ArticlePDF Available

Transdermal Gene Delivery by Functional Peptide Conjugated Cationic Gold Nanoparticle Reverses the Progression and Metastasis of Cutaneous Melanoma

Authors:

Figures

Content may be subject to copyright.
Transdermal Gene Delivery by Functional Peptide-Conjugated
Cationic Gold Nanoparticle Reverses the Progression and Metastasis
of Cutaneous Melanoma
Jie Niu,
Yang Chu,
Yan-Fen Huang,
Yee-Song Chong,
Zhi-Hong Jiang,
Zheng-Wei Mao,
§
Li-Hua Peng,*
,,
and Jian-Qing Gao*
,
Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P. R. China
State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, P. R. China
§
MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering,
Zhejiang University, Hangzhou 310027, P. R. China
ABSTRACT: Permeability barrier imposed by stratum corneum makes an
extreme challenge for the topical delivery of plasmid DNA (pDNA), which is
widely used in gene therapy. Existing techniques to overcome the skin
barrier for bio-macromolecules delivery rely on sophisticated mechanical
devices. It is still a big challenge to treat the skin cancer, for example,
melanoma, that initiates in the dermal layer by topical gene therapy. To
facilitate the skin penetration of pDNA deeply into the melanoma tissues, we
here present a cell-penetrating peptide and cationic poly(ethyleneimine)
conjugated gold nanoparticle (AuPT) that can compact the pDNAs into
cationic nanocomplexes and penetrate through the intact stratum corneum
without any additional enhancement used. Moreover, the AuPT is highly
ecient in stimulating the intracellular uptake and nuclear targeting of the
pDNAs in cells, which guarantees the eective transfection. This study
provides evidence that penetrating peptide conjugated cationic gold
nanoparticle oers a promising vehicle for both the skin penetration and transfection of pDNAs, possessing great potential in
topical gene therapy.
KEYWORDS: stratum corneum penetration, plasmid DNA, cationic gold nanoparticle, cell-penetrating peptide, melanoma
1. INTRODUCTION
Existing techniques to overcome the skin barrier for topical
delivery of bio-macromolecules, like plasmid DNA (pDNA)
and protein, rely on sophisticated mechanical devices,
1
such as
the ultrasonic apparatus,
2
iontophoresis,
3
microneedles, and
electroporation.
4
Recently, the utilization of nanoparticles in
biomedicine holds great potential for topical drug delivery.
5,6
However, most reported topical strategies with nanoparticles
still require the combination of photoinducement, thermal
ablation, or magnetism to enhance the skin penetration
eciency
7,8
so as to induce signicant therapeutic eects.
9,10
Bio-macromolecules are usually hydrophilic with large size and
will be mostly blocked by skin. Zheng et al. have recently
reported the topical delivery of siRNA as eective treatment for
skin melisma,
11,12
which reminded us of the potential of topical
gene therapy for cutaneous melanoma. As we know, pDNAs are
frequently utilized in gene therapy because of their higher
stability than that of SiRNA, MicroRNA, and DNA.
13,14
However, until now, topical delivery of pDNAs for the
cutaneous melanoma treatment has never been reported
elsewhere.
Contributing to their small size, general nontoxicity, ease of
functionalization, and high surface-to-volume ratio, gold
nanoparticles caused increasing attention from nonviral gene
delivery and therapy. Recently, Conde et al. investigated gold
nanoparticles (AuPT)-based triple-combination therapy, con-
sisting of therapeutic gene, drugs, and photo-based treatments.
The triple-combination therapy was demonstrated to eciently
inhibit tumor regression and reversed disease-specic traits to
prompt selective and personalized therapies for colon cancer.
15
In another study, unimer polyion complex (uPIC) was
conjugated on the gold nanoparticles surface. The intravenous
injection of uPIC-AuNPs carrying siRNA signicantly
enhanced the accumulation and penetration of siRNA into
solid tumor with longer blood circulation.
16
Researchers also
invented the rst example of reversible ligation of DNA on gold
nanoparticles. This conjugation can protect the DNA from
degradation, and the DNAs can be reversibly released by using
light as an external stimulus, which could potentially be widely
utilized in drug delivery, catalysis, sensing, and photonics.
17
All
this evidence reminded us of the great application potential of
gold nanoparticles in gene therapy.
Received: December 21, 2016
Accepted: March 2, 2017
Published: March 2, 2017
Research Article
www.acsami.org
© XXXX American Chemical Society ADOI: 10.1021/acsami.6b16378
ACS Appl. Mater. Interfaces XXXX, XXX, XXXXXX
Cutaneous melanoma is one of the most deadly cancers in
clinic that is still lacking eective therapies.
18,19
Advanced
treatment options include chemotherapies, antiprogrammed
death-1, and targeted therapies like BRAF inhibitors.
2022
These treatments have provided new options to treat this
deadly tumor but also cause serious side eects and easily
become drug-resistant. Gene therapy has emerged as a
promising technique for numerous tumor types. In gene
therapy, miRNA possesses the ability to regulate more than one
gene, which is dierent from siRNA and may change multiple
tumor-associated moleculessimultaneously.Muchmore
evidence has suggested that melanoma is characterized by
distinct molecular mutations, which provide unique oppor-
tunities for targeted therapy. Recently, microRNA-221
(miRNA-221) was identied to be abnormally expressed in
malignant melanoma cells, and it favors the induction of the
malignant phenotype through down-modulation of c-Kit
receptor and blocking p27 translation.
23
In the progression of
melanoma, up to 70% of metastases lack the c-Kit receptor and
can consequently escape c-Kit-triggered apoptosis.
24
Similarly,
p27 expression is lost during progression from benign nevi to
metastatic cells, and its reduction causes the poor survival,
25
because microRNA-based short hairpin still suers from
limitations such as stability. DNA-based RNAi drugs, however,
have the potential of being stably introduced when used in
plasmid DNA.
26
Antisense sequence of miRNA-221 was
constructed and inserted into the plasmid to reduce the
expression of miRNA-221, based on the transfection of pDNA
into B16F10 cells. It was demonstrated that repression of p27 is
a consequence of direct binding of miRNA-221 sites in the
3UTR.
27
It was shown that not only miR-222 but also miRNA-
221 was able to reduce viability and induce apoptosis mediated
by the KIT, AKT, and BCL2 signaling cascade.
28
MiRNA-221
has been proposed as a potential tumor suppressor for
melanoma therapy. Therefore, the inhibition of miRNA-221
expression that up-regulates the c-Kit receptor and p27 protein
is thought to be a novel treatment for advanced melanoma with
clinical translation.
29,30
Moreover, topical delivery miRNA-221
inhibitor gene can avoid or decrease reticuloendothelial system
(RES) uptake, reduce systemic toxicity, and provide targeted
gene delivery to the tumor site located at the skin subcutaneous
layer. However, stratum corneum always poses a formidable
challenge to bio-macromolecules penetration. PDNAs, because
of their large size, hydrophilic nature, and fast degradation, are
normally precluded from percutaneous absorption.
To circumvent these problems that confront the current
methods, herein, for the rst time, we present a novel strategy
for the cutaneous melanoma therapy by topical delivery of a
pDNA encoded with miRNA-221 inhibitor gene through HIV-
1 twin-arginine translocation peptide (TAT) conjugated
catonic gold nanoparticles (AuPT). In this strategy, AuPT
acts as not only the topical carrier but also the gene vector for
pDNAs. TAT as a vehicle for drug delivery has been thoroughly
investigated by many studies.
31
It was shown to be non-
immunogenic, and its use in cells and animals did not elicit
toxic responses.
32
By virtue of their strong electrostatic
interaction with anionic membrane surfaces and their some-
times amphipathic nature, TAT peptides oer interfacial
activitydened as the ability to bind at the bilayerwater
interface and perturb membrane structure.
33
It can coopera-
tively disrupt the vertical segregation of hydrophobic and
hydrophilic groups in a bilayer and allows the passage of polar
molecules across the membrane.
34,35
On the one hand, TAT
was so suggested as a skin-permeable protein, and it has been
demonstrated to transport the attached proteins into the skin
for strong transcutaneous immunization.
36
On the other hand,
by transient plasma membrane disruption
37
or spontaneous
translocation,
38
TAT can help the bio-macromolecules bypass
Figure 1. Schematic illustration of the transdermal delivery of pDNAs encoding microRNA-221 inhibitor gene (Mi221) by AuPT nanoparticles for
skin cutaneous melanoma treatment. The therapy consists of four major steps, including (A) preparation of AuPT/Mi221 nanocomplexes; (B)
topical application of AuPT/Mi221 and the skin penetration of AuPT/Mi221; (C) skin penetration into melanoma, and (D) gene transfection of
AuPT/Mi221 in melanoma cells for tumor therapy.
ACS Applied Materials & Interfaces Research Article
DOI: 10.1021/acsami.6b16378
ACS Appl. Mater. Interfaces XXXX, XXX, XXXXXX
B
the endosomal degradative environment.
3941
In our previous
study, AuPT was shown to be highly ecient in transfecting
pDNAs in both epidermal stem cells and mesenchymal stem
cells for their in vitro directed dierentiation.
