Вы находитесь на странице: 1из 12

Integrative Biology

View Article Online


PAPER View Journal | View Issue
Published on 04 November 2013. Downloaded by National Dong Hwa University Library on 27/03/2014 10:23:41.

Folate conjugated silk fibroin nanocarriers for


targeted drug delivery†
Cite this: Integr. Biol., 2014,
6, 203
Bano Subia,a Sourov Chandra,b Sarmistha Talukdara and Subhas C. Kundu*a

Disease treatment processes mainly focus on the development of nontoxic, biodegradable, non-
immunogenic, biocompatible materials capable of controlled and long-term release of biomolecules. In
this work silk protein fibroin from non-mulberry tropical tasar silkworm, Antheraea mylitta, is used to
prepare nanoparticles as a drug delivery system. Folate is a vitamin, which is brought into healthy and
cancerous cells by folate receptors. The efficiency of silk fibroin–folate nanoparticles loaded with
anticancer drug doxorubicin was evaluated by analysing the cell viability, proliferation and endocytosis.
Consequently the effects of pro-inflammatory responses by cytokines such as TNF-a, IL-1b and nitric
Received 9th September 2013, oxide were checked by stimulating the macrophages with folate conjugated silk fibroin nanoparticles.
Accepted 31st October 2013 The fibroin–folate nanocarriers are nontoxic, easily taken up by cells and capable of sustained drug
DOI: 10.1039/c3ib40184g release. Nanoparticles loaded with anticancer drug doxorubicin target cancer cells. The results show
that silk fibroin–folate nanoparticles may serve as promising nanocarriers for different biomedical and
www.rsc.org/ibiology nanotechnology applications in cancer research.

Insight, innovation, integration


Silk fibroin folate conjugated nanoparticulate system is reported for first time. Silk biomaterials have versatile properties such as biocompatibility,
biodegradability, low immunogenicity and non-toxicity. The efficiency of silk fibroin–folate nanoparticles loaded with an anticancer drug are evaluated by
analysing the cell viability, mechanism of cellular uptake, pro-inflammatory responses and pH dependent drug release. This demonstrates the broad
applicability of the silk fibroin–folate nanoparticulate system. The system facilitates interaction with the cells and retention at the site of action for a longer
period of time, which is advantageous for sustained release. The silk fibroin–folate nanoparticulate system leads to new insight for the potentiality of a carrier
for targeted drug delivery.

1. Introduction They should have properties to enhance their circulation time


in blood and actively target the cancerous cells.2 The surface of
In the past few years nanotechnology has been used in the these sub-microscopic particles can be easily modified with
improvement of conventional chemotherapy for cancer treatment. targeting molecules to improve in vivo and in vitro treatment.3
The fundamental advantage of nanoscale particles is that they Over the past decades versatile polymeric nanoparticles have
have a greater surface area to volume ratio. Recent advances been used for drug delivery systems. Polymeric nanoparticles
suggest that nanotechnology is capable of engineering and have excellent biocompatibility, biodegradability, high drug
manipulating materials at the nanoscale level. Nanoscale particles encapsulation efficiency, excellent endocytosis efficiency, passive
reach at target site at a proper concentration to destroy diseased tumor-targeting, controlled drug release and long circulation
cells while minimizing damage to normal cells.1 Nanocarriers half life.4 Among them natural protein silk fibroin are used in
designed to deliver a drug at cancerous cells should be non- the field of nanotechnology to modify the interface of nano-
toxic, non-immunogenic and with high drug loading efficiency. particles and control the growth of particles.5 The silk fibroin
obtained from the Indian tropical tasar silkworm Antheraea
mylitta has been used for the fabrication of nanoparticles. Silk
a
Department of Biotechnology, Indian Institute of Technology, Kharagpur, 721302, fibroin from this tasar silk has the versatile properties of a
India. E-mail: kundu@hijli.iitkgp.ernet.in; Fax: +91-3222-27843
b
natural polymer.6 The fibroin nanoparticles are helpful for the
Department of Chemistry, Indian Institute of Technology, Kharagpur,
721302, India
delivery of enzymes and drugs.7 They have several active amino
† Electronic supplementary information (ESI) available: Video on cellular uptake groups and tyrosine residues that help the conjugation of
of nanoparticles. See DOI: 10.1039/c3ib40184g drug and other macromolecules.8 Coating or conjugating with

This journal is © The Royal Society of Chemistry 2014 Integr. Biol., 2014, 6, 203--214 | 203
View Article Online

Paper Integrative Biology

targeting molecules to the silk fibroin overcomes the major as a targeted drug carrier system. The doxorubicin release profile is
shortcomings of the nanoparticles by increasing the specificity examined as a function of pH with an emphasis on the acid
of the nanoparticles for cell surface attachment.9 This non- cleavability. The tumor cell targeting, endocytosis mechanism of
mulberry fibroin is naturally gifted with RGD sequences which cellular uptake and antitumor activity of the prodrug are examined.
interact with surface integrin molecules and enhance cellular
attachment.10,11 The addition of RGD with this fibroin is
Published on 04 November 2013. Downloaded by National Dong Hwa University Library on 27/03/2014 10:23:41.

