufacturer’s instructions and analysed by BD flow cytometer equipped with an argon laser (Cyan ADP; Dako Denmark A/P, Glostrup, Denmark).

Journal of Nanomaterials 12 hours

Propidium iodide

103 102 101 100

10

0

10

1

2

10 10 Annexin V FITC

24 hours

104

3

10

103 102 101 100 100

4

10

1

10 10 Annexin V FITC

3

10

103 102 101 100

4

104

103

103

103

101

101 102 103 Annexin V FITC

102 101 100 100

104

Propidium iodide

104

102

101 102 103 Annexin V FITC

(d)

10

Propidium iodide

103 102 101 100

100

101 102 103 Annexin V FITC

101 100

104

100

104

10

103 102 101 100

100

101 102 103 Annexin V FITC

(g)

(h)

101 102 103 Annexin V FITC

104

(f)

4

Propidium iodide

10

104

102

(e)

4

103 101 102 Annexin V FITC (c)

104

100 100

100

(b)

Propidium iodide

ZER Propidium iodide

(a)

ZER-NLC Propidium iodide

2

48 hours

104

Propidium iodide

104

Control Propidium iodide

5

104

4

103 102 101 100

100

101 102 103 Annexin V FITC

104

(i)

Figure 5: MDA-MB-231 cell treated with ZER and ZER-NLC and labeled with FITC-conjugated Annexin V and propidium iodide. (a, b, c) untreated cells, (d, e, f) cells treated with ZER, and (g, h, i) cells treated with ZER-NLC. Cells were treated for 12, 24, and 48 hours.

4

2.6. Cell Cycle Analysis. The determination of cell cycle distribution of ZER- and ZER-NLC-treated MDA-MB-231 cell was by flow cytometry after staining with PI according to the protocol explained elsewhere [19]. 2.7. Caspases Assay. The commercial colorimetric assay kit (Genscript Corporation Inc., Piscataway, NJ, USA) was used to determine the activity of caspase-3, caspase-8, and caspase9 in MDA-MB-231 cells treated with ZER and ZER-NLC

for 12, 24, and 48 hours, according to the manufacturer’s instructions.

2.8. TUNEL Assay. The Tdt-mediated dUTP nick-end labeling (TUNEL) with the apoptotic detection kit (DeadEnd fluorometric TUNEL system; Promega, Madison, WI, USA) was applied to determine the mode of cell death induced by ZER and ZER-NLC. The assay quantifies fragmented DNA in

6

Journal of Nanomaterials 24 hours

300 240

240 G2 + M

Counts

Sub-G0/G1

120

180 120

S

60 0

0

G2 + M Sub-G0/G1 S

60

200 400 DNA content

0

600

400 200 DNA content

0

(a)

G0/G1

Sub-G0/G1 S

60 0

G2 + M

Counts

ZER Counts

240

120

0

120

G0/G1

0

600

S

0

300

240

240 G2 + M

G0/G1 Sub-G0/G1

120

S

60

200 400 DNA content

0

600

0

(d)

180

300

240

240

600

Sub-G0/G1

120

S

60

200 400 DNA content

0

600

G2 + M

G0/G1

0

(e)

300

400 200 DNA content (c)

300

180

G2 + M

Sub-G0/G1

(b)

300

180

180

60

Counts

Control Counts

180

240

G0/G1

G0/G1

48 hours

300

Counts

12 hours

300

200 400 DNA content

600

(f)

300 240

120

G2 + M

Sub-G0/G1

G2 + M

180 120

Sub-G0/G1

S

60 0

S

60

0

200 400 DNA content

600

0

Counts

G0/G1

180

Counts

ZER-NLC Counts

G0/G1

0

(g)

200 400 DNA content (h)

G0/G1

180 120

Sub-G0/G1 S

60

600

0

G2 + M

0

200 400 DNA content

600

(i)

Figure 6: Cell cycle analysis of MDA-MB-231 cells treated with (d, e, f) ZER and (g, h, i) ZER-NLC after 24, 48, and 72 hours. (a), (b), and (c) are untreated cells after 24, 48, and 72 h, respectively.