42,43
In the present
study, using AuPT as a multifunctional vehicle, the penetration
of pDNAs through the dierent skin layers and the transfection
of these pDNAs in the melanoma cells at the subcutaneous site
were investigated. Briey, both the penetration and distribution
of the pDNAs and gold nanoparticles in the dierent skin layers
after the topical application of AuPT/pDNAs were studied in
vitro and in vivo. Transfection of the pDNAs encoding miRNA-
221 inhibitor gene (Mi221) by AuPT in melanoma cells and
melanoma xenograft in mice, as well as its regulation on c-Kit
and p27 genes expression of cells and tumor tissues, were
evaluated. On the basis of that, the therapeutic eects of AuPT/
Mi221 through topical application were evaluated by
investigating the tumor cell apoptosis, metastasis, and
interference in cell cycles. The histology of the tumor tissues
was also analyzed by hematoxylin and eosin (HE), Tunel, and
EdU staining (see Figure 1AD as a schematic design).
2. EXPERIMENTAL DETAILS
2.1. Materials. Chloroauric acid (HAuCl4), sodium borohydride
(NaBH4), poly(ethelyimine) (25 kD), amiloride-HCl, chlorpromazine
(CPZ), methyl-β-cyclodextrin (MBC), 4,6-diamidino-2-phenylindole
(DAPI), and methylthiazoletetrazolium (MTT) were purchased from
Sigma (Sigma-Aldrich, St. Louis, MO, USA). Micro-bicinchoninic acid
(BCA) protein assay kit was purchased from Beyotime Biotechnology
Inc., China. PDNA encoding luciferase (PGL3) was obtained from
Institute of Infectious Diseases, Zhejiang University, China. PDNAs
encoding GFP and miRNA-221 inhibitor genes were purchased from
Genepharma Company. TAT peptide (H-Cys-Cys-Tyr-Gly-Arg-Lys-
Lys-Arg-Arg-Gln-Arg-Arg-Arg-OH, Mw = 1559), FITC-DNA, Cy5.5-
pDNA were constructed by Sangon Biotechnology Inc., China. Four-
week-old nude mice were supplied by Shanghai SLAC Laboratory
Animal Co. Ltd., China. B16F10 cell line was purchased from the
Institute of Biochemistry and Cell Biology, Chinese Academy of
Sciences (China). Dulbeccos modied Eagles medium (DMEM),
0.25 wt % trypsin with 0.02 wt % ethylenediaminetetraacetic acid, fetal
bovine serum (FBS), streptomycin, and penicillin were obtained from
Gibco BRL (USA).
2.2. Synthesis of AuPT. HAuCl4(150 μg/mL) was reduced by
NaBH4(10 mg/mL) in the presence of PEI (3 mg/mL). The reaction
solution was stirred vigorously for 15 min at room temperature and
stored for at least 1 h to get the AuP suspension. Then AuPT was
prepared by adding TAT into the AuP solution under continuous
stirring overnight. TAT peptides were mixed with AuP (100 μg/mL)
for ligand exchange, with a variable peptide concentration of 0100
μg/mL. Excess PEI and TAT were removed by dialysis.
2.3. Characterization of AuPT and AuPT/pDNAs Nano-
complexes. To quantify the PEI amount on the AuP nanoparticles,
thermogravimetric analysis was performed for powder samples using a
TGA/SDTA851, SWRTZER LAND thermogravimetric analyzer.
Samples between 5 and 15 mg were heated from 30 to 400 °Cata
heating rate of 10 °C/min in air. TGA derivative curves show distinct
transitions for dierent samples between 30 and 400 °C. To quantify
the TAT amounts on the AuPT nanoparticles, micro-BCA assay was
used to detect the amount of TAT peptide on the surface of
nanoparticles.
44
Briey, 20 μL of various AuPT aqueous samples
containing numerous amounts of peptides were added into 96-well
plates. Each well was lled with 200 μL of micro-BCA working liquid
followed by incubating them at 37 °C for 30 min. Absorbance was
measured at 570 nm by microplate spectro-photometer. TAT
concentration of the sample was determined according to the standard
curve based on protein standard solution. The concentration of AuPT
was measured by inductively coupled plasma mass spectrometry (ICP-
MS) and adjusted to 100 μg/mL. Then dierent volume of
nanoparticle solution was added into a persistent bulk of pDNAs
solution (100 μg/mL) to prepare AuPT/pDNAs nanocomplexes. The
mixture immediately vortexed for 15 s and incubated for 30 min at 37
°C. The morphologies of AuPT and AuPT/pDNA nanocomplexes
(1:8, w/w) were observed by a JEM-1200EX transmission electron
microscope (TEM). The polyplex diameters were measured by
dynamic light scattering (DLS) on a Brookhaven particle size analyzer
(90plus) at room temperature. An aqueous dip cell in the automatic
mode (Zetasizer 3000, Malvern Instruments, Southborough, MA) was
used to measure the zeta potential of nanoparticles. All measurements
were performed three times.
2.4. Transfection Assay. Reporter gene pDNA-PGL3 (PGL3)
was used for the transfection assay. B16F10 cells (5 ×104) were seeded
on 24-well plate. After cells reached to 80% conuence, the cultural
medium was removed, and cells were washed with phosphate-buered
saline (PBS) twice. Each well received 0.5 mL of DMEM without FBS.
Then dierent groups of AuPT/PGL3 containing 1 μg of PGL3 and
various amounts of AuPT were added to cells. Cells and nano-
complexes were incubated for 6 h at 37 °C before the cultural medium
was changed by DMEM containing FBS to remove the AuPT/PGL3
nanocomplexes. The cells were incubated for another 18 h and washed
with PBS twice. A luciferase assay kit (Beyotime, China) and a
luminometer (Promega, USA) were used to measure the PGL3
luciferase intensity. BCA protein assay reagent kit was used to measure
the total protein of each well. Final results were expressed as luciferase
intensity of per milligram of total protein of the tested cells.
2.5. Cellular Uptake. B16F10 cells were seeded on 24-well plate
with DMEM containing 10% FBS. After cells were incubated overnight
reaching 80% conuence, the medium was changed by DMEM
without FBS. Then the vector/FITC-pDNAs nanocomplexes or naked
FITC-pDNAs were added to each well. Cells were incubated with
nanocomplexes for various time periods. Before measuring the cellular
uptake of FITC-pDNAs, cells were washed twice with PBS, and ow
cytometry was used to determine the mean uorescence intensity per
cell. To directly observe the cellular uptake of nanocomplexes at
dierent time points, cy5.5-labeled pDNAs were used and tracked by
confocal laser scanning microscopy (CLSM). Briey, 3 ×104 B16F10
cells were seeded on 15 mm plate and incubated overnight. Then the
cultural solution was replaced by medium without FBS. Naked cy5.5-
labeled pDNAs or their nanocomplexes with AuPT were added to the
cells and were observed at 1, 3, 6, and 9 h, respectively. After that, cells
were washed twice and incubated with 4% paraformaldehyde and
DAPI for 15 min. Finally, cells were washed with PBS three times and
observed by CLSM.
2.6. Intracellular Pathway of AuPT/pDNAs Nanocomplexes.
For the illumination of the cellular uptake mechanism, cells were
treated with 4 °C to clarify energy dependence. Pharmacological
inhibitors like 50 μM amiloride-HCl, 10 μg/mL of chlorpromazine
(CPZ), and 10 mg/mL of methyl-β-cyclodextrin (MBC) were also
utilized for the mechanism studies. Briey, B16F10 cells were
pretreated with these inhibitors for 30 min at 37 °C. Cells were
then incubated with AuPT/GFP (pDNAs encoded with GFP)
nanocomplexes for 6 h; then, the medium was replaced by DMEM
with 10% FBS. After another culture of 18 h, mean uorescence
intensity per cell was accessed by ow cytometry.
2.7. Analysis of in Vitro and in Vivo Gene Expression. Total
RNA was extracted from B16F10 cells or melanoma tissues using
DxGeneTM Tissue and Cell Total RNA Extraction Kit (GenePharma,
China). MiRNA-221 was quantied by Hairpin-itTM miRNAs qPCR
quantitation kit (GenePharma, China). As for the analysis of c-Kit and
P27 expression, the primers and glyceraldehyde phosphate dehydro-
genase (GAPDH) were synthesized by Sangon Biotech (Shanghai,
China). The expressions of miRNA-221, c-Kit, and P27 were detected
with the CFX-Touch PCR detection system (Bio-Rad Laboratories,
CA, USA) complying with the manufacturers instructions.
2.8. Antiproliferation and Apoptosis Assays. B16F10 cells
were seeded on 96-well plate at a density of 1 ×104 cells/well and
cultivated overnight. The medium was replaced by fresh Opti-MEM
medium, and 10 μL of complexes solution with dierent
concentrations were added to each well cultivating for 6 h. Cells
ACS Applied Materials & Interfaces Research Article
DOI: 10.1021/acsami.6b16378
ACS Appl. Mater. Interfaces XXXX, XXX, XXXXXX
C
were incubated for 18 h further. Then the medium was replaced with
DMEM containing 3-[4, 5-dimethyl-thiazolyl-2]-2, 5-diphenol tetra-
zolium bromide (MTT, Sigma; 0.5 mg/mL). After 4 h, the supernatant
was removed, and 200 μL of dimethyl sulfoxide (DMSO, Sigma) was
added to each well. Then the plate was micro-oscillated for 30 s, and
absorbance was measured at 570 nm. The cell viability was normalized
to that of only solvent-treated cells.