required for improving the cell attachment efficiency. 2. Materials and methods
Folic acid/folate is conjugated with fibroin to further 2.1 Materials
increase the specificity and targeted delivery of nanoparticles.
Mature late 5th instar larvae of non-mulberry tropical tasar
The fundamental role of folic acid is essential for nucleotide
silkworm Antheraea mylitta were collected from our experi-
synthesis that promotes rapidly proliferating cells and tissue.3
mental farm, folic acid (Sigma-Aldrich), cellulose tubing of
This includes advanced stages of cancer, and it is also critical
MWCO 12 kDa (Pierce, USA), acetone (Merck), glutaraldehyde,
for the ‘‘1-carbon pathway’’, a key initiating step in the synthesis
doxorubicin, DMSO (dimethyl sulfoxide), MTT (3-(4,5-dimethyl-
of nucleotide precursors as building blocks for DNA synthesis in
thiazol-2-yl)-2,5-diphenyltetrazolium bromide), rhodamine B
the S-phase of the cell cycle.3 Folic acid/folate is often selected as
isothiocyanate (RITC), 2,2 0 -(ethylenedioxy)bis(ethylamine) (EDBE),
a model molecular probe for the targeted delivery of drugs to
N-hydroxysuccinimide (NHS), cytochalasin B, nystatin (Sigma),
cancer cells.12 However, active targeting of membrane bound
sodium azide (Merck) and a live/dead viability/cytotoxicity kit
tumor associated receptors is achieved by the conjugation of
(Invitrogen), were purchased for these experiments.
nanoparticles with folate.12 Recently, the folate mediated target-
ing technique has been established as a non-invasive approach 2.2 Isolation of silk fibroin protein from Antheraea mylitta
for the detection of cancers. The cancerous cells have an inherent
property of overexpressing folate receptors due to their enormous The fully-grown 5th instar larvae were dissected for extraction of the
requirements for folate.13 Tumor selective targeting is achieved silk protein fibroin following the earlier described method.6 The
by combining folic acid conjugates with liposomes/nanoparticles non-bioengineered silk fibroin was isolated from the silkworms
to enhance their uptake and targeting ability.14 Folate receptors just prior to spinning into cocoon fibers. In brief, the posterior silk
have high-affinity membrane folate-binding proteins, which glands were isolated and washed in deionized water to remove
enables them to rapidly bind and be internalized by the process traces of sericin. The glandular tubes were then squeezed with fine
of endocytosis15 or through a hypothetical pathway involving forceps to extrude out the protein. Isolated silk fibroin protein was
endocytosis proposed for targeted delivery.16 Recently it has been dissolved using a mild anionic surfactant SDS following the
proposed that folate receptor mediated delivery can enhance standard method. Excess surfactant was removed by dialysis
cellular uptake, sustained drug release and promote effective against water using dialysis membranes (12 kDa). Fibroin
cytotoxicity towards cancer cells both in vitro and in vivo.17 solution was collected and the concentration was determined
Doxorubicin is used as a model anticancer drug and is a by weighing the remaining solid after drying at 60 1C.
member of the anthracycline ring antibiotics.18 It is crucial for
2.3 Fabrication of silk fibroin nanoparticles
the treatment of acute leukaemia, malignant lymphoma and
breast cancer.19 This drug has a narrow therapeutic index and its Silk fibroin protein nanoparticles were prepared by a desolvation
clinical use is hampered by several side effects such as cardio- technique using acetone as the desolvating agent. To induce the
toxicity and myelosuppression.20 To reduce the side effects of the desolvation process, 20 ml of acetone were added in a small
drug attempts are being made to deliver the drug to its target container and regenerated silk fibroin solution (2%, w/v) was
sites by nanoparticle carriers, which are receiving much atten- added dropwise (50 ml per drop) with constant stirring. After
tion for targeted drug delivery. desolvation, the formation of silk nanoaggregates was visible in
Nanocarriers of silk fibroin–folate–doxorubicin conjugates have the form of precipitates. Nanoaggregates were then centrifuged at
been fabricated as a targeted drug delivery system to minimize the 16 000 rpm for 20 min to collect the precipitates. SF precipitates
risk of drug toxicity and increase drug efficacy. It is hypothesized were then further purified with repeated centrifugation at
that doxorubicin conjugated to nanocarriers through linkers could 16 000 rpm for 15 min with deionized water. The purified nano-
be readily delivered into cells by an endocytosis mechanism. particle pellets were then redispersed in deionized water and soni-
Within the acidic endosomal compartment the doxorubicin may cated. The nanoparticles were filtered through a 0.45 mm syringe
be cleaved in an intact form from the prodrug complex.21 Based on and lyophilized to obtain freeze-dried nanoparticles for further use.
this hypothesis, we have designed and synthesized a prodrug com-
plex, i.e., silk fibroin–folate–doxorubicin by using folic acid as a 2.4 Synthesis of silk fibroin–2,2 0 -(ethylenedioxy)-
targeting agent. Fibroin is a biocompatible, hydrophobic carrier, bis(ethylamine) conjugates (SF–EDBE)
and forms a pH-sensitive linkage with the anticancer drug doxo- 10 mg silk nanoparticles was completely dispersed into 40 ml of
rubicin. The major goal of cancer therapy is to achieve the active water with the assistance of a bath sonicator for 1 h. Then the
targeting of cancer cells and minimize drug side effects. particles were activated with 5 mg EDC–NHS (1 : 1) at pH 6 with
In this work, to achieve effective tumor targeting, silk fibroin– constant stirring for 3 h in the dark. After that 1 ml of EDBE was
folate nanoparticles are synthesized, characterized and investigated added dropwise into the above reaction mixture and the stirring

204 | Integr. Biol., 2014, 6, 203--214 This journal is © The Royal Society of Chemistry 2014
View Article Online

Integrative Biology Paper

was again continued for 10 h. EDBE conjugated SF NPs were mixture was stirred for 3 h in the dark. After that silk–EDBE–
separated out by centrifugation at 4000 rpm for 10 min. Finally RITC was added dropwise to the above solution and the pH was
the product was washed with deionized water several times to adjusted to 8 by the addition of a small amount of pyridine. The
remove excess EDBE followed by lyophilization for further use. reaction mixture was stored in the dark and stirring was
continued for 10 h. Finally the product (silk–EDBE–RITC–FA)
2.5 Synthesis of silk fibroin–2,2 0 -(ethylenedioxy)bis- was isolated by centrifugation followed by washing with deionized
Published on 04 November 2013. Downloaded by National Dong Hwa University Library on 27/03/2014 10:23:41.

(ethylamine)–folate nanoconjugates (SF–EDBE–FA) water several times.


10 mg of folic acid was sonicated in 10 ml of 1 : 1 aqueous
DMSO for 30 min. After that the dispersion was activated by using 2.10 Drug loading and entrapment
5 mg EDC–NHS (1 : 1) at pH 6 with vigorous stirring for 3 h in a 2.5 mg Doxorubicin (DOX) was loaded into 10 ml of an aqueous
closed dark container. After activation, the aqueous dispersion of (deionized water) dispersion of folic acid conjugated silk nano-
folic acid was mixed dropwise into the amine functionalized silk– particles (25 mg), with mixing along with continuous stirring
EDBE nanoparticles. The pH of the mixture was adjusted to 8 by the for 12 h in the dark. The drug (DOX) loaded silk–EDBE–FA nano-
addition of a small amount of pyridine. Stirring was again continued particles were separated out by centrifugation at 10 000 rpm for
for 10 h and finally the product (silk–EDBE–FA) was isolated by 10 min. The particles were then washed thrice with deionized
centrifugation at 4000 rpm for 10 min followed by washing several water to remove excess DOX molecules. Finally the drug loading
times with deionized water and collection after lyophilization. was evaluated spectrophotometrically by measuring the absor-
bance at 482 nm of a standard DOX solution and the super-
2.6 Particle characterization natant respectively.
Folate conjugated silk fibroin nanoparticles were analyzed by
transmission electron microscopy (TEM) for measurements of 2.11 Cumulative drug release
size and surface morphology. TEM samples were prepared by Cumulative drug release from silk fibroin–EDBE–FA was studied
putting 10 ml of diluted nanoparticles after dispersion in at two different pH values. pH 4.5, which is comparable to the
deionized water. The grid containing nanoparticles was air dried lysosomal pH of cancerous cells and pH 7.4, the extracellular
in a dust free environment at room temperature. Then the samples physiological pH of normal cells. Briefly, 3 mg of the DOX loaded
were analyzed under JEOL-JEM 2100, TEM at 80 kV (voltage). The SF–EDBE–FA was dispersed in 2 ml of buffer solution and sealed
average surface charge and polydispersity index were determined inside a dialysis bag (molecular cut-off 50 kDa), which was then
using dynamic light scattering (DLS). For the particle size analysis subsequently immersed in 50 ml of external buffer solution
nanoparticles were resuspended in deionized water and sonicated. (PBS). Dialysis was further continued at 37 1C and drug release
Particle stability and dispersity were calculated from the poly- was monitored spectrophotometrically by measuring the absor-
dispersity index and zeta potential and data were collected from bance of the external buffer at 482 nm. The results of triplicate
several measurements (n = 3). test data were used to calculate cumulated drug release.