apoptotic cells using the FACSCalibur flow cytometer (BD) equipped with an argon laser (BD). 2.9. Western Blotting Analysis. MDA-MB-231 cells (1 × 106 ) treated with ZER and ZER-NLC were lysed in the radioimmunoprecipitation assay (RIPA) buffer (Thermo Fisher Scientific, Waltham, MA, USA) and expressions of cytochrome c, Bcl-2, Bcl-xL and Bax proteins, and proliferating cell nuclear antigen (PCNA) determined by western blotting. Briefly, protein (25 𝜇g) from treated and untreated cells was separated on 10% on sodium dodecyl sulfate polyacrylamide gel and transferred to polyvinylidene difluoride membranes

(Bio-Rad Laboratories, Hercules, CA, USA). The membrane was blocked with 5% nonfat dry milk in phosphate-buffered saline with 0.05% Tween-20 for 60 minutes at room temperature. The membrane was probed by incubation with the primary rabbit anti-Bcl-2, -Bcl-xL, -Bax, -cytochrome c, and -PCNA antibodies (Abcam, USA) followed by horseradish peroxidase-conjugated secondary rabbit anti-goat antibody (Abcam, USA). The antibody binding was detected by chemiluminescence system (ECL Western blot substrate; Abcam, Cambridge, UK). The membrane was also probed with rabbit anti-𝛽-actin antibody (Abcam, USA) as a loading control. Finally Image J software was used to quantify the bands.

Journal of Nanomaterials 24 hours

TUNEL FITC

103 102 101 100

0

200

400 600 DNA-area

48 hours

104

800

103 102 101 100

1000

0

200

800

102 101 100

1000

104

103

103

103

102 101

200

400 600 DNA-area

800

102 101 100

1000

TUNEL FITC

104

0

0

200

(d) 10

TUNEL FITC

103 102 101

0

200

400 600 DNA-area

800

1000

400 600 DNA-area

800

1000

100

10

103 102 101

0

200

(g)

800

1000

0

200

400 600 DNA-area

800

1000

(f)

4

100

400 600 DNA-area

101

(e)

4

100

200

102

TUNEL FITC

10

0

(c)

104

100

ZER-NLC TUNEL FITC

103

(b)

TUNEL FITC

ZER TUNEL FITC

(a)

400 600 DNA-area

72 hours

104

TUNEL FITC

104

Control TUNEL FITC

7

400 600 DNA-area (h)

800

1000

4

103 102 101 100

0

200

400 600 DNA-area

800 1000

(i)

Figure 7: TUNEL flow cytometric analysis of MDA-MB-231 cells treated with (d, e, f) ZER and (g, h, i) ZER-NLC after 24, 48, and 72 hours. (a), (b), and (c) are untreated cells after 24, 48, and 72 h, respectively.

2.10. Statistical Analysis. All experiments were performed in triplicate and results were presented as mean ± standard deviation. Statistical package for the social sciences (SPSS), version 19.0 (SPSS Inc., Chicago, USA), and analysis of variance (ANOVA) were utilized for statistical analysis.

3. Results and Discussion 3.1. Zerumbone-Loaded NLC. The ZER-NLC was shown to be physically stable, with a particle size of 52.68 ± 0.1 nm, a zeta potential of −25.03 ± 1.24 mV, and a polydispersity index of 0.29 ± 0.0041 𝜇m [10, 14].

3.2. Cytotoxic Effect of ZER and ZER-NLC on MDA-MB-231 Cells. Several studies showed that ZER exerts antiproliferative effects on several types of cancer cell while minimally affecting normal cells [20, 21]. The IC50 for ZER on MDAMB-231 cells were 10.15 ± 0.9, 8.3 ± 0.15, and 5.96 ± 0.13 𝜇g/mL, while for ZER-NLC they were 11.45 ± 0.12, 9.4 ± 0.18, and 6.01 ± 0.11 𝜇g/mL after 24, 48, and 72 hours of incubation, respectively. On the MCF-10A cell, a normal epithelial mammary gland cell line, the IC50 of ZER and ZER-NLC at the same incubation periods were similarly low (Figures 1 and 2). This study also suggests that incorporation of ZER into NLC did not change the

8

Journal of Nanomaterials 0.7

There was a significant shift (𝑃 < 0.05) from early to late apoptosis in the distribution of treated MDA-MB-231 cells after ZER and ZER-NLC treatments. This shift in cell distribution is accompanied by decrease in distribution of viable cells, particularly after 48 hours of treatment. The Annexin V assay further confirms that both ZER and ZERNLC induce apoptosis in MDA-MB-231 cells in a timedependent manner.