2.9. Cell Cycles Analysis. After B16F10 cells were transfected
with Mi221 as mentioned above, cells were collected and xed with
70% ethanol in 4 °C overnight. After they were washed twice with
PBS, cells were mixed with 100 μL of RNase in 37 °C for 30 min.
Then 400 μL of propidium (PI) was added in 4 °C in the dark and
incubated with the cells for another 30 min. Results were determined
by ow cytometry at the wavelength of 488 nm.
2.10. Cells Migration Assay. After dierent groups of trans-
fection, the cell migration was tested by Transwell assay. B16F10 cells
were digested and resuspended in serum-free medium at a density of 3
×105 cells per milliliter, and 100 μL of the cell suspension was seeded
in the upper chamber. Medium containing 10% FBS was added to the
lower chamber. The chambers were incubated at 37 °C for 72 h. After
the incubation, the medium and cells remaining in the upper chamber
were removed. Finally, crystal violet (Beyotime, Shanghai, China) was
used to stain cells left on the lower side of the membrane for 20 min.
Cells were observed and counted by light microscope (Nikon, Japan).
2.11. In Vitro Skin Penetration Test. The skins of nude mice
(four weeks old) were used for in vitro penetration test with Franz
diusion cells. Mice were anesthetized using 10% chloral hydrate, and
then the dorsal skin hair of all mice was removed using hair removal
cream. The skin was used once it was removed from mice. Skin was
cut to a suitable size and mounted on the receptor compartment of the
diusion cell containing PBS buer (pH 7.4). The receptor was
maintained at 37 °C and constantly stirred with a magnetic bar. The
amount of total FITC-pDNAs was 10 μg, and the ratio between
vectors to FITC-pDNAs was 1:8 (w/w). The concentration of vectors
was adjusted to 100 μg/mL. Then dierent vectors solution was added
into FITC-pDNAs solution (100 μg/mL), incubating for 30 min
before adding into donor cells. PBS was added to the diusion
chamber without bubble. Vector solution loaded with FITC-pDNAs
(0.5 mL) was added to each donor cell. Samples (100 μL) were
withdrawn from diusion chamber at dierent intervals (1, 3, 6, 9, and
24 h), and fresh PBS was replenished. The samples were extracted
onto 96-well solid black microplate, and then uorescence intensity
was detected by uorescence microplate reader. Other skin samples
that treated with FITC-pDNAs were xed to glass coverslips and
observed by CLSM at 24 h.
2.12. Penetration and Distribution of Vector/pDNAs Nano-
complexes in Xenograft Tumor Tissue. Transdermal and
distribution of vector/pDNA in the tumor tissues were imaged with
TEM. For the TEM observation, tumor samples were washed twice
with PBS and then xed for 1 h with 3.5% (v/v) glutaraldehyde.
Postxation was performed for 1.5 h in 1% (v/v) osmium tetroxide at
room temperature. The samples were dehydrated in graded series of
ethanol and propylene oxide. Then the samples were embedded in
Durcupan (Fluka, Sigma-Aldrich). Thickness of section was 60 μm.
Sections were mounted on nickel grids and stained with uranyl acetate
before examination under a JEM-1200EX microscope.
2.13. In Vivo Transdermal Delivery of Mi221 and Anti-
Tumor Eects. Nude mice (four weeks old) were purchased from
Figure 2. (A) Weight loss of AuPT nanoparticle at 400 °C with dierent TAT feeding concentrations. (B) TAT contents on the AuPT
nanoparticles. (C) Diameters and polydispersity indices of the AuPT nanoparticles. (D) Zeta potentials of AuPT nanoparticles. (E) Mean
uorescence intensity per cell (derived from the expressed GFP in transfected cells) of B16F10 cells after transfection at dierent TAT
concentrations, **p< 0.01. (F) Diameters of AuPT/pDNA nanocomplexes at dierent weight ratios of vector to pDNA.
ACS Applied Materials & Interfaces Research Article
DOI: 10.1021/acsami.6b16378
ACS Appl. Mater. Interfaces XXXX, XXX, XXXXXX
D
Shanghai SLAC Laboratory Animal Co. Ltd., China. All animal
experimental procedures were performed in obedience to guidelines
and protocols of the Animal Experimental Ethics Committee of
Zhejiang University. B16F10 cells (1 ×106)wereinoculated
subcutaneously at right ank of nude mice to set up the melanoma
model. The melanoma-bearing nude mice were then randomly divided
into six groups, and each group included six nude mice. For the
preparation of AuPT/Mi221, AuP/Mi221, and AuPT/shNC samples,
vectors were puried by dialysis to remove excess ligand molecules.
The concentrations of AuP and AuPT were adjusted to 100 μg/mL
and veried by inductively coupled plasma mass spectrometry (ICP-
MS). Vectors (12.5 μL) were added into pDNAs solution including 10
μg of pDNAs, vortexed for 15 s, and incubated for 20 min prior to
applying to the dorsal skin of the mice, under which the melanoma
xenograft existed. Nude mice were given topical treatment with AuPT/
Mi221, AuP/Mi221, AuPT, AuPT/shNC, naked Mi221, or PBS,
respectively, twice a day. The therapy dose was 100 μL containing 10
μg of Mi221 or shNC. The body weights of nude mice were tested
every day. All mice were well-tolerated with the tested treatments over
the course of research with no noticeable body weight loss or any signs
of poisonousness such as diarrhea or edema.
2.14. H&E, TUNEL, and EdU staining. On the last day of
treatment, tumors were collected and prepared into paran and frozen
in slices 6 μm thick by standardized protocols. Afterward, the paran
slices were analyzed by H&E and TUNEL staining. The processed
paran slices were imaged and analyzed using a light microscope.
Frozen slices were xed using 4% paraformaldehyde. Consistent with
the manufacturersinstructions(Beyotime,Shanghai,China),
uorimetric TUNEL (TdT-mediated dUTP Nick-End Labeling)
staining was used to detect the presence of apoptotic cells. The brief
procedure was that frozen sections were treated with 20 μg/mL of
proteinase K. Then a nucleotide mixture of uorescein-12-dUTP and
terminal deoxynucleotidyl transferase (TdT) was added to the frozen
sections incubating for 90 min. After cell nucleus was stained by DAPI
(1 μg/mL), uorescence images of apoptotic cells (red) and cell nuclei
(blue) were obtained by CLSM analysis. EdU (keyFluor488 Click-iT
EdU Kit, keygen BioTECH, Nanjing, China) was used to label cells
nuclei that have undergone S phase of DNA synthesis. Briey, tumor
sections were xed with 4% paraformaldehyde for 15 min. Next, Click-
iT reaction mixture containing CuSO4, keyFluor 488 azide, and
buering was prepared according to the instructions of Click-iT EdU
Kit. Then the prepared mixture was incubated with each tumor section
for 30 min in darkness. Sections were nally washed with PBS twice
and observed under CLSM.
2.15. Statistical Analysis. Data are expressed as mean ±standard
deviation (SD). A statistically signicant value was set as p< 0.05
based on the Studentsttest.
3. RESULTS AND DISCUSSION
3.1. Characterization of AuPT Nanoparticles and
AuPT/pDNAs Polyplexes. PEI was proved to be densely
conjugated on the surface of gold nanoparticles (Figure 2A),
indicating 150 PEI molecules were covered on the gold
surface (0.8 molecule per square nanometer). PEI conjugated
to the nanogold could stabilize the hydrophobic gold
Figure 3. (A) Luciferase activity per milligram of total protein expressed by the transfected B16F10 cells with the tested vectors/pDNAs. (B) Mean
FITC uorescence intensity per B16F10 cell after incubation with various vectors/FITC-pDNAs. Co-localization of the cy5.5-pDNAs and DAPI
stained nucleus at 1, 3, 6, and 9 h of the naked pDNAs (C1C4), PEI/pDNAs (D1D4), AuP/pDNAs (E1E4), and AuPT/pDNAs (F1F4) treated
group, respectively. Scale bar is 10 μm. (G) B16F10 cells were pretreated with various endocytosis inhibitors and then transfected with AuPT/
pDNAs. *and ** indicate the signicant dierence at p< 0.05 and p< 0.01, respectively.
ACS Applied Materials & Interfaces Research Article
DOI: 10.1021/acsami.6b16378
ACS Appl. Mater. Interfaces XXXX, XXX, XXXXXX
E
nanoparticles and kept the nanogold colloid system stable. TAT
peptides could replace PEI on the surface of gold nanoparticles
under the reaction of ligand exchange as TAT peptide
sequences contain thiol. During ligand-exchange process, the
capping ligands are displaced by thiol-TAT due to a stronger
AuS linkage and an energy gain associated with the
intermolecular interaction. And this stronger AuS linkage
has been proved by many scientists in previous studies.