2.7 Fourier transform infrared spectroscopy (FTIR) 2.12 Cell culture


The conformation transitions of lyophilized silk fibroin, and Human breast adenocarcinoma cell line (MDA-MB-231) was
folate conjugated silk-fibroin nanopowders were obtained by used for intracellular localization and cytotoxic studies. Breast
the use of Perkin Elmer FTIR spectroscopy in the spectral adenocarcinoma cells were maintained inside an incubator at
region 500–4000 cm1 at the resolution of 4 cm1 in KBr pellet. 37 1C with air supplemented with CO2 (5%). The cells were
All the samples were analyzed in transmission mode. cultured in DMEM with FBS (10%) and antibiotics (1%) and
incubated for 24 h. Cultures were split when they reached 70–80%
2.8 Synthesis of silk fibroin–2,2 0 -(ethylenedioxy)bis- confluence with medium change every 2–3 days in between
(ethylamine)–rhodamine isothiocyanate (SF–EDBE–RITC) culturing.
1 mg of rhodamine isothiocyanate (RITC) was dissolved in 1 : 1 2.12.1 Intracellular localization. To observe the folate conju-
aqueous DMSO and added dropwise into an aqueous dispersion gated silk fibroin nanoparticles, human breast adenocarcinoma
of silk–EDBE (5 mg in 0.1 (M) sodium bicarbonate solution) with cells (MDA-MB-231) were incubated with nanoparticles. Briefly,
continuous stirring. The pH of the solution was kept at 8 and 2  104 cells per well were seeded into 24 well culture plates in
stirring was continued overnight in the dark. Finally SF–EDBE– DMEM culture medium (FBS 10% and antibiotics 1%) for 24 h.
RITC was recovered by centrifugation at 4000 rpm for 10 min and After that the growth medium was removed and replaced with
washed several times with deionized water to remove excess RITC. fresh medium containing RITC loaded SF–FA incubated for 48 h.
RITC loaded pure SF nanoparticles were taken as a control.
2.9 Synthesis of folate conjugated silk fibroin–2,2 0 - Specific uptake of RITC labelled folate conjugated silk fibroin
(ethylenedioxy)bis(ethylamine)–rhodamine isothiocyanate nanoparticles by breast cancer cells (MDA-MB-231) was further
labelled nanoparticles (SF–EDBE–RITC–FA) evaluated in a competitive inhibition assay. RITC labelled SF–FA
After conjugation with RITC, SF–EDBE–RITC and folic acid (FA) conjugated nanoparticles were incubated with 2 mM of free folic
were dissolved in 1 : 1 aqueous DMSO separately. Folic acid was acid and incubated for 12 h. Blocking of the folate receptor with
then activated with the addition of 1 : 1 EDC–NHS and the an excess of free folate was analysed by confocal microscopy.

This journal is © The Royal Society of Chemistry 2014 Integr. Biol., 2014, 6, 203--214 | 205
View Article Online

Paper Integrative Biology

In the absence of free folate, RITC labelled silk fibroin folate loaded nanoparticles. 100 ml of MTT solution (5 mg ml1 in PBS
nanoparticles were taken as control. pH 7.4) was added in each well plate and incubated for 4 h.
2.12.2 Confocal microscopy. Attachment and spreading of Following this, the MTT solution was removed and 500 ml
MDA-MB-231 was observed using confocal laser microscopy. of DMSO was added to dissolve the formazan crystals. The
Cells were fixed with 4% formaldehyde for 15 min. The samples absorbance of samples was measured by a microplate reader
were washed with PBS and treated with 0.1% Triton-X for at 595 nm.
Published on 04 November 2013. Downloaded by National Dong Hwa University Library on 27/03/2014 10:23:41.

membrane permeabilization. After that the whole constructs 2.12.6 Flow cytometry analysis. MDA-MB-231 breast cancer
were washed with PBS and treated with 1% bovine serum cells were seeded at a density of 1  106 cells per well and
albumin (BSA) for 30 min. The cell nuclei were stained with incubated at 37 1C inside an incubator. The cells were pre-
Hoechst 33342 (5 mg ml1) for 30 min. Samples were imaged incubated on TCP for 24 h to allow cell attachment. Culture
using confocal laser scanning microscopy (CLSM, Olympus FV medium was removed and replaced with incomplete medium to
1000 equipped with an inverted microscope IX 81, Japan) equipped starve the cells. Following this cells were incubated with nano-
with argon (488 nm) and HeNe (534 nm) lasers. Two-dimensional particles loaded with pure silk fibroin (500 mg ml1), silk fibroin–
multichannel image processing was done using FV 1000 Advance doxorubicin (10 mg ml1) and pure doxorubicin (10 mg ml1) for
software version 4.1 (Olympus, Japan). 12 h. Wells without nanoparticles were taken as the control. After
that, the cells were harvested by addition of 1 ml of 0.25%
Video image analysis. Movies of SF–FA conjugated nano- trypsin–EDTA to the samples. They were then incubated at 37 1C
particle uptake analysis by MDA-MB-231 were recorded using in an incubator until all cells were detached from the flask. The
confocal laser microscopy at different time intervals. trypsinized cell suspension was neutralized with 3 ml complete
2.12.3 Nanoparticle uptake in the presence of endocytosis medium, followed by centrifugation at 1000 rpm for 10 min. The
inhibitors. Known inhibitors of endocytosis were used to elucidate cell pellet was resuspended in cold PBS and centrifuged for
the mechanism of cellular uptake of silk fibroin–folate conjugated 10 min at 1000 rpm. To the cell pellet 70% chilled ethanol was
nanoparticles. Human breast adenocarcinoma cells were seeded added in shaking conditions and incubated at 4 1C. The cell
in 24 well culture plates at the density of 1  104 cells per well and pellet was resuspended in 200 ml PBS containing 0.1 mg ml1
grown to 70% confluence. The cells were pre-incubated with the RNase and 40 ml of PI solution (1 mg ml1 PI) and incubated for
inhibitors, NaN3 (0.1%), nystatin (15 mg ml1), and cytochalasin D 30 min at 37 1C. The resultant cell suspension was analyzed with
(5 mg ml1) for 1 h. The cells were washed 3 with cold sterile a flow cytometer (FACS Calibur, BD using Cell Quest Pro soft-
PBS. Following this, the cells in each well were resuspended in ware) at 488 nm excitation and a 560 nm band pass filter for red
1 ml DMEM and incubated with folate conjugated nano- fluorescence of propidium iodide.
particles (200 mg ml1) for 6 h. The mechanism of endocytosis 2.12.7 Live/dead assay. A live/dead assay was performed for
was elucidated with confocal laser microscopy. the analysis of cell viability after cellular uptake of nanoparticles.
2.12.4 Cell viability and cytotoxicity. MTT (3-(4,5-dimethyl- Pure SF–FA nanoparticles without drug and nanoparticles conju-
thiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay was used gated with DOX drug were analyzed in the live/dead assay. Samples
to measure the cell cytotoxicity. Human breast adenocarcinoma were stained with calcein and ethidium bromide to visualize the
cells (MDA-MB-231) were seeded at the desired density 2  populations of live and dead cells, after 48 h incubation of nano-
104 cells per well into 24 well plates at 5% CO2 at 37 1C. The particles. Briefly, nanoparticle encapsulated cells were incubated at
cells were pre-incubated on tissue culture plates (TCP) for 24 h 37 1C in PBS containing calcein AM and ethidium homodimer-1
to allow cell attachment. The culture medium was removed and (Molecular Probes) for 20 min to stain viable and non-viable cells,
replaced with incomplete medium. Following this, nanoparticles respectively. The samples were washed thrice with PBS for 10 min
with different concentrations (100, 250 and 500 mg ml1) were each, and imaged by confocal microscopy.
added and incubated for 48 h. After that 100 ml of MTT dye
solution was added (5 mg ml1 MTT in PBS at pH 7.4) in each 2.13 Determination of TNF-a, IL-1b and nitric oxide (NO)
well and incubated for 4 h. The MTT solution was then removed production
and 500 ml of DMSO was added in each well to solubilize the The immunogenicity of the SF–FA nanoparticles was evaluated
formazan crystals. The absorbance of 96 well tissue culture by in vitro quantitative determination of human tumour necrosis
plates was recorded with a microplate reader at 595 nm. factor alpha (TNF-a), IL-1b and NO concentrations in cell culture
2.12.5 Cytotoxicity. Doxorubicin conjugated silk fibroin– supernatant. Two sets of experiments were designed to analyze
folate nanoparticle uptakes were evaluated using cancer cells. in vitro immune response with different SF–FA concentra-
The cancer cells with a density of 2  104 cells per well were tions (100, 250 and 500 mg ml1) for long and short term
seeded in 24 well tissue culture plates. After 24 h, culture culture (1 and 7 days). Briefly macrophage cells with density
medium was replaced with incomplete culture medium to starve 1  105 cells per well were seeded into TCP and incubated for
the cells. Nanoparticles with different drug concentrations 48 h. After 48 h of cell seeding, the complete medium was
(0.1, 1 and 10 mg ml1) were added into fresh medium (10% FBS replaced with incomplete medium. Nanoparticles with concen-
and 1% antibiotics) and incubated for 48 h in a humidified trations 100, 250 and 500 mg ml1 were added into each well to
atmosphere containing 5% CO2 at 37 1C. After that, an MTT stimulate the macrophages. The cell supernatant was collected
assay was carried out to evaluate the cytotoxicity of the DOX to analyze TNF-a and IL-1b levels using TNF-a and IL-1b