Caspase activity

Optical density (450 nm)

0.6 0.5 0.4 0.3 0.2 0.1

ZER

48 h ZER-NLC

24 h ZER-NLC

12 h ZER-NLC

Control

48 h ZER

24 h ZER

12 h ZER

Control

0

ZER-NLC Time (hours)

Caspase-9 Caspase-3

Caspase-8

Figure 8: Caspases activity in MDA-MB-231 cells treated with ZER and ZER-NLC for 12, 24, and 48 h.

cytotoxic effect of the compound on MDA-MB-231 cells, thus maintaining its anticancer activity. This finding is consistent with that reported early in human lymphoblastic leukemia cells [10]. 3.3. Morphological Characterization Using Normal Inverted Microscope. Both ZER and ZER-NLC treatments suppressed proliferation of MDA-MB-231 cells while the proliferation of untreated cells (control) increased and remained confluent during all incubation periods. Morphologically, MDA-MB231 cells treated with ZER and ZER-NLC showed irregular shape, membrane blebbing, cell shrinkage, chromatin condensation, and presence of apoptotic bodies (Figure 3).

5

3.4. Morphological Characterisation Using Acridine Orange/Propidium Iodide (AO/PI). Using the AO and PI double-staining method, the breast cancer cells treated with ZER and ZER-NLC showed apoptotic features in a time-dependent manner, while untreated cells showed green nuclear staining suggesting intact nuclear structure (Figure 4). Early apoptosis in treated cells indicated DNA fragmentation after 24 hours, as shown by the bright-green color. Chromatin condensation was also observed by the appearance of orange color in the cells indicating formation of apoptotic bodies, characteristic of late apoptosis. On the other hand, necrotic cells, characterised by reddish-orange nuclei, were also observed after 72 hours of treatments. 3.5. Apoptosis Detection by Annexin Assay. MDA-MB-231 cells treated with ZER and ZER-NLC showed apoptosis, while viable cells number decreased with period of treatment (Figure 5; see Supplementary Table S1 in Supplementary Material available online at http://dx.doi.org/10.1155/2014/742738).

3.6. Cell Cycle Analysis Using Flow Cytometry. The results demonstrated that ZER and ZER-NLC induced depletion of the G1 phase and S phase of MDA-MB-231 cells with concomitant accumulation of the sub-G0/G1 and G2/M phases at all periods of treatment (Figure 6 and Table S2). The study also showed that both ZER and ZER-NLC caused marked arrest of (𝑃 < 0.05) MDA-MB-231 cells at G2/M phase that increased with duration of treatment. Concurrently, the sub-G0/G1 peak became evident beginning as early as 12 hours of treatment in the ZER-NLC-treated cells. This peak appeared later, at 24 hours with ZER treatment. The results show that both ZER and ZER-NLC are potent in the induction of apoptosis of the MDA-MD-231 cells. Cell cycle arrest is a normal regulatory pathway that allows for tissue maintenance, such as repair of cell damage. Cell cycle arrest is also dependent on availability of essential hormones, growth factors, and nutrients [22]. In disease, gene mutation and tumorigenesis can be induced by defects in these cell cycle checkpoints leading to uncontrolled proliferation. With the treatment of ZER and ZER-NLC, MDA-MB-231 cell cycle arrest at G2/M phase is proposed to occur through increased expression of cyclin B1, Cdk1, Cdc25C, and Cdc25B proteins [23]. 3.7. Apoptosis Detection Using TUNEL Assay. Nuclear fragmentation was observed in MDA-MB-231 cells treated with either ZER or ZER-NLC. The DNA fragmentation shows that the rate of apoptosis of MDA-MB-231 cells increased gradually and significantly (𝑃 < 0.05) during the treatment periods (Figure 7). However, untreated cells proliferated without showing apoptosis (Table S3). 3.8. Detection of Caspase Protease Activity. Caspases are among essential enzymes of the apoptosis-signaling pathway. Caspase-3, initiated by caspase-8, plays a key role in the death receptor pathway, while caspase-9 is a major player in the mitochondrial pathway [24, 25]. Thus these caspases were chosen to determine the mechanism of apoptosis in the MDA-MB-231 cells treated with ZER and ZER-NLC (Figure 8). In this study, the activity of caspase-3 and caspase9 increased significantly (𝑃 < 0.05) with time in ZER- and ZER-NLC-treated cells. However, there was no increase in caspase-8 activity in the MDA-MB-cells during the same period of treatment (Table S4). The results suggest that ZER and ZER-NLC induced apoptosis of MDA-MB-231 via the intrinsic pathway. 3.9. Western Blotting. Determination of pro- and antiapoptotic proteins is another method of ascertaining the mode