45,46
As
the result of ligand exchange with TAT peptide, the amount of
TAT on AuPT increased from 0% to 6.7% with the TAT
feeding concentration increased (Figure 2B). No signicant
change was observed for the diameters and zeta potentials of
the AuP nanoparticles after the TAT anchoring on them
(Figure 2C,D). So, AuP nanoparticles with dierent amounts of
TAT conjugation were further tested with transfection
eciency in B16F10 cells to determine the best TAT feeding
concentration. It was found that highest uorescence intensity
derived from GFP expression appeared when TAT concen-
tration reached 25 μg/mL (Figure 2E). The results
demonstrated that certain amount of TAT could help improve
the transfection eciency, while too much TAT may exchange
more PEI molecules and reduce the ability of AuPT in gene
delivery. As Duchardt et al. had investigated the inuence of
dierent kinds of cell-penetrating peptides (concentration)
including TAT in cellular uptake, it was found that when cells
were incubated with the TAT at concentrations of 15μM
(equal to 1.57.5 μg/mL), the peptide was kept being located
only in vesicles, which could not enhance the transfection
eciency signicantly. However, above a concentration thresh-
old of 240 μM, which was nearly equivalent to 360 μg/mL,
this peptide could, on the one hand, internalize predominantly
through a process that leads to a rapid distribution of peptides
into the cytoplasm and nucleus.
47
On the other hand, according
to our previous study,
42
too much TAT will inhibit the
transfection eciency. Therefore, this concentration range of
0100 μg/mL was selected for investigation in the present
study. However, according to Figure 2F, diameters of AuPT/
pDNAs nanocomplexes decreased with the increase of AuPT.
The smallest diameter of AuPT/pDNAs polyplex was 199 ±
7.76 nm, and PDI was 0.27 ±0.02 when the weight ratio of
AuPT to pDNA was 1:8.
3.2. In Vitro Transfection of B16F10 with Reporter
pDNAs. B16F10 is known as a hard-to-transfect murine tumor
cell line.
48,49
However, from Figure 3A, transfection eciency
of AuPT in B16F10 reached highest of 1.71 ×107RLU/mg
total protein when the ratio of vector to pDNA is 1:8 (w/w).
The zeta potential of the nanocomplexes with this ration was
16.81 ±0.56 mV. Along with the priority of nanoparticles with
small size in transdermal delivery, this ratio was further
investigated in the following study. This transfection eciency
is signicantly higher than those of AuP, PEI, and Lipofect-
amine 2000 (Lipo 2k) by 1.3-fold (p< 0.01), 2.5-fold (p<
0.01), and 1.9-fold (p< 0.01), respectively. To clarify the
transfection mechanism, cellular uptake kinetics, nuclear
targeting property, and intracellular pathway of AuPT/
pDNAs in B16F10 were investigated. As shown in Figure 3B,
the average uorescence intensity per cell increased along with
the prolongation of culture time and took up much more
pDNAs than the naked pDNAs, PEI/pDNAs, and AuP/pDNAs
treated groups within the tested 9 h. This might be attributed to
the attachment of TAT peptides on the AuP surface, which
resulted in higher TAT local concentration and subsequently
enhanced cell penetrating ability. This consequence was also
clearly observed in CLSM images. As shown in Figure 3CF,
cells obviously ingested much more uorescent cy5.5-pDNAs
(Figure 3F1F4) with many localized at the nucleus, as white
arrows indicated. By contrast, no obvious uorescence in
B16F10 cells could be observed in the naked pDNA-treated
group (Figure 3C1C4). Even with the PEI/cy5.5-pDNAs
(Figure 3D1D4) and AuP/cy5.5-pDNAs (Figure 3E1E4)
treatment, less cy5.5-pDNAs were identied in the cytoplasm
without nuclear targeting property (Figure 3D1D4,E1E4).
These results reminded us that AuPT could disrupt the
endosomal membrane and release the pDNAs to cytoplasm, as
well as translocate them to the nucleus. The endosome escape
eect of AuPT might be partly contributed to the conjugation
of PEI, which was a well-known polymer capable of protonation
and can disturb the lysosomes.
50
A comprehensive evaluation of
the PEI/TAT/pDNA (as a control group), including the
characterization, toxicity, transfection eciency, and targeting
ability to cell nucleus was investigated in our previous study.
42
It was shown that the PEI-TAT/pDNA complexes had lower
zeta potential, larger particle size, higher toxicity, and lower
transfection eciency in cells than Au-PEI-TAT/pDNA.
AuPT/pDNA-based vectors can deliver pDNA not only into
cells but also target to cell nucleus. On the contrary, the
uorescence observed in PEI-TAT/pDNA group expressed
limited cell penetrating and gene transfection ability, although
they have similar ability to condense pDNA and generate
vector/pDNA complexes. The results suggested that only
tethered TAT molecules on AuP gold nanoparticle can
generate pores on cell membrane and facilitate cellular uptake
and gene transfection due to locally enhanced concentration.
Additionally, the larger particle size of PEI-TAT/pDNA
complexes was not advantageous for the transdermal eciency.
PEI/TAT/pDNA was so not set as a control group in this
study.
Pharmacological inhibitors are often used to investigate
which endocytic mechanism is responsible for cellular uptake of
the various nanoparticles. Dierent pharmacological inhibitors
have been discussed and summarized by Iversen. Many
endocytosis pathways have been reported including caveolae
pinching, RhoA-mediated endocytosis, raft-mediated endocy-
tosis, macropinocytosis, and clathrin-independent and choles-
terol-dependent endocytosis.
51
Several dierent types of
endocytosis have been well-investigated in the intracellular
mechanisms of vesicles derived from plasma membrane, which
are clathrin-independent endocytosis (CIE), raft-mediated
endocytosis, and micropinocytosis. Especially, methyl-β-cyclo-
dextrin (MBC) is a cyclic oligomer of glucopyranoside that
inhibits cholesterol-dependent endocytic processes by rever-
sibly extracting the steroid out of the plasma membrane. MBC
is regularly used to determine whether endocytosis is
dependent on the integrity of lipid rafts.
52
Chlorpromazine is
a cationic amphiphilic drug that is believed to inhibit clathrin-
coated pit formation by a reversible translocation of clathrin
and its adapter proteins from the plasma membrane to
intracellular vesicles.
53
It has been known for years that
amiloride and amiloride derivatives block micropinocytosis.
These chemicals, known to inhibit the Na+/H+ exchanger,
were shown to inhibit the initiation of micropinocytosis by
lowering the pH locally, close to the membrane, and thereby
inhibit recruitment of Rac.
54
From Figure 3G, with 4 °Cor
methylated β-cyclodextrin pretreatments, the transfection
eciency of AuPT/pDNA was signicantly decreased. These
results indicated that caveolin-mediated endocytotic pathway
ACS Applied Materials & Interfaces Research Article
DOI: 10.1021/acsami.6b16378
ACS Appl. Mater. Interfaces XXXX, XXX, XXXXXX
F
also contributed to the ingestion of AuPT/pDNA in melanoma
cells besides energy-dependent process.
PEI-capped gold nanoparticles can increase the transfection
eciency compared with PEI. This result had been previously
proved,
42
and the mechanism has been explained by some
scientists.
55,56
(1) The conjugating PEI to gold nanoparticles
would increase its eective molecular weight, consequently
enhancing pDNA binding and condensation and therefore
improving the transfection. (2) The super gene transfection
performance is also likely due to the biomimetic design of the
particles that has a size more or less similar to the nucleosome
core proteins, having a large proportion of basic residues that
form electrostatic bonding with the phosphate backbone of
pDNA. Those PEI-capped gold nanoparticles may redirect the
intracellular distributions of nucleic acid drug and enhance the
gene therapy eect of nucleic acid drug compared with PEI. (3)
PEI-capped gold nanoparticles showed good cytocompatibility,
which suggests that PEI-capped gold nanoparticles are suitable
for transfecting cells with lower toxicity leading to better
cellular state.
3.3. Gene Interference in B16F10 by Mi221 Trans-
fection. To investigate whether Mi221 could regulate gene
expression of melanoma cells, resulting in inhibition of cell
cycles and cell migrations to induce cytotoxicity in vitro,
dierent groups carrying Mi221 treated B16F10 cells. The
results were in agreement with gene transfection consequences
mentioned above. From Figure 4A,B, the AuPT/Mi221
transfection signicantly decreased the miRNA-221 level
along with the obvious up-regulation of c-Kit and p27
expression in B16F10 cells. However, PEI/Mi221 and AuP/
Mi221 groups did not express regulating ability as eectively as
AuPT/Mi221. In addition, control groups nearly had no
inuence on B16F10 cells, which proved the regulation of
miRNA-221 was intervention result of Mi221. It has been
found that inhibition of c-Kit receptor permits uncontrolled cell
proliferation leading to melanoma progression.
57
P27 plays an
important role in regulating the cell cycle transition from G1 to
Figure 4. (A) MiRNA-221 gene expression levels, (B) P27 and c-Kit genes expression in the melanoma cells after 24 h of transfection. (C) Cell
cycles percentage of the tested cells after transfection. (D) Cell viability after the 24 h of transfection. (E) Cells migrations after the 24 h of
transfection. (F) Migrated cell numbers in each group were quantied. Scale bar = 100 μm. *and ** indicate signicant dierence at p< 0.05 and p
< 0.01, respectively.
ACS Applied Materials & Interfaces Research Article
DOI: 10.1021/acsami.6b16378
ACS Appl. Mater. Interfaces XXXX, XXX, XXXXXX
G
S phase.