206 | Integr. Biol., 2014, 6, 203--214 This journal is © The Royal Society of Chemistry 2014
View Article Online

Integrative Biology Paper

quantification kits (Invitrogen). Data accumulations were carried II and III bands (1630, 1516, 1231 cm1). No characteristic
out at 450 nm and 600 nm using a microplate reader. To quantify peak for carboxylic acid is observed in such silk fibroin as
the NO production cell supernatants were collected and incubated these are fewer in comparison to the amide groups. Only the
with Griess reagents (Sigma) for 10 min. Data accumulations were terminal part of the silk fibroin contains a –COOH moiety,
done at 548 nm and 600 nm using a microplate reader. which is used for the conjugation of folic acid. Fig. 2b
reveals the FTIR spectrum of silk fibroin after treatment with
Published on 04 November 2013. Downloaded by National Dong Hwa University Library on 27/03/2014 10:23:41.

2.14 Statistical analysis 2,2 0 -(ethylenedioxy)bis(ethylamine), in which the peaks for


Statistical differences were performed using one-way ANOVA. amide I and II are shifted to higher wavelengths (1652 and
Data are presented as mean  SD. p-Values o0.05 were con- 1524 cm1 respectively). An additional broad peak at 3417 cm1
sidered as statistically significant. clearly signifies the presence of 11 amine (–NH2) groups
on the surfaces of those particles. Finally Fig. 2c illustrates
the conjugation of folic acid with silk fibroin–EDBE nano-
3. Results particles revealing the formation of amide bonds in the
3.1 Surface properties of silk fibroin–folate conjugated presence of EDC–NHS acting as the coupling agent. The amide
(SF–FA) nanoparticles I, II and III bands in silk fibroin–EDBE–FA are recorded at 1637,
Protein based nanoparticles were fabricated by a desolvation 1524, and 1239 cm1 respectively. The absence of free amine in
method followed by conjugation with folate by successive steps. silk fibroin–EDBE–FA suggests that all –NH2 groups are con-
The size and zeta potential of SF–FA conjugated nanoparticles verted to amide bonds after conjugation with the –COOH
were determined using transmission electron microscopy and a groups of FA. A large and broad peak at 3437 cm1 confirms
dynamic light scattering particle size analyzer respectively. TEM the presence of –OH groups in SF–EDBE–FA coming from FA
images show that the particles are monodisperse and o200 nm (Fig. 2a–c).
in diameter. The particles are almost uniform and spherical in
3.3 Cellular uptake and localization of silk fibroin–folate
shape (Fig. 1). The stability of the particles is confirmed from
nanoparticles on cancer cells
their zeta potentials. The surface potentials of SF and SF–FA
conjugated nanoparticles are 22  5 and 32  3 respectively. In this study, it was hypothesized that cellular uptake of folic
acid is enhanced by conjugation of folic acid with SF nano-
3.2 FTIR analysis of nanoparticles particles. The cellular binding and uptake of rhodamine iso-
The FTIR spectrum of silk fibroin (Fig. 2a) illustrates the thiocyanate (RITC) labelled folate silk fibroin nanoparticles
presence of an amide A band (3296 cm1) along with amide I, were analyzed on human breast adenocarcinoma cells by con-
focal laser microscopy. The carboxylic group of folate is
covalently conjugated with the amino group of silk fibroin by
using the 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide
(EDC) coupling method. Uptake efficiencies of RITC labelled
pure silk fibroin nanoparticles and covalently folate conjugated
silk fibroin nanoparticles were analyzed at different incubation
periods (0 and 24 h). RITC labelled pure silk fibroin nano-
particles were chosen as a control (Fig. 3A). However, cellular
uptake of RITC labelled silk fibroin nanoparticles by cancer
cells was analysed after 12 h. It showed that in the presence of
competitive inhibitor (free folate, 2 mM), nanoparticle binding
to the folate receptor is less (Fig. 3B). Whereas the same
Fig. 1 TEM pictographs of (a) silk protein fibroin nanoparticles and (b) silk nanoparticles effectively bind with cancer cells in the absence
fibroin–folate nanoparticles. of competitive inhibitor (free folate).

Fig. 2 FTIR spectra of (a) pure silk fibroin; (b) silk fibroin–2,2 0 -(ethylenedioxy)bis(ethylamine) nanoconjugates (SF–EDBE); and (c) silk fibroin–2,2 0 -
(ethylenedioxy)bis(ethylamine)–folate nanoconjugates (SF–EDBE–FA).