6

Journal of Nanomaterials Control

12 hours

9 24 hours

48 hours

Control 12 hours 24 hours 48 hours

Bcl-2

Bcl-2

Bcl-xL PCNA

Bcl-xL PCNA

Bax Bax Cytochrome c

Cytochrome c

𝛽-Actin

𝛽-Actin

(a)

(b)

Figure 9: Western blot expression apoptosis-regulating proteins in MDA-MB-231 cells treated with (a) ZER and (b) ZER-NLC after 12, 24, and 48 h. The treated cells were compared with untreated cells (control).

7

of apoptosis in treated cells. In this study, Bax, Bcl-2, and Bcl-xL protein expressions in MDA-MB-231 cells treated with ZER and ZER-NLC were determined. Other proteins, PCNA and cytochrome c, that play roles in cell proliferation and death were also estimated. The treatment of MDA-MB-231 cells with ZER and ZER-NLC caused significant (𝑃 < 0.05) downregulation of Bcl-2 and Bcl-xL proteins and PCNA, while the Bax protein was upregulated and cytochrome c protein expression increased (Figure 9). The relative density of Bax and cytochrome c bands in both ZER- and ZER-NLCtreated groups increased with duration of treatment, whereas the relative density of Bcl-2, Bcl-xL, and PCNA decreased in comparison with their control (untreated) groups (Tables S5 and S6). Cytochrome c, released by the mitochondria, is responsible for the activation of caspase-3 and caspase-9. It is suggested that the anticancer effect of ZER and ZER-NLC is through the upregulation of Bax, cleaving poly(adenosine diphosphate-ribose) polymerase (PARP) with concurrent downregulation of Bcl-2 [14]. The apoptosis of MDA-MB231 cells induced by ZER and ZER-NLC that occurs through the intrinsic pathway is the result of increased permeability of mitochondrial membrane and release of cytochrome c that subsequently activates proapoptotic Bax and inhibits antiapoptotic Bcl-2 and Bcl-xL. These results are consistent with that obtained by a previous study [23]. Therefore, the study shows that ZER and ZER-NLC induced intrinsic apoptotic pathway via downregulation of antiapoptotic and upregulation of apoptotic proteins facilitated the permeabilization of outer mitochondrial membrane and release of cytochrome c. This event activates caspase-9, followed by activation of caspase-3 and cleavage of PARP [23]. Concurrently, ZER and ZER-NLC treatments decreased PCNA resulting in suppression of cancer cell proliferation [28].

protein expressions. Loading of ZER into NLC will increase the bioavailability of the insoluble ZER in the treatment of cancers.

4. Conclusion

[5] H.-Y. Weng, M.-J. Hsu, C.-C. Wang et al., “Zerumbone suppresses IKK𝛼, Akt, and FOXO1 activation, resulting in apoptosis of GBM 8401 cells,” Journal of Biomedical Science, vol. 19, no. 1, article 86, 2012.

This study shows that loading of ZER into NLC did not compromise the antiproliferative effect of ZER. Both ZER and ZER-NLC significantly induced apoptosis via the intrinsic pathway in time-dependent manner. The proposed mechanism of apoptosis of cancer cells induced by ZER and ZERNLC is via activation of caspase-9 and caspase-3, inhibition of antiapoptotic protein, and stimulation of proapoptotic

Conflict of Interests The authors declare that there is no conflict of interests regarding the publication of this paper.

Acknowledgments This research was granted by National Cancer Council (MAKNA) and Institute of Bioscience/Universiti Putra Malaysia. The authors would like to appreciate their kind help and support.

