Accepted Manuscript Gold-Nanorods-siRNA Nanoplex for Improved Photothermal Therapy by Gene Silencing Bei-Ke Wang, Xue-Feng Yu, Jia-Hong Wang, Zhi-Bin Li, Peng-Hui Li, Huaiyu Wang, Li Song, Paul K. Chu, Chengzhang Li PII:

S0142-9612(15)00921-7

DOI:

10.1016/j.biomaterials.2015.11.025

Reference:

JBMT 17202

To appear in:

Biomaterials

Received Date: 1 August 2015 Revised Date:

26 October 2015

Accepted Date: 12 November 2015

Please cite this article as: Wang B-K, Yu X-F, Wang J-H, Li Z-B, Li P-H, Wang H, Song L, Chu PK, Li C, Gold-Nanorods-siRNA Nanoplex for Improved Photothermal Therapy by Gene Silencing, Biomaterials (2015), doi: 10.1016/j.biomaterials.2015.11.025. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT

Gold-Nanorods-siRNA Nanoplex for Improved Photothermal Therapy by Gene Silencing

RI PT

Bei-Ke Wanga,1, Xue-Feng Yub*, Jia-Hong Wangc, Zhi-Bin Lib, Peng-Hui Lib,e, Huaiyu Wangb, Li Songd, Paul K. Chue, Chengzhang Lia*

The State Key Laboratory Breeding Base of Basic Science of Stomatology,

SC

a

M AN U

Hubei-MOST & Key, Laboratory of Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, P. R. China b

Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, P. R. China

d

School of Physics and Technology, Wuhan University, Wuhan 430072, P. R. China

TE D

c

Department of Stomatology, The Second Affiliated Hospital to Nanchang University, Nanchang 330006, P. R. China

Department of Physics and Materials Science, City University of Hong Kong, Tat

EP

e

AC C

Chee Avenue, Kowloon, Hong Kong, China

*Corresponding authors: Tel: +86-87686212; Fax: +86-87873260 (C. Li) E-mails: [email protected] (C. Li); [email protected] (X. F. Yu)

1

ACCEPTED MANUSCRIPT Abstract

Nanomaterials-mediated photothermal therapy (PTT) often suffers from the

RI PT

fundamental cellular defense mechanism of heat shock response which leads to therapeutic resistance of cancer cells and reduces the therapeutic efficacy.

Herein, a

gold nanorods (GNRs)-siRNA platform with gene silencing capability is produced to After surface modification, the GNRs show the ability

SC

improve the PTT efficiency.

M AN U

to deliver siRNA oligos targeting BAG3 which is an efficient gene to block the heat-shock response.

The synthesized GNRs-siRNA nanoplex exhibits excellent

ability in the delivery of siRNA into cancer cells with high silencing efficiency which is even better than that of commercial Lipofectamine 2000.

The in vitro and in vivo

TE D

studies demonstrate the ability of the GNRs-siRNA nanoplex to sensitize the cancer cells to PTT under moderate laser irradiation by down-regulating the increased BAG3 expression and enhancing apoptosis.

The GNRs-siRNA mediated PTT has large

EP

potential in clinical cancer therapy due to the elimination of therapeutic resistance and It also

AC C

enhanced photothermal therapeutic efficacy by means of gene silencing. suggests an efficient platform for gene delivery and controllable gene therapy.

Keywords: gold nanorods; photothermal therapy; RNA interference; oral cancer; biomedical applications 2

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

TOC Graphic

3

ACCEPTED MANUSCRIPT 1. Introduction

Nanomaterials-mediated photothermal therapy (PTT) by means of near-infrared Owing to small light

RI PT

(NIR) illumination is an emerging tool in cancer therapy.

scattering and absorption from intrinsic chromophores in tissues, NIR light can penetrate tissues with sufficient intensity and high spatial precision [1-3]. Therefore,

SC

PTT treatment provides an efficient approach to convert photon energy into cytotoxic

surgery is difficult [1,4,5].

M AN U

heat to destroy cancer cells with high selectivity, especially in vital regions where Compared with conventional cancer treatment

approaches, PTT with highly localized heat delivery is minimally invasive and rapid and can be combined with chemotherapy and drug/gene delivery.

Despite these In

TE D

advantages, the full potential of PTT is still hindered by some challenges.

particular, to achieve sufficient heating for complete killing of cancer cells, a high-power laser or large agent concentration has been suggested [6-8]. However, a

EP

high laser power or agent concentration produces safety risks including uncertain

AC C

cytotoxicity, unnecessary morbidity due to collateral damage, and lack of patient tolerance at a high temperature [9-11].

It is especially true for some special cancer

such as head and neck cancer due to their special anatomic sites, vital biological functions, and cosmetic requirements [12].

Therefore, it is imperative to develop a

general strategy to improve the PTT efficiency without a high laser power or large agent amount. Heat shock response is a mechanism to prevent cancer cells from hyperthermia 4

ACCEPTED MANUSCRIPT and has been shown to undermine the therapeutic efficacy of thermal therapy due to their cytoprotective and antiapoptotic effects, termed as thermoresistance [13-15]. Recent studies indicate that PTT can trigger the heat shock response in cancer cells

RI PT

consequently decreasing the therapeutic efficacy by suppressing apoptosis [16-19]. Several families of heat shock proteins (such as HSP70) and BAG3 (Bcl-2 associated athanogene domain 3, also known as Bis and CAIR) induced by the heat shock

The application of RNA interference to cancer

M AN U

role in the thermoresistance [20-23].