This journal is © The Royal Society of Chemistry 2014 Integr. Biol., 2014, 6, 203--214 | 207
View Article Online

Paper Integrative Biology


Published on 04 November 2013. Downloaded by National Dong Hwa University Library on 27/03/2014 10:23:41.

Fig. 3 (A) Confocal images of human breast adenocarcinoma cells incubated with 1 mg ml1 of RITC labelled nanoparticles (a–d) 0 h; (e–h) RITC
labelled pure silk fibroin nanoparticles; and (i–l) RITC labelled silk fibroin–folate nanoparticles. Represents cell attachment, proliferation and viability at
37 1C. Scale bars represent 50 mm. (B) Confocal laser microscopy images after 12 h incubation of RITC labelled silk fibroin folate nanoparticles with
MDA-MB-231 cells, in (a) excess free folate (2 mM); (b) the absence of free folate. Scale bars represent 50 mm diameter. (C) Blocking the cellular uptake of
silk fibroin–folate nanoparticles by using endocytosis inhibitors. The cells are pre-treated with endocytosis inhibitors for 1 h, followed by addition of silk
fibroin–folate nanoparticles for 6 h (a) control; (b) RITC labelled SF–FA nanoparticles; (c) NaN3; (d) cytochalasin B; (e) nystatin; (f) mean fluorescence
intensity of nanoparticles after treatment with endocytosis inhibitors. Scale bars represent 50 mm diameter.

208 | Integr. Biol., 2014, 6, 203--214 This journal is © The Royal Society of Chemistry 2014
View Article Online

Integrative Biology Paper

3.4 Nanoparticle uptake mechanism 3.6 Cytotoxicity assay


A variety of endocytosis mechanisms operated in cells includ- In vitro cytotoxicity assays of SF–FA and DOX loaded SF–FA NPs were
ing calthrin-mediated, caveolin-mediated, calthrin or caveolin carried out on human breast adenocarcinoma cells. The cytotoxi-
independent endocytosis and macropinocytosis. The cells are cities of folate conjugated silk fibroin nanoparticles with different
pre-incubated with various endocytosis inhibitors and incu- concentrations (100, 250 and 500 mg ml1) were evaluated by MTT
Published on 04 November 2013. Downloaded by National Dong Hwa University Library on 27/03/2014 10:23:41.

bated with silk fibroin–folate nanoparticles. Nanoparticle entry assay. The results demonstrate that the cell viability and prolifera-
into cells under these conditions is evaluated by measuring the tion are not significantly affected by different nanoparticle concen-
fluorescence intensity. To investigate the processes, MDA-MB- trations (Fig. 5b). In contrast, cell inhibitory effects were evaluated
231 cells are pre-incubated in the presence of sodium azide with free drug (DOX) and drug loaded nanoparticles (DOX–SF–FA)
(energy dependent endocytosis), nystatin (calthrin or caveolin with different drug concentration (0.1, 1.0 and 10 mg ml1) on
mediated endocytosis), cytochalasin D (macropinocytosis) and human breast cancer cells. Cell viabilities were determined by MTT
then treated with SF–FA nanoparticles. An inhibited inter- assay. The MTT assay data more likely suggest a change in the cell
nalization is observed after treatment with these inhibitors. metabolic activity or enzyme activity in the mitochondrial respira-
The result shows that the internalization of nanoparticles into tory chain in treated cells. Free DOX and DOX–SF–FA nanoparticles
breast cancer cells pre-treated with these inhibitors is compara- show dose dependent cytotoxicity (Fig. 5c).
tively less than non-treated cells (Fig. 3C).
3.7 Cell cycle analysis
3.5 Live/dead assay As shown in Fig. 6, SF–FA–DOX creates cytotoxicity in breast cancer
To study the biocompatibility of silk fibroin–folate conjugated cells, whereas the effect of SF–FA nanoparticles is almost similar
nanoparticles and doxorubicin (DOX) loaded SF–FA nano- to that of the control. Doxorubicin creates immediate effects on
particles, live/dead assays were performed on MDA-MB-231 cells. breast cancer cells (10 mg ml1), while SF–FA conjugated DOX
SF–FA nanoparticles with concentration (100, 250 and 500 mg ml1) creates slow and effective cytotoxicity. Thus, the maximum of the
and DOX loaded SF–FA nanoparticles with concentration cells seems to be influenced by the cytotoxic effect of doxorubicin
(0.1, 1.0 and 10 mg ml1) were used for live/dead assays released from the folate conjugated silk fibroin nanoparticles.
respectively. From Fig. 4a–d it is clearly observed that a large FACS showed that the SubG0 of the apoptotic cells was 0.76%,
percentage of live cells are observed on SF–FA NPs with 4.28%, 3.50%, 25.85% for the control, SF–FA, SF–FA–DOX nano-
concentration 100, 250 and 500 mg ml1. This indicates that particles and pure DOX respectively. On the other hand the
nanoparticles are nearly non-toxic to the cells (Fig. 4a–d). proportion of cells in the G1 phase was 79.47%, 70.23%, 13.21%,
Similarly, SF–FA–DOX conjugate nanoparticles are able to 43.21% and in the G2 phase of the cell cycle was 9.03%, 13.55%,
effectively target the cancer cells (Fig. 4e–h). In this study, the 65.27%, and 8.72% respectively in all four groups.
loading ratio of DOX is evaluated as 1.0 mg per mg of SF–FA
nanoparticles. The results suggest that with increasing drug 3.8 Cytokine production assay
concentration, cell viability decreases in comparison to non- To analyze the immunogenic response of silk fibroin folate
treated cells (Fig. 4e–h). nanoparticles mouse bone marrow macrophages were used.

Fig. 4 Live/dead assay on human breast cancer cells incubated with silk fibroin–folate nanoparticles with different concentration: (a) control; (b) 100 mg ml1;
(c) 250 mg ml1; (d) 500 mg ml1 and doxorubicin loaded silk fibroin folate nanoparticles with different drug concentration: (e) control; (f) 0.1; (g) 1.0;
(h) 10 mg ml1 after 48 h of incubation, represents cell attachment, proliferation and viability at 37 1C. Scale bars represent 100 mm.

This journal is © The Royal Society of Chemistry 2014 Integr. Biol., 2014, 6, 203--214 | 209
View Article Online

Paper Integrative Biology


Published on 04 November 2013. Downloaded by National Dong Hwa University Library on 27/03/2014 10:23:41.

Fig. 5 (a) Schematic representation of the mechanism of action of doxorubicin (DOX) conjugated nanoparticles on cancer cells (modified from
Mansoori et al., 2010, ref. 14); (b) viability of human breast adenocarcinoma cells after 48 h exposure to silk fibroin–folate nanoparticles with different
concentration (100, 250 and 500 mg1 ml); and (c) cytotoxicity effect of doxorubicin loaded pure silk fibroin and silk fibroin–folate conjugated
nanoparticles with different drug concentration (0.1, 1, 10 mg ml1) on human breast cancer cells after 48 h exposure as determined by MTT assay.
The data represent mean  SD (n = 3), * represents statistically significant differences (p o 0.05).