58
From Figure 4C, in contrast to the normal cells,
PEI/Mi221 and AuP/Mi221 treated cells, of which the cell
distributions in G1 and S phases were 70.0% and 20.0%, the
AuPT/Mi221 transfected cell distributions in G1 and S phases
were 79.3% and 14.1%, respectively, demonstrating more cells
were detained in G1 phases. This result gave evidence that
much more AuPT/Mi221 treated B16F10 cells were retarded
in G1 phase and that the cells in S phase were signicantly
decreased. Subsequently, cell viability was decreased to 68.7%,
62.2%, and 48.2%, respectively, for the PEI/Mi221, AuP/
Mi221, and AuPT/Mi221 treated cells (Figure 4D). As the
AuPT vector control, AuPT/shNC and naked Mi221 had no
cytotoxicity (Figure 4D), the antiproliferation eect was
identied to be induced by the decrease of miRNA-221 level
and the up-regulation of c-Kit and p27 genes, which initiated
the apoptosis.
B16F10 is a melanoma cell with highly aggressive property
and may metastasize from a primary subcutaneous site to the
lungs, bones, etc.
59
The invasion of the transfected cells was
evaluated by transwell assay. From Figure 4E,F, in contrast to
the blank control, cells treated with naked Mi221 or AuPT/
shNC, the migration of PEI/Mi221, AuP/Mi221, and AuPT/
Mi221 transfected cells were signicantly decreased with 79.1%,
85.0%, and 89.1%, respectively, as black arrows headed. This
Figure 5. Skin penetration of the FITC-pDNAs delivered in vitro and in vivo. (A) TEM images of AuPT nanoparticles, (B) AuPT/pDNAs
nanocomplexes, and (C) higher magnication micrograph of AuPT/pDNAs nanocomplexes. (D) CLSM images of the lateral skin sections. Scale bar
= 100 μm. (E) Quantitative analysis of the FITC-pDNAs that had penetrated through the skins by in vitro skin diusion test. Data refer to average
value of three times. TEM images of the melanoma xenograft tissues collected from the mice after treated with blank (F1,G
1), naked Mi221 (F2,G
2),
AuPT (F3,G
3), AuPT/shNC (F4,G
4), AuP/Mi221 (F5,G
5), and AuPT/Mi221 (F6,G
6). Black arrows indicated the Au nanoparticles distributed in
melanoma tissues. Scale bars of F1F6and G1G6represent 2 μm and 200 nm, respectively. ** indicates signicant dierence at p< 0.01.
ACS Applied Materials & Interfaces Research Article
DOI: 10.1021/acsami.6b16378
ACS Appl. Mater. Interfaces XXXX, XXX, XXXXXX
H
migrating inhibition eect of AuPT/Mi221 provides a great
priority for its application in preventing the clinical melanoma
metastasis.
3.4. Skin Penetration of pDNAs by AuPT Delivery.
Round-shaped nanoparticles were also found in skin follicles by
CLSM (Figure 5D), which permits simultaneous examination
of the distribution of uorescent dye inside skin.
8,60
The xz
optical sections of the skin after treatment with FITC-pDNAs
were shown in Figure 5D. Images starting from 40 μm were
collected, because obvious uorescence could be tested until
the depth of z-axle reached 40 μm at the tested time point. In
the naked FITC-pDNAs, PEI/FITC-pDNAs, and AuP/FITC-
pDNAs treated groups, weak FITC uorescence was only
observed at 40 and 60 μm in depth of the skin (Figure 5D).
By contrast, when the same dose of FITC-pDNAs was
condensed by AuPT and applied to the skin, strong
uorescence was observed beyond the epidermis and into the
dermal layer up to 80 μm(Figure 5D). In the images of Figure
5D as white arrows headed, a large amount of round-shaped
AuPT/FITC-pDNAs complexes uniformly dispersed in the hair
follicles without aggregation, which provided robust evidence
that AuPT was an excellent transdermal vehicle for bio-
macromolecules, utilizing the hair follicle duct as a transport
route. Transmission electron microscope (TEM) image (Figure
5A) showed that AuPT nanoparticles are round in shape and
well-dispersed with no aggregation observed. The average
diameter of AuPT is 18 nm. In Figure 5B, AuPT was shown
to closely attach to the surface of pDNA and condense them
into round nanostructures. All the pDNAs were incorporated in
the center of the vector/pDNAs nanocomplexes (see Figure 5C
as an enlarged view), whose round shape was consistent with
images shown in Figure 5D. The plasmid DNA was negatively
charged, so it would be condensed and surrounded by
positively charged gene vector AuPT, whose zeta potential
was 35 mV as shown in Figure 5B,C. Gold nanoparticles are
adsorbed in the outermost layer of AuPT/pDNA complexes, so
the color of the outermost layer was black. However, the pDNA
do not have high electron density; therefore, pDNA was almost
transparent under TEM
61,62
and was wrapped in the core of
complexes. Considering the protection and gene transfection
eects were attributed to PEI and TAT modied on the surface
of gold nanoparticles, PEI and TAT may not dissociate upon
the addition of pDNA. It has also been proved by Z. Chen that
shielded PEI on Au-PEI/pDNA complex not only condensed
pDNA completely but also protected DNA from DNase I
enzymatic degradation.
63
In conclusion, these nanostructures
will protect nucleic acids from enzymatic degradation, increase
the bioavailability of miRNA-221 inhibitor, and enhance their
target delivery to specic cells.
As we all know, stratum corneum is a challenging barrier for
the transdermal/topical drug delivery. Hair follicles directly
penetrate into the dermal layer, which could help the
nanoparticles reach deeply into the subcutaneous tissues.
6466
For the transdermal mechanism of the cationic gold nano-
particles, on the one hand, AuP and AuPT can interact with the
negatively charged skin lipids by electrostatic interaction, and
TAT may help cause membrane destabilization. On the other
hand, the small size of the metal gold particles with good
stability is also advantageous for the skin penetration. As shown
in Figure 5D, compared with other control groups, AuPT
stimulated much more entrapped FITC-pDNAs to diuse into
the deeper layers of skin tissues. Accordingly, signicantly
higher FITC-pDNAs concentrations were detected in the
diusion solution of AuPT/FITC-pDNAs than that of other
control groups within the 24 h (Figure 5E). In vivo studies
were also performed to testify if the AuPT/pDNAs complexes
Figure 6. Antitumor eects in skin melanoma xenograft-bearing mice. (A) Tumor volumes were monitored every day (*p< 0.05). (B) Body weights
were assessed after the rst treatment every day. (C) MicroRNA-221 gene expression level in the tumor tissues collected on day 8 by qRT-PCR (**p
< 0.01). (D) P27 and c-Kit gene expression levels were analyzed for the tumor tissues collected on day 8 by qRT-PCR (**p< 0.01).
ACS Applied Materials & Interfaces Research Article
DOI: 10.1021/acsami.6b16378
ACS Appl. Mater. Interfaces XXXX, XXX, XXXXXX
I
could penetrate deeply into melanoma tissues. With the TEM
observation, from Figure 5F,G, nanocomplexes with several
hundreds of nanometers (black arrows headed) were found to
be localized in the tumor slices of AuPT (Figure 5G3,F3),
AuPT/shNC (Figure 5G4,F4), AuP/Mi221 (Figure 5G5,F5),
and AuPT/Mi221 (Figure 5G6,F6) treated groups, which were
absent in the blank (Figure 5G1,F1) and naked Mi221 (Figure
5G2,F2) control groups. These results together demonstrated
the strong ecacy of AuPT in stimulating the skin penetration
of pDNAs into the subcutaneous tissues.
Gold nanoparticles have been indicated for their nano-
biological interactions with membrane lipids to enhance
transdermal eciency.
67,68
The nanoparticles have also been
proved to modulate membrane lipid phase transitions so as to
increase the lipid uidity.
69,70
Because the skin barrier is
actually governed by the physical state and structural
organization of stratum corneum extracellular lipids, the lipid-
uidizing functions of nanoparticles play an important role in
their ability to alter skin permeability.
71
Metal-based nano-
particles, including gold nanoparticles,
5
quantum dots
72
and
iron oxide nanoparticles,
73,74
were revealed to be capable of
penetrating through intact skin, due largely to the interaction
with the lipid in extracellular space, which was absent for
polymers such as PEI. Hence, the altered condition of skin
accompanied by percutaneous penetration of nanoparticles
leads to the breach of skin barrier as the result of usage of gold
nanoparticles in our study.
3.5. In Vivo Anti-Tumor Eects. As shown in Figure 6A,
despite that no signicance was obtained, AuP/Mi221
expressed a certain tumor inhibition trend. Compared with
the blank group, AuP/Mi221 treatment inhibited the tumor
growth with 11.5%38.7% at the tested period. By contrast, the
tumor inhibition rates expressed by AuPT/Mi221-treated
group were 38.5%52.3%, with signicance received at day 2,
4, and 8 (p< 0.05). AuPT-treated group showed a similar
prole to that of saline group, which indicated that metal
particles themselves have no inuence on the tumor growth.