SC

response exert central cytoprotective effects preventing cell death and play a major

therapy has recently attracted attention as a promising therapeutic modality [24,25]. By interfering with the expression of specific genes, the small-interfering RNA (siRNA) acts as an effective vehicle in RNA interference thereby suggesting a

TE D

possible strategy to inhibit the heat shock response and renders the cancer cells more susceptible to PTT by silencing the expression of HSPs or BAG3. Among the various nano-agents for PTT, gold nanorods (GNRs) with the unique

EP

surface plasmon resonance (SPR) bands have been studied extensively on account of

AC C

their strong NIR absorption and high photothermal conversion efficiency [26], and PTT mediated by GNRs has been suggested to be a promising treatment for oral cancer [27].

Moreover, possessing a large surface area to volume ratio and being

easily and controllably surface functionalized, GNRs are attractive nanocarriers for different types of drugs to cells [28-31], photosensitizers [27,32-35], and small biomolecules [36,37]. The effectiveness of GNRs in a gene delivery system has been demonstrated in vitro and also preliminarily in vivo [38-45]. 5

It has further been

ACCEPTED MANUSCRIPT shown that GNRs can penetrate the blood brain barrier and silence expression of DARPP-32 in the delivery system for specific siRNAs into the neuron cells [46]. Owing to the versatility, GNRs can play an important role in a single multifunctional With regard to

RI PT

nanotherapeutic platform for combined PTT and gene delivery.

cancer therapy, it has shown that different kinds of synthesized multifunctional GNRs-siRNA nanoplex can be applied to treat different types of cancer such as breast

In these studies, GNRs-siRNA complexes not only function as nanovectors

M AN U

[51].

SC

cancer [38,47-49], pancreatic adenocarcinoma [50], as well as head and neck cancer

[52-54] for siRNA and chemotherapy agents delivery [55-57], but also act as photothermal or imaging agents for theranostic purposes (summarized in Table S1) [55-57].

TE D

In this work, the GNRs-siRNA nanoplex is utilized in a therapeutic platform to introduce gene silencing technology to improve the PTT efficacy (see Scheme 1). The CTAB-GNRs are prepared by a seed-mediated growth method followed by surface

modification

EP

sequential

with

negatively-charged

poly(sodium

AC C

4-styrenesulfonate) (PSS) and positively-charged poly(diallyldimethylammonium chloride) (PDDAC) to form the GNRs/PSS/PDDAC with cationic charges.

The

GNRs/PSS/PDDAC can complex with anionic charged siRNA oligos targeting BAG3 via electrostatic interaction to form the GNRs-siRNA nanoplex.

The nanoplex

shows the ability to silence the increased BAG3 expression induced by a high temperature both on the mRNA and protein levels and produces good NIR-activated photothermal properties.

Their ability to silence the increased BAG3 expression 6

ACCEPTED MANUSCRIPT attenuates the heat shock response induced by PTT sensitizing the cancer cells to easier PTT-mediated cell death.

Our in vitro and in vivo studies demonstrate the

ability of the GNRs-siRNA nanoplex to overcome the thermoresistance during PTT

TE D

M AN U

SC

RI PT

and produce the maximal tumoricidal effects with minimal damage to normal tissues.

AC C

platform.

EP

Scheme 1. Schematic illustration of the design of GNRs-siRNA in the improved PTT

7

ACCEPTED MANUSCRIPT 2. Materials and Methods 2.1 Materials The Dulbecco's modified Eagle's medium (DMEM) was obtained from Hyclone

USA).

RI PT

(UT, USA) and fetal bovine serum (FBS) was purchased from Gibco (Carlsbad, CA, MTT (3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide),

serum-free

medium

(opti-MEM),

(4 ′ ,6-diamidino-2-phenylindole),

DAPI

The BAG3 siRNA and fluorescently labeled siRNA

M AN U

Invitrogen (Carlsbad, CA).

SC

lipofectamine 2000 (lipofectamine), and Trizol reagent were purchased from

(BAG3 siRNAFAM) were synthesized by GenePharma (Shanghai, China). sequences

were

as

follows:

AAGGUUCAGACCAUCUUGGAA-3’.

BAG3

siRNA:

The 5’-

The primary antibodies against human

TE D

BAG3 were obtained from proteintech (Wuhan, China), HSP27, HSP60, HSP70, and HSP90 were purchased from ABclonal (Wuhan, China), cleaved PARP was purchased from Cell Signaling Technology (Danvers, MA, USA), and GAPDH was

EP

purchased from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA).

Chloroauric acid

AC C

(HAuCl4·4H2O, 99.99%), sodium chloride (NaCl, 96.0%), and hydrochloric acid (HCl, 36–38%) were bought from Sinopharm Chemical Reagent Co. Ltd. (Shanghai, China).