Fig. 6 Flow cytometry analysis of cell phase distribution: (a) control; (b) SF–FA nanoparticles (500 mg ml1); (c) pure DOX (10 mg ml1); (d) SF–FA–DOX
NPs (10 mg ml1); (e) overlay of SF–FA NPs and DOX; (f) overlay of SF–FA–DOX NPs and pure DOX incubated with cancer cells for 12 h, determined by
flow cytometry using FACS Diva software after the cells were labelled by PI preceding RNase treatment and the percentage of apoptosis was calculated.

These macrophages constitute a homogeneous population of done with SF–FA nanoparticles with concentration (100, 250 and
primary cells that respond to proliferate or activating stimuli 500 mg ml1) for short and long day culture periods (1 and 7 days).
under some circumstances by becoming apoptotic. The pro- For nitric oxide production, a macrophage stimulation assay
inflammatory response of the cells with nanoparticles was was observed by using Griess reagents. It clearly indicated that
studied by the production of cytokines such as TNF-a, IL-1b and RAW 264.7 cell stimulation with SF–FA shows minimal immune
NO by using ELISA kit. Briefly, an immune response study has response, while lipid polysaccharide (LPS) reflects high cytokine

210 | Integr. Biol., 2014, 6, 203--214 This journal is © The Royal Society of Chemistry 2014
View Article Online

Integrative Biology Paper


Published on 04 November 2013. Downloaded by National Dong Hwa University Library on 27/03/2014 10:23:41.

Fig. 7 (a) TNF-a production by RAW 264.7 murine macrophages; (b) IL-1b production; and (c) nitric oxide (NO) production. RAW 264.7 cells were
stimulated with silk fibroin–folate (SF–FA) nanoparticles with different concentrations (100, 250, 500 mg ml1). Lipopolysaccharide from Escherichia coli
was added (100 ng ml1) to each experimental set as a positive stimulant and media as a negative control. Error bars represent the standard error of the
mean (n = 3).

production (Fig. 7a–c). LPS was chosen as a positive control for pH values. The patterns reveal that the release profile of
an inflammatory response due to its known ability to activate doxorubicin varies with pH. The overall release of the drug is
antigen presenting cells. much higher at pH 4.5 than at pH 7.4.

3.9 In vitro drug release


The administration of chemotherapeutic drugs needs a proper 4. Discussion
delivery system to achieve effective killing of cancer cells. The Our aim is the conjugation of folate with silk protein fibroin
encapsulation of anticancer drugs with nanoparticles can over- (SF) to accomplish targeted drug delivery. However, in this work
come the issues and help in localization within a carcinoma and for effective drug targeting SF is conjugated with amine functional
stimulating the uptake.22 The use of polymeric nanoparticles groups. The folate is conjugated with amine functionalized
as drug delivery vehicles is gaining importance in biomedical nanoparticles by using carbodiimide chemistry by free radical
applications. They enable the encapsulation and successful polymerization.23 Recent studies have shown that the smallest
delivery of drugs with poor aqueous solubility profiles such as silk fibroin–folate conjugated (SF–FA) nanoparticles (B200 nm)
antitumor agents. In vitro release studies were conducted at with higher zeta potential take advantage of the enhanced
pH 7.4 and pH 4.5 by using PBS as a release medium to simulate permeability retention effect and circulate in the intercellular
the physiological pH and endosomal pH of tumour cells res- spaces of blood capillaries without any aggregation.24 Spherical
pectively. The release medium shows a typical release profile shaped nanoparticles experience faster uptake than rod shaped
followed by a sustained release pattern. Fig. 8 represents the nanoparticles.25
in vitro cumulative release profile of doxorubicin at different However, the cellular uptake and fluorescence of folate
decorated silk fibroin nanoparticles is higher in comparison
to pure silk fibroin (Fig. 3A and Movie S1 and S2, ESI†).
Fluorescent labelled SF–FA nanoparticles get internalized by
the cells through folate receptors.26 The particles reside in the
cytoplasm and perinuclear space of the cells and their fluores-
cent intensity and internalization increases with time. It has
been proposed that folate is often a limiting nutrient in human
serum; up-regulation of high affinity FRs on cancer cells may
enable malignant cells to compete more aggressively for the
vitamin.27 Hence, folate not only increases the retention of the
nanoparticles at the tumour site but also promotes cellular
uptake in drug delivery systems.28 The involvement of folate
cell surface receptors in the binding and cellular uptake of silk
fibroin nanoparticles with cancer cells is validated by the
blocking of folate receptors with excess folic acid. It seems to
Fig. 8 Release profile of doxorubicin from doxorubicin–silk fibroin–folate
be interesting that uptake of the silk fibroin folate nano-
nanoparticles in PBS solution at pH 7.4 and 4.5 at 37 1C, observed for a particles is much less in the presence of competitive inhibitor
period of 21 days. (free folate). Whereas in the absence of free folate, the binding

This journal is © The Royal Society of Chemistry 2014 Integr. Biol., 2014, 6, 203--214 | 211
View Article Online

Paper Integrative Biology

affinity of RITC labelled folate conjugated silk fibroin nano- signify that SF–FA conjugated with doxorubicin more effectively
particles to the cancer cell is high. On the basis of that we release the drug as compared to pure DOX. Doxorubicin release
conclude that excess free folate (competitive inhibitor) blocks from the SF–FA matrices is most likely due to the presence of
the folate receptors. It inhibits the binding and cellular uptake enzymes and pH variations. It accelerates degradation of
affinity of folate conjugated silk fibrin nanoparticles to the matrices and allows the diffusion of DOX from the matrix
receptor sites of cancerous cells. and exhibits cytotoxicity.34,35 DOX conjugated nanoparticles
Published on 04 November 2013. Downloaded by National Dong Hwa University Library on 27/03/2014 10:23:41.