Neither signicant tumor inhibition was observed in the naked
Mi221 nor AuPT/shNC treated group. On the one hand, these
results provided the evidence that the AuPT/Mi221 trans-
fection played important role in the antitumor eect. Of which,
TAT was critical in promoting the skin penetration and gene
transfection of the pDNAs by AuP in topical application. On
the other hand, mice in all the tested groups showed similar
body weights growth (Figure 6B), indicating that the topical
delivery of AuPT/Mi221 and other control groups had no
cytotoxic eect on the animals. The MicroRNA-221 regulated
target genes expression in vivo was also identied and shown in
Figure 6C,D. In accord with the observed delay in tumor
growth, the miRNA-221 expression of melanoma in the AuPT/
Mi221 and AuPT/Mi221 treated groups were inhibited, while
p27 and c-Kit were up-regulated obviously compared with
other control groups.
In keeping with this, a further examination of tumor tissues
was conducted by H&E staining. As shown in Figure 7A1A4,
Figure 7. H&E staining images of the tumor tissues harvested from mice treated with (A1) blank, (A2) naked Mi221, (A3) AuPT, (A4) AuPT/shNC,
(A5) AuP/Mi221, and (A6) AuPT/Mi221, on day 8. Scale bar represents 100 μm. TUNEL staining (red uorescence) images of the tumor tissues
harvested from mice treated with (B1) blank, (B2) naked Mi221, (B3) AuP, (B4) AuPT/shNC, (B5) AuP/Mi221, and (B6) AuPT/Mi221. EdU
staining (green uorescence) images of the tumor tissues harvested from mice treated with (C1) blank, (C2) naked Mi221, (C3) AuP, (C4) AuPT/
shNC, (C5) AuP/Mi221, and (C6) AuPT/Mi221. Scale bar represents 50 μm. (D) Quantication of the number of melanoma cells with positive
stain in TUNEL assay. (E) Quantication of the number of melanoma cells with positive stain in EdU assay. H&E stain was observed by light
microscope. TUNEL and EdU stains were observed.
ACS Applied Materials & Interfaces Research Article
DOI: 10.1021/acsami.6b16378
ACS Appl. Mater. Interfaces XXXX, XXX, XXXXXX
J
no indication of necrotic or apoptotic regions were observed in
the robust tumor tissues of saline, naked Mi221, AuPT, and
AuPT/shNC treated groups. By contrast, signicant cell
necrosis and big vacuoles were observed in the tumors of
AuPT/Mi221 and AuP/Mi221 treated group (Figure 7A5A6,
black arrows headed), which might be contributing to the
apoptosis of cells inside the tumor. The enhanced inhibition in
xenografted melanoma expressed by the AuPT/Mi221 treat-
ment can be partly attributed to the enhanced skin penetration
and increased transfection of Mi221 in the tumor cells. High in
vivo transdermal eciency of AuPT/pDNAs nanocomplexes
had been previously proven in Figure 5F,G.
DNA fragmentation in apoptosis is usually associated with
ultrastructural changes in cellular morphology.
75
Apoptosis
resulting from DNA fragmentation can be examined by TdT-
mediated dUTP Nick-End Labeling (TUNEL) assay. It was
shown that many apoptotic cells indicated by the red
uorescence (white arrows headed) in AuP/Mi221 and
AuPT/Mi221 tumor slices were observed (Figure 7B5B6).
On the contrary, only a small amount of red uorescence was
identied in other four control groups (Figure 7B1B4, white
arrows headed). Quantitatively, there are 14% cell that were
apoptotic in tumor slices of AuPT/Mi221-treated group, which
is 1.3-fold (P< 0.01) and 7-fold (P< 0.01) higher than that of
AuP/Mi221 and control groups, respectively (Figure 7D).
Apoptosis is usually associated with cell cycle inhibition, which
was so examined by Click-iT EdU assay. Particularly, cells in S-
phase of DNA synthesis were stained to express green
uorescence, and subsequent visualization by uorescent
microscopy gives biologically important data about cell cycle.
As shown in Figure 7C1C4, the four control groups
demonstrated massive green uorescence (white arrows
headed), suggesting many cells were in S-phase of DNA
synthesis. A certain amount of proliferation signal was observed
from the AuP/Mi221 and AuPT/Mi221 tumor slices groups,
but the intensity is much less than that of the control groups
(Figure 7C5,C6, white arrows headed). The EdU positive cells
were calculated and shown in Figure 7E. It was found that only
8% melanoma cells of AuPT/Mi221 group were in S phases,
compared with the 10% in AuP/Mi221 group and 18% in
other control groups, suggesting more cells were retarded in G1
phase upon AuP/Mi221 or AuPT/Mi221 treatment. These
results also suggested the highest transfect eciency of AuPT,
which induced the strongest MicroRNA-221 inhibition,
resulting in the inhibition of DNA synthesis in the cells.
Collectively, these results provided further evidence for the
eciency of AuPT as a vehicle for the in vivo transdermal
delivery of pDNAs and as the vector for the pDNAs
transfection in vivo.
Melanoma has poor prognosis with high mortality when
patients are in advanced stages.
7678
Current therapeutic ways
for skin melanoma were usually not eective resulting in only
less than 10% survival rate. Two main reasons explained the
poor eciency. The rst one lies in that chemotherapeutic ways
would easily cause drug resistance and make melanoma
insensitive to them. The other reason is that melanoma is
associated with multiple gene changes, so targeting only one
gene is not easy to receive satisfactory antitumor eects.
23,79
Therefore, in the present study, we use miRNA, which can
control multiple targets involved in melanoma by mainly
targeting c-Kit and p27 simultaneously. Multiple genes
regulation can inuence a series of protein amount in
melanoma cells and change signaling pathways. Such synergy
can achieve better antitumor results theoretically. Whats more,
latest studies have explored the expression of microRNA in
drug-resistant cancer cells. They found drug-resistant mecha-
nism was closely associated with miRNA levels. In two kinds of
drug-resistant cancer cell lines, the expression of a certain
miRNA was signicantly higher than those of drug-sensitive cell
lines.
80
In addition, Denmark Santaris pharma has started the
phase I clinical trials to target miRNA molecules (miR-122),
suggesting miRNA was a promising target in cancer gene
therapy.
81
All this research progress reminded us of the great
potential of miRNA-based novel gene therapy in cancer gene
therapy. In the present study, we aim to deliver miRNA by a
novel strategy, transdermal delivery, for the topical skin cancer.
Until recently, transdermal bio-macromolecules delivery is
reported scarcely and less than the delivery by other strategies
like injection and oral administrations. Siu et al. have recently
reported the design and application of noncovalently function-
alized carbon nanotubes (CNT) in topical siRNA delivery, in
which, PEI was conjuguated to CNT to deliver siRNA.
Through transdermal method, the target gene in the tumor
tissue was silenced eectively.
82
Another study formulated the
eective cationic lipidpolymer hybrid nanoparticles compris-
ing of an anti-inammatory drug Cap and siRNA against TNF-
αto treat dicult skin inammatory conditions in vivo.
83
These
two studies both prepared nanoparticles carrying siRNA with
favorable transdermal ability. But it was known that pDNAs are
used more often in clinic as it remained stable in vivo. However,
transdermal delivery of pDNAs with higher molecular weight
faced much bigger challenge than siRNA. In this study, AuPT
was rst reported to deliver the pDNAs by transdermal
pathway. AuPT played two indispensable roles in not only
enhancing the transdermal ability of pDNAs, making the
AuPT/pDNAs can enter into deep skin layers, but also acting
as the ecient nonviral vector to transfect the melanoma cells.
The in vitro transfection eciency and mechanism of AuPT in
dicult-to-transfect cells (bone marrow derived mesenchymal
stem cells) were investigated in our precious work.
42
In the
present study, the transdermal eciency and in vivo application
of AuPT was systematically investigated. Considering the scarce
report on transdermal pDNA delivery, the present study also
provides, for the rst time, the evidence for the new way in
utilizing the transdermal pDNAs delivery for gene therapy.
4. CONCLUSIONS
Transmembrane peptide-conjugated cationic gold nanoparticles
(AuPT) was shown a highly ecient carrier for the transdermal
delivery of pDNAs. Along with the robust gene transfection
eciency of AuPT, transdermal delivery of AuPT/pDNA-
Mi221 provides a novel topical gene therapy strategy for skin
cancer with great priority to reverse both the progression and
metastasis of advanced melanoma.
AUTHOR INFORMATION
Corresponding Authors
*E-mail: lhpeng@zju.edu.cn. Phone:+86-571-88208437. (L.-
H.P.)
*E-mail: gaojianqing@zju.edu.cn. (J.-Q.G.)
ORCID
Li-Hua Peng: 0000-0001-9763-383X
Notes
The authors declare no competing nancial interest.
ACS Applied Materials & Interfaces Research Article
DOI: 10.1021/acsami.6b16378
ACS Appl. Mater. Interfaces XXXX, XXX, XXXXXX
K
ACKNOWLEDGMENTS
The study was supported by National Natural Science
Foundation of China (Project No. 81473145) and the Macau
Science and Technology Development Fund, Macau Special
Administrative Region, China (Open Project of State Key
Laboratory of Quality Research in Chinese Medicine, Macau
University of Science and Technology, Project No. MUST-
SKL-2016-11).
REFERENCES
(1) Li, N.; Peng, L.; Chen, X.; Nakagawa, S.; Gao, J. Transcutaneous
Vaccines: Novel Advances in Technology and Delivery for Over-
coming the Barriers. Vaccine 2011,29, 61796190.