Hexadecyltrimethylammonium

bromide

(CTAB,

99.0%),

sodium

borohydride (NaBH4, 96%), silver nitrate (AgNO3, 99.8%), L-ascorbic acid (99.7%), Poly(sodium

4-styrenesulfonate)

(PSS,

MW

~70,000

g/mol),

and

Poly(diallyldimethylammonium chloride) solution 20 wt% in H2O (PDDAC, MW 100,000~200,000 g/mol) were obtained from Aldrich (America). 8

All the chemicals

ACCEPTED MANUSCRIPT were used as received without purification.

Ultrapure water with a resistivity of

about 18.25 MΩ·cm was used as the solvent in the experiments.

RI PT

2.2 Synthesis of GNRs-siRNA The GNRs were synthesized in an aqueous solution using a seed-mediated growth method as previously described [58].

For preparation of the gold seed

SC

particles with a size of 3-4 nm, 5 ml of 0.5 mM HAuCl4 were mixed with 5 ml of 0.2

M AN U

M CTAB and 600 µl of freshly prepared ice-cold 10 mM NaBH4 were immediately injected into the solution under vigorously stirring. to bright brown.

The color changed from yellow

The seed solution was left at least 2 h before use.

In the GNR

synthesis, 1.2 ml of 5 mM HAuCl4 and 15 µl of 0.1 M AgNO3 were added to 6 ml of

TE D

0.2 M CTAB and then 15 µl of 1.2 M HCl and 720 µl of 10 mM ascorbic acid were added and gently swirled until the color changed from dark orange to colorless. Afterwards, 12 µl of the seed solution were added.

Finally, the GNRs were collected by

EP

mixed and left undisturbed for 8 h.

AC C

centrifugation at 12,000 rpm for 15 min.

The supernatant was removed and

precipitate was resuspended in ultrapure water. estimated to be about 0.65 nM.

The resulting solution was gently

The GNRs concentration was

In the synthesis of GNRs-siRNA, GNRs were first

coated with PSS by the method previously reported [59].

2 ml of 10 mg/mL PSS

dispersed in 1 mM NaCl and 1 ml of 10 mM NaCl were added to 10 ml of GNRs and stirred for 1 h at room temperature.

A centrifugation cycle of 10,000 rpm for 10 min

was performed to remove the excess PSS and NaCl, obtaining PSS-coated GNRs 9

ACCEPTED MANUSCRIPT (GNR/PSS).

The PDDAC was further coated using a similar method.

The

prepared cationic GNRs/PSS/PDDAC (GNRs) were mixed with different amounts of siRNAs by pipetting and incubated for 30 min at room temperature before the

RI PT

operation to form the nanoplex electrostatically.

2.3 Characterizations

SC

Transmission electron microscopy (TEM) was performed on a JEOL 2010 (HT) The zeta

M AN U

transmission electron microscope at an accelerating voltage of 200 kV.

potentials of the samples were determined on a Zeta sizer (Nano ZS90, Malvern Instruments, UK) at 25 ○C and the absorption spectra were taken on a TU-1810 UV-Vis-NIR spectrophotometer (Purkinje General Instrument Co. Ltd. Beijing,

TE D

China).

2.4 Agarose gel electrophoresis

EP

The GNRs-siRNA nanoplexes with different GNRs/siRNA ratios were prepared

AC C

by addition of the 1.5 µg BAG3 siRNA to the assigned amounts of 0.65 nM GNRs solution (ranging from 0-27 µl) for 30 mins at room temperature.

After incubation,

the RNase free water was added to the GNR-siRNA nanoplexes with different binding ratios to form the sample solution (30 µl total).

The solutions were centrifuged and

the supernatants were loaded onto 1% agarose gels containing ethidium bromide (0.5 mg/ml) in the 1 × TAE buffer (Tris-acetate-EDTA buffer) and electrophoresed at 100

10

ACCEPTED MANUSCRIPT V for 20 min.

The gel was imaged under UV light on a GelDoc 2000 imager system

(Bio-Rad, Munich, Germany).

RI PT

2.5 Photothermal conversion measurement The photothermal efficiency was measured on a homemade setup as described A 1 cm quartz cuvette containing 2 ml of the sample was covered

with a foam cap.

The cuvette was clamped on the top part above the sample surface

SC

previously [27].

stirrer.

M AN U

and the bottom of the cuvette was kept at approximately 0.5 cm above the magnetic A fiber-coupled continuous semiconductor diode laser (810 nm,

KS-810F-8000, Kai Site Electronic Technology Co., Ltd. Shaanxi, China) with a power density of 2.7 W·cm-2 and beam diameter of approximately 0.5 cm illuminated A digital thermometer (TX3001, Xintengxing, Wuhan, China) was used

TE D

the cuvette.

to monitor the temperature change.

The head of the thermometer was completely

submerged in the solution and carefully prevented from direct illumination by the Each sample in the cuvette was irradiated for 20 min under rigorous stirring

EP

laser.

AC C

and the temperature was recorded per 30 s.

2.6 Cell culture

The human oral squamous cell carcinoma cell line Cal-27 (CRL-2095, ATCC)

was obtained from the Shanghai Research Institute of Stomatology, Affiliated Ninth People’s Hospital, Shanghai Jiaotong University, Shanghai, China.