However, endocytosis inhibitors are used for the further may enhance cell internalization and lead to the delivery of
assessment of the cellular uptake mechanism of nanoparticles. drug closer to the intracellular site of action.36 These findings
Endocytosis inhibitor, sodium azide, blocks cellular ATP suggest that SF–FA conjugated with doxorubicin might effec-
synthesis.29 In this result a remarkable internalization inhibi- tively kill cancer cells. Although further studies are needed to
tion effect is observed. However, the inhibitory internalization of understand the exact mechanism of toxicity of the drug loaded
nanoparticles into breast cancer cells pre-treated with nystatin nanoparticles against the cancer cells. However, more detailed
and cytochalasin D remains approximately the same.30 On the investigations are needed to further explore the mechanism of
basis of the above experimental results, we conclude that the DOX conjugated silk fibroin and DOX conjugated silk fibroin
admission of silk fibroin–folate nanoparticles into breast cancer folate nanoparticles through cellular uptake, intracellular
cells is predominated by endocytosis (Fig. 3B). It seems that trafficking, and the cytotoxic mechanism of DOX delivered by
none of the specific inhibitors leads to greater than 90% inhibi- this system.
tion of internalization. Interactions of blood occur when drug loaded nanoparticles
Biocompatibility and non-toxicity of SF–FA conjugated nano- are inoculated intravenously and come in contact with macro-
particles are the other considerations for targeted drug delivery. phages. By internalization of exogenous substrate macrophages
Live/dead and cell viability assays evaluate that nanoparticles could secretes cytokines, chemical messengers for regulating
are nearly non-toxic and compatible with cells ( p o 0.05) innate and adaptive immune systems.37 Their concentrations
(Fig. 4a–d and 5b). However, DOX loaded SF–FA nanoparticles are low in cells in the absence of exogenous stimuli but become
create toxicity to human breast cancer cells (Fig. 4e–h). It seems synthesized de novo when the cell is activated.38 Stimulation of
to be interesting that folate conjugated silk fibroin nano- macrophages with SF–FA nanoparticles failed to respond with
particles are nearly less toxic to fibroblast cells. While DOX consistently elevated levels of TNF-a, IL-b and NO production
conjugated silk fibroin folate nanoparticles (DOX–SF–FA) create (Fig. 7a–c). Macrophages cultured in the presence of SF–FA
cytotoxicity to cancer cells, with a neutral effect on fibroblast nanoparticles did not up-regulate transcript levels for a wide
cells (Fig. S4b, ESI†). This might be due to the presence of folate range of pro-inflammatory cytokines to any significant degree.39
receptors on cancer cells that promote/increase the cellular The maximum levels of TNF-a and IL-b release from macro-
uptake of doxorubicin loaded nanoparticles. phages are less than 500 pg ml1 and for NO it is o15 mM. It has
The in vitro cell inhibitory effects of DOX loaded SF–FA been already reported that this level of cytokines would not cause
conjugated nanoparticles show superior cytotoxic activity as any inflammatory response and not prevent proliferation of the
compared to free DOX ( p o 0.05). The IC50 value (50% cells.40,41 The lower cytokine secretion from SF–FA particles
inhibitory concentration) for DOX loaded nanoparticles is lower confirms the absence of any significant inflammatory activity.
(IC50 0.611  1.05 mg ml1) in comparison to free DOX ( p o 0.05). It could therefore be concluded that nanoparticles would not
SF–FA nanoparticles enhance 3-fold (approximately) cytotoxicity of elicit significant macrophage response and it may confirm the
DOX in comparison to free DOX. The efficacy of DOX-loaded less immunogenic behaviour of the nanoparticles.
SF–FA nanoparticles is due to ready internalization of nano- Doxorubicin conjugated silk fibroin folate nanoparticles
particles by endocytosis compared to a passive diffusion mecha- could be used as a pH-stimulated drug delivery system.42,43
nism of free DOX.31 However, nanoparticles enhance the uptake It improves the drug’s half-life in plasma and increases the
and retention of DOX at the effective site. Fig. 5a shows a drug’s accumulation at a tumour site.44 Though, a lesser
hypothetical representation of the internalization and diffusion amount of drug is released slowly at pH 7.4 due to poor
of DOX loaded SF–FA nanoparticles in cancer cells. The cell solubility of DOX at basic pH, where dissociation of the drug
viability of human breast adenocarcinoma cells is decreased; from nanoparticles is less.45 It is assumed that the DOX release
this may be due to apoptosis induction and inhibition of the rate is maximum and quicker at pH 4.5 because of weak
cell cycle. It has already been reported that DOX prevents binding between DOX and the carboxylic group of polymer
cancer cell growth by inducing DNA breakage as well as cell and the reprotonation of the amino groups of DOX (Fig. 8).
cycle arrest.32 It arrests the cell cycle at the G2/M phase.33 It is When the environmental pH is lower, polypeptide segments
observed that both SubG0/G1 and G2/M phases increase in become hydrophilic because under acidic conditions, H+ in
DOX loaded NP treated cells with respect to a control at 12 h solution would compete with the hydrogen bond forming group
incubation of cancer cells as shown in Fig. 6. On the other hand and weaken hydrogen bond interactions, which make the polymer
G1 and G2 phases decrease in a dose dependent way without more soluble in water.46 Both drug and polymer carry positive
significant alteration in the S phase. Pure DOX showed cell charge at lower pH, which provides the necessary repulsion
cycle arrest in the G1 phase, whereas DOX loaded SF–FA between them.47,48 These factors control the higher and prolonged
nanoparticles arrest the cell cycle in the G2 phase. The results drug release in the acidic environment of a tumour site and the

212 | Integr. Biol., 2014, 6, 203--214 This journal is © The Royal Society of Chemistry 2014
View Article Online

Integrative Biology Paper

lower drug release in other tissues. Moreover, there is more 12 D. Zhao, X. Zhao, Y. Zu, J. Li, Y. Zhang and R. Jiang,
accelerated release inside the endosome/lysosome of tumour Int. J. Nanomedicine, 2010, 5, 669–677.
cells. The results showed that nanoparticles could not only 13 S. J. Yang, F. H. Lin, K. C. Tsai, M. F. Wei, H. M. Tsai,
solubilize the poorly soluble drug but also control sustained J. M. Wong and M. Shieh, Bioconjugate Chem., 2010, 21,
drug release. Therefore, a pH-dependent release function is 679–689.
especially useful for achieving tumour-targeted drug delivery. 14 G. A. Mansoori, K. S. Brandenburg and A. Shakiri-Zadeh,
Published on 04 November 2013. Downloaded by National Dong Hwa University Library on 27/03/2014 10:23:41.

Cancers, 2010, 2, 1911–1928.