(2) van Rooy, I.; Mastrobattista, E.; Storm, G.; Hennink, W.;
Schiffelers, R. Comparison of Five Different Targeting Ligands to
Enhance Accumulation of Liposomes into the Brain. J. Controlled
Release 2011,150,3036.
(3) Chen, Z.; Zheng, J.; Yuan, M.; Zhang, L.; Yuan, W. A Novel
Topical Nano-Propranolol for Treatment of Infantile Hemangiomas.
Nanomedicine 2015,11, 11091115.
(4) Lakshmanan, S.; Gupta, G.; Avci, P.; Chandran, R.; Sadasivam,
M.; Jorge, A.; Hamblin, M. Physical Energy for Drug Delivery;
Poration, Concentration and Activation. Adv. Drug Delivery Rev. 2014,
71,98114.
(5) Fernandes, R.; Smyth, N.; Muskens, O.; Nitti, S.; Heuer-
Jungemann, A.; Ardern-Jones, M.; Kanaras, A. Interactions of Skin
with Gold Nanoparticles of Different Surface Charge, Shape, and
Functionality. Small 2015,11, 713721.
(6) Li, N.; Peng, L.; Chen, X.; Zhang, T.; Shao, G.; Liang, W.; Gao, J.
Antigen-Loaded Nanocarriers Enhance the Migration of Stimulated
Langerhans Cells to Draining Lymph Nodes and Induce Effective
Transcutaneous Immunization. Nanomedicine 2014,10, 215223.
(7) Chen, M.; Lin, Z.; Ling, M. Near-Infrared Light-Activatable
Microneedle System for Treating Superficial Tumors by Combination
of Chemotherapy and Photothermal Therapy. ACS Nano 2016,10,
93101.
(8) Chen, Y.; Cun, D.; Quan, P.; Liu, X.; Guo, W.; Peng, L.; Fang, L.
Saturated Long-Chain Esters of Isopulegol as Novel Permeation
Enhancers for Transdermal Drug Delivery. Pharm. Res. 2014,31,
19071918.
(9) Rao, Y.; Chen, W.; Liang, X.; Huang, Y.; Miao, J.; Liu, L.; Lou, Y.;
Zhang, X.; Wang, B.; Tang, R.; Chen, Z.; Lu, X. Epirubicin-Loaded
Superparamagnetic Iron-Oxide Nanoparticles for Transdermal Deliv-
ery: Cancer Therapy by Circumventing the Skin Barrier. Small 2015,
11, 239247.
(10) Guo, L.; Yan, D.; Yang, D.; Li, Y.; Wang, X.; Zalewski, O.; Yan,
B.; Lu, W. Combinatorial Photothermal and Immuno Cancer Therapy
Using Chitosan-Coated Hollow Copper Sulfide Nanoparticles. ACS
Nano 2014,8, 56705681.
(11) Zheng, D.; Giljohann, D.; Chen, D.; Massich, M.; Wang, X.;
Iordanov, H.; Mirkin, C.; Paller, A. Topical Delivery of Sirna-Based
Spherical Nucleic Acid Nanoparticle Conjugates for Gene Regulation.
Proc. Natl. Acad. Sci. U. S. A. 2012,109, 1197511980.
(12) Yi, X.; Zhao, G.; Zhang, H.; Guan, D.; Meng, R.; Zhang, Y.;
Yang, Q.; Jia, H.; Dou, K.; Liu, C.; Que, F.; Yin, J. Mitf-Sirna
Formulation Is a Safe and Effective Therapy for Human Melasma. Mol.
Ther. 2011,19, 362371.
(13) Winstel, V.; Kuhner, P.; Krismer, B.; Peschel, A.; Rohde, H.
Transfer of Plasmid DNA to Clinical Coagulase-Negative Staph-
ylococcal Pathogens by Using a Unique Bacteriophage. Appl. Environ.
Microbiol. 2015,81, 24812488.
(14) Smith, L.; Wloch, M.; Ye, M.; Reyes, L.; Boutsaboualoy, S.;
Dunne, C.; Chaplin, J.; Rusalov, D.; Rolland, A.; Fisher, C.; Al-
Ibrahim, M.; Kabongo, M.; Steigbigel, R.; Belshe, R.; Kitt, E.; Chu, A.;
Moss, R. Phase 1 Clinical Trials of the Safety and Immunogenicity of
Adjuvanted Plasmid DNA Vaccines Encoding Influenza a Virus H5
Hemagglutinin. Vaccine 2010,28, 25652572.
(15) Conde, J.; Oliva, N.; Zhang, Y.; Artzi, N. Local Triple-
Combination Therapy Results in Tumour Regression and Prevents
Recurrence in a Colon Cancer Model. Nat. Mater. 2016,15, 1128
1138.
(16) Kim, H.; Takemoto, H.; Yi, Y.; Zheng, M.; Maeda, Y.; Chaya,
H.; Hayashi, K.; Mi, P.; Pittella, F.; Christie, R.; Toh, K.; Matsumoto,
Y.; Nishiyama, N.; Miyata, K.; Kataoka, K. Precise Engineering of Sirna
Delivery Vehicles to Tumors Using Polyion Complexes and Gold
Nanoparticles. ACS Nano 2014,8, 89798991.
(17) Harimech, P.; Gerrard, S.; El-Sagheer, A.; Brown, T.; Kanaras, A.
Reversible Ligation of Programmed DNA-Gold Nanoparticle
Assemblies. J. Am. Chem. Soc. 2015,137, 92429245.
(18) Chapman, P.; Hauschild, A.; Robert, C.; Haanen, J.; Ascierto, P.;
Larkin, J.; Dummer, R.; Garbe, C.; Testori, A.; Maio, M.; Hogg, D.;
Lorigan, P.; Lebbe, C.; Jouary, T.; Schadendorf, D.; Ribas, A.; ODay,
S.; Sosman, J.; Kirkwood, J.; Eggermont, A.; Dreno, B.; Nolop, K.; Li,
J.; Nelson, B.; Hou, J.; Lee, R.; Flaherty, K.; McArthur, G. Improved
Survival with Vemurafenib in Melanoma with Braf V600e Mutation. N.
Engl. J. Med. 2011,364, 25072516.
(19) Hauschild, A.; Grob, J.; Demidov, L.; Jouary, T.; Gutzmer, R.;
Millward, M.; Rutkowski, P.; Blank, C.; Miller, W.; Kaempgen, E.;
Martin-Algarra, S.; Karaszewska, B.; Mauch, C.; Chiarion-Sileni, V.;
Martin, A.; Swann, S.; Haney, P.; Mirakhur, B.; Guckert, M.;
Goodman, V.; Chapman, P. Dabrafenib in Braf-Mutated Metastatic
Melanoma: A Multicentre, Open-Label, Phase 3 Randomised
Controlled Trial. Lancet 2012,380, 358365.
(20) Karabulut, Y.; Erdogan, S.; Sayar, H.; Ergen, A.; Ertoy Baydar,
D. Primary Malignant Melanoma of the Urinary Bladder: Clinical,
Morphological, and Molecular Analysis of Five Cases. Melanoma Res.
2016,26, 616624.
(21) Bhatia, S.; Margolin, K. Disparate Clinical Activity of Pd-1
Blockade in Melanoma Subtypes: Know Thy Enemy! Cancer 2016,
122, 32633266.
(22) Richard, G.; Dalle, S.; Monet, M.; Ligier, M.; Boespflug, A.;
Pommier, R.; de la Fouchardiere, A.; Perier-Muzet, M.; Depaepe, L.;
Barnault, R.; Tondeur, G.; Ansieau, S.; Thomas, E.; Bertolotto, C.;
Ballotti, R.; Mourah, S.; Battistella, M.; Lebbe, C.; Thomas, L.;
Puisieux, A.; Caramel, J. Zeb1-Mediated Melanoma Cell Plasticity
Enhances Resistance to Mapk Inhibitors. EMBO Mol. Med. 2016,8,
11431161.
(23) Felicetti, F.; Errico, M.; Bottero, L.; Segnalini, P.; Stoppacciaro,
A.; Biffoni, M.; Felli, N.; Mattia, G.; Petrini, M.; Colombo, M.; Peschle,
C.; Care, A. The Promyelocytic Leukemia Zinc Finger-Microrna-221/-
222 Pathway Controls Melanoma Progression through Multiple
Oncogenic Mechanisms. Cancer Res. 2008,68, 27452754.
(24) Hussein, M.; Haemel, A.; Wood, G. Apoptosis and Melanoma:
Molecular Mechanisms. J. Pathol. 2003,199, 275288.
(25) Li, W.; Sanki, A.; Karim, R.; Thompson, J.; Soon Lee, C.;
Zhuang, L.; McCarthy, S.; Scolyer, R. The Role of Cell Cycle
Regulatory Proteins in the Pathogenesis of Melanoma. Pathology 2006,
38, 287301.
(26) Aagaard, L.; Rossi, J. Therapeutics: Principles, Prospects and
Challenges. Adv. Drug Delivery Rev. 2007,59,7586.
(27) Gillies, J.; Lorimer, I. Regulation of P27kip1 by Mirna 221/222
in Glioblastoma. Cell Cycle 2007,6, 20052009.