11

The cells were

ACCEPTED MANUSCRIPT maintained in DMEM supplemented with 10% FBS in a humidified atmosphere of 5% CO2 at 37 °C.

RI PT

2.7 Selection of the moderate laser irradiation power density for PTT To select the optimal light irradiation conditions for PTT, the cell viability of Cal-27 cells in the presence of GNRs irradiated with different laser power density was The GNRs were diluted in the complete DEME 8 × 103 Cal-27 cells/well were

M AN U

medium to achieve the concentration of 97.5 pM.

SC

assessed by the MTT assay.

seeded onto 96-well plates and incubated overnight.

Afterwards, the cells were

treated with the prepared GNRs medium for 24 h and irradiated by the 810 nm laser at power densities of 0 J·cm-2, 200 J·cm-2, 400 J·cm-2, 600 J·cm-2, 800 J·cm-2, and 1,000 The untreated cells were irradiated with a power density of 1000 J·cm-2.

TE D

J·cm-2.

Afterwards, the cells were incubated in a complete medium up to 24 h. Finally, the cell

viability

was

determined

using

EP

3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT, Sigma-Aldrich) The formula, (ODtreated/ODcontrol) × 100%, was

AC C

assay as previously described [60].

used to calculate the cell viability and the experiments were conducted in triplicate. The cells without any treatment serve as the viability control.

2.8 Heat shock response detection To confirm the occurrence of heat shock response in PTT at a moderate laser power, real-time PCR and western blots were performed to determine the expression 12

ACCEPTED MANUSCRIPT of HSP27, HSP60, HSP70, HSP90, BAG3 at different time points after the photothermal treatment.

Briefly, the Cal-27 cells were precultured on 24-well plates The cells were treated

with the 97.5 pM GNRs medium and incubated for 24 h.

After incubation, the

RI PT

at the confluence of 70% and allowed to adhere overnight.

treated cells were illuminated by the 810 nm laser with a power density of 600 J·cm-2. The irradiated cells were incubated in the complete medium and collected at 0 h, 2 h,

2.9 Cell uptake of GNRs-siRNA

M AN U

SC

4 h, 8 h, and 12 h for real time PCR and western blots.

According to the fluorescence of BAG3 siRNAFAM (492 nm excitation and 518 nm emission), the cellular uptake of GNR-siRNA was analyzed using both

TE D

fluorescence microscopy and flow cytometry.

In fluorescence microscopy, the

Cal-27 cells were seeded on glass coverslips at 40–50% confluency on 24-well plates and incubated overnight before transfection.

On the next day, the GNRs (97.5 pM),

EP

naked siRNAFAM (0.28 pM), and GNR-siRNAFAM nanoplexes (containing 97.5 pM

AC C

GNRs and 0.28 pM siRNAFAM) were added to the complete DMEM medium and incubated with cells for 6 h, respectively.

The untreated cells served as the negative

control and cells transfected with BAG3 siRNAFAM by lipofectamine 2000 (invitrogen) served as the positive control following the manufacturer‘s instructions. Thereafter, the cells were rinsed twice with 1 × cold phosphate buffered saline (PBS) and fixed with 4% paraformaldehyde for 15 min at room temperature.

After

washing with PBS buffer solution, the cells were mounted using an aqueous mounting 13

ACCEPTED MANUSCRIPT medium with DAPI (Invitrogen).

The cells were imaged under the the Leica

DM4000B fluorescence microscope (Leica, Nussloch, Germany).

In flow cytometry,

the Cal-27 cells were seeded on 6-well plates at a density of 5 × 105 cells/ml and

fluorescent microscopy.

After attachment, the cells were transfected for

RI PT

incubated under standard conditions.

Six hours after treatment, the cells were harvested and

analyzed on the FACS Calibur flow cytometer (Becton Dickinson, Franklin Lakes,

SC

NJ) at the excitation wavelength of 488 nm and emission wavelength range of

M AN U

515-545 nm and analyzed by the Cell Quest Software (Becton Dickinson).

2.10 Evaluation of gene silencing efficiency of BAG3 siRNA via different vectors: Before transfection, the Cal-27 cells were seeded on 24-well plates at the density 1.875 µg of the BAG3 siRNA

TE D

of 5 × 104 cells/well and allowed to adhere overnight.

oligos were added to one aliquot of the 48.75 fmol GNRs in the eppendorf tubes and incubated for 30 min at room temperature to form the GNR-siRNA nanoplexes.

The

EP

GNRs,siRNA, and prepared GNR-siRNA were gently dipped into the complete

AC C

DMEM medium making sure that the GNRs concentration and siRNA concentration in the final medium was 97.5 pM and 0.28 pM, respectively.

After aspirating the

media, the cells were treated with the prepared GNRs medium, siRNA medium, and GNRs-siRNA medium for 24 h according to the indicated treatment. cells were regarded as the control.

The untreated

Lipofectamine 2000 was used as the positive

control to transfect BAG3 siRNA and was performed according to the manufacturer’s instruction.

Briefly, 3 µl Lipofectamine diluted in 50 µl opti-MEM were mixed with 14

ACCEPTED MANUSCRIPT 5 µg of the siRNA diluted in 50 µl of the opti-MEM.