15 B. Stella, S. Arpicco, M. T. Peracchia, D. Desmaële,
5. Conclusions J. Hoebeke, M. Renoir, J. D’Angelo, L. Cattel and
Silk fibroin–folate conjugated nanoparticles were fabricated for P. Couvreur, J. Pharm. Sci., 2000, 89, 1452–1464.
targeted delivery to tumour cells. The fabricated nanoparticles 16 S. Sabharanjak and S. Mayor, Adv. Drug Delivery Rev., 2004,
loaded with doxorubicin were taken up by breast cancer cells 56, 1099–1109.
via endocytosis. The particles are less than B200 nm in 17 Y. Zheng, S. G. Song and M. Darby, J. Biotechnol., 2010, 145,
diameter, stable, non-toxic and less immunogenic to the cells. 47–53.
The pH dependent release pattern of doxorubicin suggests that 18 S. Bisht and A. Maitra, Wiley Interdiscip. Rev.: Nanomed.
the prepared nanoparticles can serve as effective drug delivery Nanobiotechnol., 2009, 1, 415–425.
agents. The experimental observations based on the fabricated 19 A. Rahman, A. Joher and J. R. Nafees, Br. J. Cancer, 1986, 54,
natural protein fibroin–folate conjugated nanoparticles promise 401–408.
a potential, effective and targeted drug delivery system. 20 A. Kakinoki, Y. Kaneo, Y. Ikeda, T. Tanaka and K. Fujita,
Biol. Pharm. Bull., 2008, 31, 103–110.
21 C. Du, D. Deng, L. Shan, S. Wan, J. Cao, J. Tian, S. Achilefu
Acknowledgements and Y. Gu, Biomaterials, 2013, 34, 3087–3097.
22 E. J. Yeo, Y. S. Chun, Y. S. Cho, J. Kim, J. C. Lee, M. S. Kim
We are thankful to Dr Tuli Dey for FACS analysis in this work. and J. W. Park, J. Natl. Cancer Inst., 2003, 95, 516–525.
The work is financially supported by the Department of Bio- 23 A. S. Wadajkar, J. U. Menon, Y. S. Tasi, C. Gore, T. Dobin,
technology, through its Bioinformatics SUB-DIC and NER twin- L. Gandeeh, K. Kangasniemi, M. Takahashi, B. Manandhar,
ning programmes, Govt. of India, New Delhi. The discussions J. M. Ahn, J. T. Hsieh and K. T. Nguyen, Biomaterials, 2013,
and suggestions in this work by the expertise from the Indian 34, 3618–3625.
Council of Medical Research are thankfully acknowledged. 24 A. J. Ditto, K. N. Shah, N. K. Robishaw, M. J. Panzer,
W. J. Youngs and Y. H. Yun, Mol. Pharmaceutics, 2012, 9,
References 3089–3098.
25 M. L. Hans and A. M. Lowman, Curr. Opin. Solid State Mater.
1 Y. Cao, Y. Gu, H. Ma, J. Bai, L. Liu, P. Zhao and H. He, Sci., 2002, 6, 319–327.
Int. J. Biol. Macromol., 2010, 46, 245–249. 26 K. S. Ho and M. S. Shoichet, Curr. Opin. Chem. Eng., 2013, 2, 1–7.
2 X. H. Peng, X. Qian, H. Mao, A. Y. Wang, Z. G. Chen, S. Nie 27 W. L. Suen and Y. J. Chau, J. Controlled Release, 2013, 167,
and D. M. Shin, Int. J. Nanomedicine, 2008, 3, 311–321. 21–28.
3 A. E. Garcia-Bennett, C. Xiao, C. Zhou, T. Castle, K. Miyasaka 28 Y. Zhang, H. Hong, D. V. Myklejord and W. Cai, Small, 2011,
and O. Terasaki, Chem.–Eur. J., 2011, 17, 13510–13516. 7, 3261–3269.
4 C. He, Y. Hu, L. Yin, C. Tang and C. Yin, Biomaterials, 2010, 29 J. Panyam and V. Labhasetwar, Pharm. Res., 2003, 20, 2.
31, 3657–3666. 30 W. Xia and P. S. Low, J. Med. Chem., 2010, 53, 6811–6824.
5 B. Subia, S. Mukharjee, R. P. Bahadur, V. M. Correlo, 31 H. Yang, C. Lou, M. Xu, C. Wu, H. Miyoshi and Y. Liu, Int.
R. L. Rui, R. Sabrinathan, K. Sekar and S. C. Kundu, Open J. Nanomedicine, 2011, 6, 2023–2032.
Tissue Eng. Regener. Med. J., 2012, 5, 9–16. 32 G. S. Wu, J. J. Lu, J. J. Guo, Y. B. Li, W. Tan, Y. Y. Dang,
6 B. B. Mandal and S. C. Kundu, Macromol. Biosci., 2008, 8, Z. F. Zhong, Z. T. Xu, P. X. Chen and T. Y. Wang, Fitoterapia,
807–818. 2012, 83, 408–414.
7 B. Subia and S. C. Kundu, Nanotechnology, 2013, 24, 33 H. Lee, S. Park, J. B. Kimb, J. Kim and H. Kim, Cancer Lett.,
035103. 2013, 332, 110–119.
8 Y. Q. Zhang, W. D. Shen, R. L. Xiang, L. J. Zhuge, W. J. Gao 34 H. S. Yoo, K. H. Lee, J. E. Oh and T. G. Park, J. Controlled
and W. B. Wang, J. Nanopart. Res., 2007, 9, 885–900. Release, 2000, 68, 419–431.
9 A. S. Gobin, C. E. Butler and A. B. Mathur, Tissue Eng., 2006, 35 A. B. E. Attia, C. Yang, J. P. K. Tan, S. Gao, D. F. Williams,
12, 3383–3394. J. L. Hedrick and Y. Y. Yang, Biomaterials, 2013, 34,
10 A. Datta, A. K. Ghosh and S. C. Kundu, Comp. Biochem. 3132–3140.
Physiol., Part B: Biochem. Mol. Biol., 2001, 129, 197–204. 36 A. Cambon, A. Ray-Rico, S. Barbosa, J. F. A. Soltero, S. J.
11 C. Patra, S. Talukdar, T. Novoyatleva, S. R. Velagala, C. Mühlfeld, Yeates, J. Brea, M. I. Loza, C. Alvarez-Lorenzo, A. Concheiro,
B. Kundu, S. C. Kundu and F. B. Engel, Biomaterials, 2012, 33, P. Taboada and V. Mosquera, J. Controlled Release, 2013,
2673–2680. 167, 68–75.

This journal is © The Royal Society of Chemistry 2014 Integr. Biol., 2014, 6, 203--214 | 213
View Article Online

Paper Integrative Biology

37 J. H. Doroshow, Proc. Natl. Acad. Sci. U. S. A., 1986, 83, 4514–4518. 43 S. Parveen, R. Misra and S. K. Sahoo, Nanomed.: Nanotechnol.,
38 R. Shukla, V. Bansal, M. Chaudhary, A. Basu, R. R. Bhonde Biol. Med., 2012, 8, 147–166.
and M. Sastry, Langmuir, 2005, 21, 10644–10654. 44 S. Yu, G. Wu, X. Gu, J. Wang, Y. Wang and H. J. Gao, Colloids
39 V. Thomas, B. A. Halloran, N. Ambalavana, S. A. Catledge Surf., B, 2013, 103, 15–22.
and Y. K. Vohra, Acta Biomater., 2012, 8, 1939–1947. 45 C. Nathan, Cell, 1995, 82, 873–876.
40 P. Aramwit, S. Kanokpanont, W. De-Eknamkul and 46 Y. Lu and P. S. Low, Adv. Drug Delivery Rev., 2002, 54,
Published on 04 November 2013. Downloaded by National Dong Hwa University Library on 27/03/2014 10:23:41.

T. Srichana, J. Biosci. Bioeng., 2009, 107, 556–561. 675–693.


41 F. Khan, S. Roychowdhury, R. Nemes, P. Vyas, P. Woster and 47 W. She, N. Li, K. Luo, C. Guo, G. Wang, Y. Geng and Z. Gu,
C. Svensson, Toxicology, 2006, 218, 90–99. Biomaterials, 2013, 34, 2252–2264.
42 R. Wilusz, J. Weinberg, F. Guilak and A. J. McNulty, 48 X. Wang, H. J. Kim, P. Xu, A. Matsumoto and D. L. Kaplan,
J. Orthop. Res., 2008, 26, 504–512. Langmuir, 2005, 21, 11335–11341.

214 | Integr. Biol., 2014, 6, 203--214 This journal is © The Royal Society of Chemistry 2014

Вам также может понравиться