References [1] E. K. O. Ng, R. Li, V. Y. Shin, J. M. Siu, E. S. K. Ma, and A. Kwong, “MicroRNA-143 is downregulated in breast cancer and regulates DNA methyltransferases 3A in breast cancer cells,” Tumor Biology, vol. 35, no. 3, pp. 2591–2598, 2014. [2] V. P. Gullo and D. E. Hughes, “Exploiting new approaches for natural product drug discovery in the biotechnology industry,” Drug Discovery Today: Technologies, vol. 2, no. 3, pp. 273–278, 2005. [3] J. Chane-Ming, R. Vera, and J. C. Chalchat, “Chemical composition of the essential oil from rhizomes, leaves and flowers of Zingiber zerumbet Smith from Reunion Island,” Journal of Essential Oil Research, vol. 15, no. 3, pp. 202–205, 2003. [4] M. N. Somchit, J. H. Mak, A. A. Bustamam et al., “Zerumbone isolated from Zingiber zerumbet inhibits inflammation and pain in rats,” ” Journal of Medicinal Plants Research, vol. 6, pp. 177– 180, 2012.

[6] S. I. Abdel Wahab, A. B. Abdul, A. S. Alzubairi, M. Mohamed Elhassan, and S. Mohan, “In vitro ultramorphological assessment of apoptosis induced by zerumbone on (HeLa),” Journal of Biomedicine and Biotechnology, vol. 2009, Article ID 769568, 10 pages, 2009.

8

10 [7] S. I. Abdelwahab, A. B. Abdul, S. Mohan et al., “Zerumbone induces apoptosis in T-acute lymphoblastic leukemia cells,” Leukemia Research, vol. 35, no. 2, pp. 268–271, 2011. [8] J.-U. A. H. Junghanns and R. H. M¨uller, “Nanocrystal technology, drug delivery and clinical applications,” International Journal of Nanomedicine, vol. 3, no. 3, pp. 295–309, 2008. [9] K. T. Savjani, A. K. Gajjar, and J. K. Savjani, “Drug solubility: importance and enhancement techniques,” ISRN Pharmaceutics, vol. 2012, Article ID 195727, 10 pages, 2012. [10] H. S. Rahman, A. Rasedee, C. W. How et al., “Zerumboneloaded nanostructured lipid carriers: preparation, characterization, and antileukemic effect,” International Journal of Nanomedicine, vol. 8, pp. 2769–2781, 2013. [11] A. Patidar, D. S. Thakur, P. Kumar, and J. Verma, “A review on novel lipid based nanocarriers,” International Journal of Pharmacy and Pharmaceutical Sciences, vol. 2, no. 4, pp. 30–35, 2010. [12] E. B. Souto, S. A. Wissing, C. M. Barbosa, and R. H. M¨uller, “Development of a controlled release formulation based on SLN and NLC for topical clotrimazole delivery,” International Journal of Pharmaceutics, vol. 278, no. 1, pp. 71–77, 2004. [13] A. Chinsriwongkul, P. Chareanputtakhun, T. Ngawhirunpat et al., “Nanostructured lipid carriers (NLC) for parenteral delivery of an anticancer drug,” AAPS PharmSciTech, vol. 13, no. 1, pp. 150–158, 2012. [14] H. S. Rahman, A. Rasedee, A. B. Abdul et al., “Zerumboneloaded nanostructured lipid carrier induces G2/M cell cycle arrest and apoptosis via mitochondrial pathway in a human lymphoblastic leukemia cell line,” International Journal of Nanomedicine, vol. 9, no. 1, pp. 527–538, 2014. [15] J. Pardeike, A. Hommoss, and R. H. M¨uller, “Lipid nanoparticles (SLN, NLC) in cosmetic and pharmaceutical dermal products,” International Journal of Pharmaceutics, vol. 366, no. 1-2, pp. 170– 184, 2009. [16] S. Selvamuthukumar and R. Velmurugan, “Nanostructured lipid carriers: a potential drug carrier for cancer chemotherapy,” Lipids in Health and Disease, vol. 11, article 159, 2012. [17] S. A. S. Sakinah, S. Tri Handayani, and L. P. A. Hawariah, “Zerumbone induced apoptosis in liver cancer cells via modulation of Bax/ Bcl-2 ratio,” Cancer Cell International, vol. 7, article 4, 2007. [18] S. I. Abdelwahab, A. B. Abdul, Z. N. M. Zain, and A. H. A. Hadi, “Zerumbone inhibits interleukin-6 and induces apoptosis and cell cycle arrest in ovarian and cervical cancer cells,” International Immunopharmacology, vol. 12, no. 4, pp. 594–602, 2012. [19] S. Syam, A. B. Abdul, M. A. Sukari, S. Mohan, S. I. Abdelwahab, and T. S. Wah, “The growth suppressing effects of girinimbine on hepg2 involve induction of apoptosis and cell cycle arrest,” Molecules, vol. 16, no. 8, pp. 7155–7170, 2011. [20] A. B. H. Adbul, A. S. Al-Zubairi, N. D. Tailan et al., “Anticancer activity of natural compound (Zerumbone) extracted from Zingiber zerumbet in human HeLa cervical cancer cells,” International Journal of Pharmacology, vol. 4, no. 3, pp. 160–168, 2008. [21] A. Deorukhkar, N. Ahuja, A. Mercado et al., “Zerumbone, a sesquiterpene from southeast Asian edible ginger sensitizes colorectal cancer cells to radiation therapy,” International Journal of Radiation Oncology, Biology, Physics, vol. 78, no. 3, supplement, p. S654, 2010.