The mixture was incubated at

37 °C for 20 min and added to the wells producing a final siRNA concentration of 0.375 pM for the positive control.

Six hours after incubation, the medium was On the following

RI PT

exchanged by the 97.5 pM GNRs medium and incubated for 24 h.

day, all the groups except control were washed by PBS and irradiated by the 810 nm laser with a power density of 600 J·cm-2.

Eight hours after irradiation, all the cells

SC

were harvested for mRNA isolation and protein extraction and the expressions of

and western blotting, respectively.

M AN U

BAG3 and GAPDH on the mRNA and protein levels were analyzed by real time PCR

2.11 Analysis of cytotoxicity and apoptosis induced by combined treatment of

TE D

GNRs-siRNA in vitro

The efficacy of the GNR-siRNA in PTT of cancer cells irradiated with a moderate-power laser was evaluated by the MTT assay, cell apoptotic detection, and

EP

western blotting for caspase-3.

In the MTT assay, the Cal-27 cells on 96-well plates

AC C

were either left untreated or treated with fresh mediea containing siRNA oligos, GNRs, and GNRs-siRNA at the siRNA concentration of 0.28 pM and GNRs concentration of 97.5 pM for 24 h.

In the positive group, 0.375 pM siRNA delivered

by lipofectamine 2000 was added to the cells, incubated for 6 h at 37 °C, and then was replaced by the 97.5 pM GNRs medium.

810 nm laser irradiation with a power

density of 600 J·cm-2 was performed on most cells at the end of incubation. cells without treatment were used as a control. 15

The

After incubation for 24 h, the cell

ACCEPTED MANUSCRIPT viability was assessed by the MTT assay as previously described.

In analyzing the

apoptotic response, the cells were treated in the same manner as in the MTT assay and harvested for apoptosis and necrosis assay and western blotting for caspase-3.

The

RI PT

apoptosis and necrosis assay was performed as previously reported and finally analyzed on a flow cytometer (Becton Dickinson, Franklin Lakes, NJ) using the Cell

SC

Quest software (Becton Dickinson) [27].

M AN U

2.12 Western blots

The harvested cells were lysed in M-PER (Pierce Chemical, Rockford, IL) containing Halt Protease Inhibitor cocktail (Pierce).

The denatured protein was

collected and the concentration was determined by the BCA Protein Assay Kit Subsequently, 20 µg of protein were separated using a 10% sodium

TE D

(Pierce).

dodecylsulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and transferred onto polyvinylidene fluoride (PVDF) membranes (Millipore Corporation, Billerica, MA).

EP

The membranes were blocked by 5% non-fat dry milk for 1 h under room temperature The blocked membranes were incubated overnight

AC C

to prevent nonspecific binding.

at 4 °C with the primary anti-HSP27 (1:2000, A0240, ABclonal), anti-HSP60 (1:2000, A0969, ABclonal), anti-HSP70 (1:2000, A0284, ABclonal), anti-HSP90 (1:2000, A0365, ABclonal), anti-BAG3 (1:2000, 10599-1-AP, proteintech), cleaved PARP (1:1000, #5625, CST), and anti-GAPDH (1:5000, sc-365062, Santa Cruz) antibodies. The immunoblots were probed with Horseradish peroxidase-conjugated goat anti-rabbit or antimouse secondary antibodies (Pierce) for 1 h at room temperature. 16

ACCEPTED MANUSCRIPT Finally, the protein were visualized with a chemiluminescence kit (Pierce) and photographed.

RI PT

2.13 RNA extraction and real-time quantitative PCR The total RNA was isolated from the cells using TRIzol Reagent (Invitrogen). 1 µg total RNA from each sample was reverse transcribed to cDNA (20 µl) primed by

SC

oligo (dT) using M-MuLV reverse transcriptase (Fermentas, Glen Burnie, MD).

The quantitative PCR

M AN U

mRNA was analyzed quantitatively by real-time PCR.

The

reaction was carried out in a 20 µl reaction volume containing 2 µl cDNA as a template, 10 µl 2 × Maxima SYBR Green PCR master mix (Takara, Kyoto, Japan), and 0.8 µl of the primer mix (10 µm forward primer, 10 µm reverse primer).

All the

TE D

reactions were conducted in triplicate on an ABI 7500 Real-Time PCR System (Applied Biosystems, Foster City, CA).

The expression was normalized to GAPDH

which was used as the endogenous control in each experiment.

The primer

AC C

EP

nucleotide sequences for PCR are presented in Table S2.

2.14 Establishment of Tumor Xenografts Model The male BALB/c nude mice (4-6 weeks old, 18-20 g, 40 animals) were

obtained from Hunan SJA Laboratory Animal Co., Ltd. (Hunan, China). animals were housed in sterilized cages and a 12 h light/dark cycle.

The

All the

experiments were approved by the ethics committee of Wuhan University and were performed according to institutional animal use and care regulations. 17

2 × 107 Cal-27

ACCEPTED MANUSCRIPT cells in 200 µl PBS were subcutaneously injected into the right flank of mice to initiate tumor growth.

The longest and shortest dimension of tumors were detected

by a digital calipers.

After the longest dimension of the tumor reached 5 mm, the

2.15 Distribution of GNR-siRNA inside the tumor

RI PT

tumor bearing mice were chosen for in vivo treatment.

SC

Twelve mice with established tumors were randomly divided into 4 groups (n = In the

M AN U

3 per group): control group, siRNA group, GNRs group, GNRs-siRNA group.

control group, the mouse was intratumorally injected with sterilized 10 mM PBS solusion.

In the other three group, the mice were intratumorally injected with the

siRNAFAM dispersion (15.2 pM siRNAFAM), GNRs dispersion (5.3 nM GNRs), and

respectively.

TE D

GNR-siRNAFAM dispersion (containing 5.3 nM GNRs and 15.2 pM siRNAFAM), The injectied volume of each type of dispersion was 200 µL per 50

mm3 tumor volume.

After 6 h post-administration, all the mice were sacrificed and The ex vivo tumors were subsequently

EP

the tumors were extracted immediately.

AC C

embedded in the Tissue-Tek OCT (Sakura Finetek), frozen in liquid nitrogen, and cut into sections 5 µm thick on the CM1850 UV cryostat (Leica Microsystems).

The

sections were mounted with DAPI containing mounting solution (Invitrogen) and imaged by the Leica DM4000B fluorescence microscope (Leica, Nussloch, Germany) at excitation wavelengths of 405 nm (DAPI) and 488 nm (siRNAFAM).

The cell

uptake of siRNAFAM in vivo was quantified by calculating the percent of the cells that took up siRNA among the total cells in each group. 18

ACCEPTED MANUSCRIPT 2.16 In vivo photothermal treatment After the longest dimension of the inoculated tumors reached ~5 mm, the mice were randomized into 5 groups (n = 10 per group).

The mice in groups 3 and 4 were

RI PT

the naïve state and served as the control.

The mice in group 1 were kept in

intratumorally injected with the siRNA solution (15.2 pM , 200 µL per 50 mm3 tumor volume) and GNRs solution (5.3 nM, 200 µL per 50mm3 tumor volume), respectively.

SC

The mice in groups 2 and 5 were intratumorally injected with the prepared

M AN U

GNRs-siRNA solution (200 µL per 50 mm3 tumor volume) at the same concentration of siRNA and GNRs as in the group 3 and group 4, respectively.

After 24 h

post-injection, the mice in groups 3, 4, and 5 were irradiated by the 810 nm near-infrared laser (Gigaa Optronics Technology Co., Ltd., Wuhan, China) with a Aftewards, the mice were returned to the animal

TE D

power density of 600 J·cm-2.

housing and 24 h later, three mice in each group were randomly selected for the BAG3 immunohistochemistry and TUNNEL assay.

EP

to measure the tumor growth volume.

The remaining mice were used

The largest and shortest tumor diameters were

AC C

determined every other day with the digital caliper.

The formula: V = 1/2 (L × W2)

was used to calculate the tumor volume, where L and W were the length (longest diameter) and width (shortest diameter), respectively.

The increased tumor percent

(Ti) was calculated according to the formula: Ti (%) = (DP - DC) / DC × 100%, where DC and DP were the tumor volumes before and after treatment, respectively. At day 18 after the treatment, the mice were euthanatized and the tumors were extracted and photographed. 19

ACCEPTED MANUSCRIPT

2.17 Immunohistochemistry and TUNEL assay Twenty four hours after the treatment, the collected tumors were fixed in 4%

RI PT

paraffin or frozen in liquid nitrogen for subsequent BAG3 immunohistochemical analysis and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL) assay.

In BAG3 immunohistochemistry, the slices were stained

A streptavidin-biotin-peroxidase complex with diaminobenzidine (DAB) as

M AN U

4 °C.

SC

with the rabbit anti-BAG3 polyclonal antibody (1:1000, Proteintech) overnight at

the peroxidase substrate was used to visualize immunoreactivity in accordance with the

manufacturer's

instructions

Peroxidase/DAB+, Rabbit/Mouse).

REAL

TM

Detection

System,

The stained sections were counterstained with

In the TUNEL assay, the slides were stained with the TUNEL

TE D

hematoxylin.

(Dako

technique on the in situ cell death detection kit (7 sea pharmtech, Shanghai, China) according to the manufacture’s instructions.

The fluorescent images were acquired

AC C

EP

on the Leica DM4000B fluorescence microscope (Leica, Nussloch, Germany).

2.18 Statistical analysis All the data were expressed as means ± standard deviations from three

independent experiments.

The One-way ANOVA and Student-Newman-Keuls were

used in the statistical analysis with p < 0.05 being considered to be statistically significant.

20

ACCEPTED MANUSCRIPT 3. Results and Discussion The CTAB-coated GNRs with an aspect ratio of about 4.2 were synthesized by a seed-mediated growth method [58].

Negatively-charged PSS was introduced to the

PSS-coated

GNRs

(GNRs/PSS)

[59].

Afterwards,

RI PT

surface of the GNRs followed by absorption of positively-charged PDDAC onto the the

positively-charged

GNRs/PSS/PDDAC possesses the ability to bind with negatively-charged BAG3 The optimal binding ratio of GNRs to BAG3 siRNA is

SC

siRNA electrostatically.