Journal of Nanomaterials [22] J. A. Pietenpol and Z. A. Stewart, “Cell cycle checkpoint signaling: cell cycle arrest versus apoptosis,” Toxicology, vol. 181182, pp. 475–481, 2002. [23] A. Sehrawat, J. A. Arlotti, A. Murakami, and S. V. Singh, “Zerumbone causes Bax- and Bak-mediated apoptosis in human breast cancer cells and inhibits orthotopic xenograft growth in vivo,” Breast Cancer Research and Treatment, vol. 136, no. 2, pp. 429–441, 2012. [24] N. O’Donovan, J. Crown, H. Stunell et al., “Caspase 3 in breast cancer,” Clinical Cancer Research, vol. 9, no. 2, pp. 738–742, 2003. [25] H. Zhao, Y.-H. Lo, L. Ma et al., “Targeting tyrosine phosphorylation of pcna inhibits prostate cancer growth,” Molecular Cancer Therapeutics, vol. 10, no. 1, pp. 29–36, 2011.

Journal of Nanomaterials

Composition Comments 1. Please check and confirm the author(s) first and last names and their order which exist in the last page. 2. Please provide valid postal code(s) to the address(es) lacking ones. 3. There is a difference between the manuscript and the new electronic version in Figures 1, 2, 4, and 8 and we followed the new electronic version. Please check. 4. We redrew some parts in Figures 5, 6, and 7. Please check. 5. We made the highlighted change(s) in the caption of Figure 4 for the sake of clarity and correctness. Please check. 6. Please provide a short description (in paragraph style) of the Supplementary Material. This description along with the Supplementary Material will be available online. 7. Please check the correctness of the highlighted part. 8. We made the highlighted change(s) for the sake of clarity and correctness. Please check.

11

Author(s) Name(s) It is very important to confirm the author(s) last and first names in order to be displayed correctly on our website as well as in the indexing databases: Author 1 Given Names: Mahnaz Last Name: Hosseinpour

Author 5 Given Names: Swee Keong Last Name: Yeap

Author 2 Given Names: Ahmad Bustamam Last Name: Abdul

Author 6 Given Names: Negin Last Name: Ahmadi

Author 3 Given Names: Heshu Sulaiman Last Name: Rahman

Author 7 Given Names: Hemn Hassan Last Name: Othman

Author 4 Given Names: Abdullah Last Name: Rasedee

Author 8 Given Names: Max Stanley Last Name: Chartrand

It is also very important for each author to provide an ORCID (Open Researcher and Contributor ID). ORCID aims to solve the name ambiguity problem in scholarly communications by creating a registry of persistent unique identifiers for individual researchers. To register an ORCID, please go to the Account Update page (http://mts.hindawi.com/update/) in our Manuscript Tracking System and aer you have logged in click on the ORCID link at the top of the page. is link will take you to the ORCID website where you will be able to create an account for yourself. Once you have done so, your new ORCID will be saved in our Manuscript Tracking System automatically.