M AN U

investigated by the agarose gel electrophoresis assay (see Figure S1).

Figure 1 shows the characteristics of the products in the GNRs-siRNA synthesis. As shown in Figure 1a, the original CTAB-GNRs exhibit two SPR bands: a weak transverse SPR (TSPR) band at about 510 nm and a strong longitudinal SPR (LSPR) A slight red-shift is observed from the LSPR band in both the

TE D

band at about 808 nm.

PSS and PDDAC modification process.

Following complexation with BAG3 siRNA,

a further red-shift of about 4 nm in the LSPR band can be observed.

It is well

EP

known that the LSPR band peak shift from the GNRs results from the local refractive

AC C

index around the GNRs, which is sensitive to the changes on the surface of the GNRs [61].

Here, the redshift of the LSPR band is probably due to binding of siRNA with

the GNRs/PSS/PDDAC forming GNRs/PSS/PDDAC-siRNA (named GNRs-siRNA). Similar results have been reported showing that binding of biological molecules can induce the LSPR band shift from the GNRs [62,63].

In the process, no obvious

broadening in the LSPR band can be found thus demonstrating good dispersion of the products.

The zeta potentials in Figure 1b present more evidence about successful 21

ACCEPTED MANUSCRIPT modification of the GNRs in different stages, i.e. +43.9 mV (CTAB-GNRs), -33.0 mV (GNRs/PSS), and +38.0 mV (GNRs/PSS/PDDAC).

After incubation of the cationic

GNRs/PSS/PDDAC substrate with anionic BAG3 siRNA, the zeta potentials reverse

GNRs in the final GNRs-siRNA.

RI PT

from positive (+38.0 mV) to negative (-24.2 mV), indicating binding of siRNA with The TEM image of GNRs-siRNA in Figure 1c

shows that the GNR morphology is almost unchanged and no aggregation can be

SC

observed, further confirming that the modification and conjugation processes do not

AC C

EP

TE D

M AN U

affect the dispersion status of the GNRs.

Figure 1. Characteristics of the synthesis of GNRs-siRNA nanoplex. spectra

and

(b)

Zeta

potential

of

original

(a) Absorption

CTAB-GNRs,

GNRs/PSS/PDDAC, and GNRs/PSS/PDDAC-siRNA (GNRs-siRNA). 22

GNRs/PSS, (c) TEM

ACCEPTED MANUSCRIPT image of GNRs-siRNA.

(d) Temperature change curves of the aqueous solutions

containing CTAB-GNRs and GNRs-siRNA, and pure water irradiated by the 810 nm

RI PT

laser for 20 min.

The photothermal conversion ability of the GNRs-siRNA under 810 nm laser irradiation is examined by using the original CTAB-GNRs and pure water as the As shown in Figure 1d, the temperature of the CTAB-GNRs and

SC

control samples.

respectively.

M AN U

GNRs-siRNA increases by 38.1 oC and 37.5 °C after NIR light irradiation for 20 min, In contrast, the temperature of pure water only increases by 3.4 °C.

The results indicate that the GNRs-siRNA retains the good photothermal performance of the original GNRs.

The

TE D

The heat shock response in oral cancer cells induced by PTT is investigated.

photothermal effect of the GNRs on Cal-27 cells is first assessed by the MTT assay (see Figure S2).

The cytotoxic effect of the GNRs on cancer cells is hampered at an

EP

irradiation power density of 600 J·cm-2 (3.3 W·cm-2 for 3 mins), suggesting that the

AC C

heat shock response may be induced under these conditions.

The expression of heat

shock response related genes in the GNRs-treated cancer cells under NIR laser irradiation at 600 J·cm-2 is evaluated on both the transcription level (mRNA) and protein level by real time-PCR and western blots (see Figure 2).

As shown in Figure

2a, the mRNA expressions of HSP27, HSP60, HSP70, HSP90, and BAG3 increase significantly after irradiation for 2 h and then decrease gradually within 24 h.

BAG3

shows the largest relative increase in the mRNA expression in the Cal-27 cells and it 23

ACCEPTED MANUSCRIPT is almost ten times that of the control cells at 2 h.

The western blots results (see

Figure 2b) show that the protein expression of BAG3 is largely increased in the GNRs treated cancer cells at 4 h post irradiation, although the protein expression of HSP27 The results indicate

RI PT

shows the largest increase among the detected HSP proteins.

that the GNRs-mediated PTT induces obvious heat shock response in the cancer cells and BAG3 is the most obviously induced on the mRNA level and also increases

SC

largely on the protein level.

M AN U

BAG3 is chosen as the target gene for the following reasons.

First of all, among

the detected heat shock related proteins, the expression of BAG3 is strongly induced on the mRNA and increases greatly on the protein level, indicating its critical role in the heat shock response at least in PTT of oral cancer cells.

The RNA interference Here,

TE D

approach is based on the endogenous degradation of mRNA of the target gene.

our results demonstrate that BAG3 mRNA is the most induced among all the selected genes.

Although the HSP27 protein expression is higher than BAG3 in the GNRs

EP

treated cancer cells after laser irradiation, BAG3 is selected as the silencing target Secondly, BAG3 as an antiapoptotic protein

AC C

gene due to the better silencing effects.

is important to the pleiotropic effects [50-52].

Generally, BAG3 can bind with

HSP70 to inhibit the chaperone activity of HSP70 [50].

By means of BAG3 and

HSP70 multiprotein complex, many antiapoptotic modulators (such as anti-apoptotic Bcl-2 family members, Bcl-XL) as HSP70 client proteins can be stabilized and protected from proteasome-mediated degradation, thereby mitigating stress-induced apoptosis and sustaining cell survival to induce thermoresistance of the cancer cells. 24

ACCEPTED MANUSCRIPT There is emerging evidence that BAG3 also increases after chemotherapy to play a protective role against apoptosis and it therefore has been used as a potential therapeutic target to enhance chemotherapy [50].

In addition, BAG3 has been

RI PT

demonstrated to be involved in the stress response to X-ray along with HSP70 and p63 [51] indicating the role in the apoptosis resistance in radiotherapy.

Therefore, as

a versatile anti-apoptotic effector, silencing the expression or inhibiting the activity of

SC

BAG3 provides an effective strategy to improve the apoptotic response to Thirdly,

M AN U

hyperthermia [52], chemotherapy [53], and even radiotherapy [54].

elevated BAG3 expressions can be detected from some malignant tumors [52,55-57]. The specific expression and activity of BAG3 in neoplastic cells versus normal tissues have been observed [58].

In our study, a moderately higher BAG3 gene expression

TE D

in the oral cancer cells compared to normal cells is observed (data not shown) and hence, BAG3 is a favorable therapeutic candidate to photo-sensitize cancer cells in

AC C

EP

order to improve the therapeutic efficacy.

25

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

TE D

Figure 2. Heat shock response induced by PTT with GNRs.

(a) Relative mRNA

expression and (b) Protein expressions of HSP27, HSP60, HSP70, HSP90 and BAG3

EP

in Cal-27 cells for the indicated time after treatment with 97.5 pM GNRs for 24 h

AC C

followed by irradiation by the 810 nm laser at a power density of 600 J·cm-2.

Cell imaging experiments are performed to investigate the uptake ability of Cal-27

cancer cells for GNRs-siRNA (see Figure 3a).

To visualize the GNRs-siRNA

nanoplexes by fluorescence microscopy, siRNA labeled with the green fluorescent FAM (siRNAFAM) is used to conjugate the GNRs.

The bare GNRs

(GNRs/PSS/PDDAC) and siRNAs are the negative control samples whereas the lipo-siRNAFAM (siRNAFAM delivered by lipofectamine) are the positive samples. 26

ACCEPTED MANUSCRIPT Although GNRs are well-known fluorescence quenchers, it has been demonstrated that attachment of fluorescent dyes through polyelectrolyte spacer can produce obvious fluorescence [38,46,49].

In the present study, the GNRs used for binding

RI PT

siRNAFAM are coated with PSS and PDDAC layer-by-layer, and the emission spectrum of FAM partly overlaps the weak TSPR band of the GNRs. obtained GNRs-siRNA nanoplexes can emit green fluorescence.

Therefore, the

As shown in Figure

SC

3, such green fluorescence can be observed from the cancer cells incubated with the

M AN U

GNRs-siRNAFAM for 6 h indicating effective delivery of siRNAFAM into the cells. The uptake of GNRs is manifested as particulates with a large thickness contrast in the GNRs and GNRs-siRNAFAM treated cells in the bright-field images. According to the merged images, the green fluorescence overlaps the location of the

TE D

GNRs in the GNRs-siRNAFAM treated cells furnishing further evidence of delivery of siRNA by the GNRs into the cells.

As the control, the cells treated with siRNAFAM

alone only show weak green fluorescent signals and as the positive group, the

EP

lipo-siRNAFAM-treated cells show considerable green fluorescent dots.

Flow

AC C

cytometry is employed to quantify the amount of siRNAFAM delivered to the cancer cells (shown in Figure 3b).

Compared to the untreated cells (blank group), about

87.6% of cells exhibit green fluorescence after incubating with the GNRs-siRNAFAM and it is comparable to that of cells treated with Lipo-siRNAFAM of about 84.9%. Since lipofectamine is a classical and recommended gene delivery vector, the results suggest that the GNRs are promising nanocarriers for the delivery of siRNA to cancer cells. 27

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

Figure 3. Cell uptake of GNRs-siRNA by Cal-27 cells.

(a) Bright field, fluorescent,

and merged optical images of the untreated Cal-27 cells (denoting as blank) and cells

TE D

incubated with GNRs, siRNA, GNRs-siRNA, and Lipo-siRNA, respectively.

The

siRNA is labeled with the green fluorescent FAM and nuclei are stained blue with (b) Flow cytometric analysis of green fluorescent FAM.

The results

EP

DAPI.

represent three independent experiments with mean ± SEM and **p

Gold-nanorods-siRNA nanoplex for improved photothermal therapy by gene silencing.

Nanomaterials-mediated photothermal therapy (PTT) often suffers from the fundamental cellular defense mechanism of heat shock response which leads to ...
565B Sizes 0 Downloads 14 Views