(28) Ihle, M.; Trautmann, M.; Kuenstlinger, H.; Huss, S.; Heydt, C.;
Fassunke, J.; Wardelmann, E.; Bauer, S.; Schildhaus, H.; Buettner, R.;
Merkelbach-Bruse, S. Mirna-221 and Mirna-222 Induce Apoptosis Via
the Kit/Akt Signalling Pathway in Gastrointestinal Stromal Tumours.
Mol. Oncol. 2015,9, 14211433.
(29) Carvajal, R.; Antonescu, C.; Wolchok, J.; Chapman, P.; Roman,
R.; Teitcher, J.; Panageas, K.; Busam, K.; Chmielowski, B.; Lutzky, J.;
Pavlick, A.; Fusco, A.; Cane, L.; Takebe, N.; Vemula, S.; Bouvier, N.;
Bastian, B.; Schwartz, G. Kit as a Therapeutic Target in Metastatic
Melanoma. JAMA, J. Am. Med. Assoc. 2011,305, 23272334.
(30) Sigalotti, L.; Covre, A.; Fratta, E.; Parisi, G.; Colizzi, F.; Rizzo,
A.; Danielli, R.; Nicolay, H.; Coral, S.; Maio, M. Epigenetics of Human
Cutaneous Melanoma: Setting the Stage for New Therapeutic
Strategies. J. Transl. Med. 2010,8, 56.
ACS Applied Materials & Interfaces Research Article
DOI: 10.1021/acsami.6b16378
ACS Appl. Mater. Interfaces XXXX, XXX, XXXXXX
L
(31) Tsui, B.; Singh, V.; Liang, J.; Yang, V. Reduced Reactivity
Towards Anti-Protamine Antibodies of a Low Molecular Weight
Protamine Analogue. Thromb. Res. 2001,101, 417420.
(32) Suhorutsenko, J.; Oskolkov, N.; Arukuusk, P.; Kurrikoff, K.;
Eriste, E.; Copolovici, D.; Langel, U. Cell-Penetrating Peptides,
Pepfects, Show No Evidence of Toxicity and Immunogenicity.
Bioconjugate Chem. 2011,22, 22552262.
(33) Wimley, W. Describing the Mechanism of Antimicrobial Peptide
Action with the Interfacial Activity Model. ACS Chem. Biol. 2010,5,
905917.
(34) He, J.; Hristova, K.; Wimley, W. A Highly Charged Voltage-
Sensor Helix Spontaneously Translocates across Membranes. Angew.
Chem., Int. Ed. 2012,51, 71507153.
(35) Palm-Apergi, C.; Lorents, A.; Padari, K.; Pooga, M.; Hallbrink,
M. The Membrane Repair Response Masks Membrane Disturbances
Caused by Cell-Penetrating Peptide Uptake. FASEB J. 2009,23, 214
223.
(36) Huang, Y.; Park, Y.; Moon, C.; David, A.; Chung, H.; Yang, V.
Synthetic Skin-Permeable Proteins Enabling Needleless Immunization.
Angew. Chem., Int. Ed. 2010,49, 27242727.
(37) Vives, E.; Brodin, P.; Lebleu, B. A Truncated Hiv-1 Tat Protein
Basic Domain Rapidly Translocates through the Plasma Membrane
and Accumulates in the Cell Nucleus. J. Biol. Chem. 1997,272, 16010
16017.
(38) He, J.; Kauffman, W.; Fuselier, T.; Naveen, S.; Voss, T.;
Hristova, K.; Wimley, W. Direct Cytosolic Delivery of Polar Cargo to
Cells by Spontaneous Membrane-Translocating Peptides. J. Biol.
Chem. 2013,288, 2997429986.
(39) LaRochelle, J.; Cobb, G.; Steinauer, A.; Rhoades, E.; Schepartz,
A. Fluorescence Correlation Spectroscopy Reveals Highly Efficient
Cytosolic Delivery of Certain Penta-Arg Proteins and Stapled
Peptides. J. Am. Chem. Soc. 2015,137, 25362541.
(40) Lin, J.; Alexander-Katz, A. Cell Membranes Open Doorsfor
Cationic Nanoparticles/Biomolecules: Insights into Uptake Kinetics.
ACS Nano 2013,7, 1079910808.
(41) Marks, J.; Placone, J.; Hristova, K.; Wimley, W. Spontaneous
Membrane-Translocating Peptides by Orthogonal High-Throughput
Screening. J. Am. Chem. Soc. 2011,133, 89959004.
(42) Peng, L.; Niu, J.; Zhang, C.; Yu, W.; Wu, J.; Shan, Y.; Wang, X.;
Shen, Y.; Mao, Z.; Liang, W.; Gao, J. Tat Conjugated Cationic Noble
Metal Nanoparticles for Gene Delivery to Epidermal Stem Cells.
Biomaterials 2014,35, 56055618.
(43) Peng, L.; Huang, Y.; Zhang, C.; Niu, J.; Chen, Y.; Chu, Y.; Jiang,
Z.; Gao, J.; Mao, Z. Integration of Antimicrobial Peptides with Gold
Nanoparticles as Unique Non-Viral Vectors for Gene Delivery to
Mesenchymal Stem Cells with Antibacterial Activity. Biomaterials
2016,103, 137149.
(44) Lou, G.; Zhang, Q.; Xiao, F.; Xiang, Q.; Su, Z.; Zhang, L.; Yang,
P.; Yang, Y.; Zheng, Q.; Huang, Y. Intranasal Administration of Tat-
Hafgf Attenuates Disease Progression in a Mouse Model of
Alzheimers Disease. Neuroscience 2012,223, 225237.
(45) Love, J.; Estroff, L.; Kriebel, J.; Nuzzo, R.; Whitesides, G. Self-
Assembled Monolayers of Thiolates on Metals as a Form of
Nanotechnology. Chem. Rev. 2005,105, 11031169.
(46) Xia, X.; Yang, M.; Wang, Y.; Zheng, Y.; Li, Q.; Chen, J.; Xia, Y.
Quantifying the Coverage Density of Poly(Ethylene Glycol) Chains
on the Surface of Gold Nanostructures. ACS Nano 2012,6, 512522.
(47) Duchardt, F.; Fotin-Mleczek, M.; Schwarz, H.; Fischer, R.;
Brock, R. A Comprehensive Model for the Cellular Uptake of Cationic
Cell-Penetrating Peptides. Traffic 2007,8, 848866.
(48) Han, S.; Gai, W.; Yancovitz, M.; Osman, I.; Di Como, C.;
Polsky, D. Nucleofection Is a Highly Effective Gene Transfer
Technique for Human Melanoma Cell Lines. Exp. Dermatol. 2008,
17, 405411.
(49) Weiss, J.; Shivakumar, R.; Feller, S.; Li, L.; Hanson, A.; Fogler,
W.; Fratantoni, J.; Liu, L. Rapid, in Vivo, Evaluation of Antiangiogenic
and Antineoplastic Gene Products by Nonviral Transfection of Tumor
Cells. Cancer Gene Ther. 2004,11, 346353.
(50) Neu, M.; Fischer, D.; Kissel, T. Recent Advances in Rational
Gene Transfer Vector Design Based on Poly(Ethylene Imine) and Its
Derivatives. J. Gene Med. 2005,7, 9921009.
(51) Iversen, T.-G.; Skotland, T.; Sandvig, K. Endocytosis and
Intracellular Transport of Nanoparticles: Present Knowledge and
Need for Future Studies. Nano Today 2011,6, 176185.
(52) Rodal, S.; Skretting, G.; Garred, O.; Vilhardt, F.; van Deurs, B.;
Sandvig, K. Extraction of Cholesterol with Methyl-Beta-Cyclodextrin
Perturbs Formation of Clathrin-Coated Endocytic Vesicles. Mol. Biol.
Cell 1999,10, 961974.
(53) Wang, L.; Rothberg, K.; Anderson, R. Mis-Assembly of Clathrin
Lattices on Endosomes Reveals a Regulatory Switch for Coated Pit
Formation. J. Cell Biol. 1993,123, 11071117.
(54) Koivusalo, M.; Welch, C.; Hayashi, H.; Scott, C.; Kim, M.;
Alexander, T.; Touret, N.; Hahn, K.; Grinstein, S. Amiloride Inhibits
Macropinocytosis by Lowering Submembranous Ph and Preventing
Rac1 and Cdc42 Signaling. J. Cell Biol. 2010,188, 547563.
(55) Rana, S.; Bajaj, A.; Mout, R.; Rotello, V. Monolayer Coated
Gold Nanoparticles for Delivery Applications. Adv. Drug Delivery Rev.
2012,64, 200216.
(56) Thomas, M.; Klibanov, A. Conjugation to Gold Nanoparticles
Enhances Polyethylenimines Transfer of Plasmid DNA into
Mammalian Cells. Proc. Natl. Acad. Sci. U. S. A. 2003,100, 91389143.
(57) Koelz, M.; Lense, J.; Wrba, F.; Scheffler, M.; Dienes, H.;
Odenthal, M. Down-Regulation of Mir-221 and Mir-222 Correlates
with Pronounced Kit Expression in Gastrointestinal Stromal Tumors.
Int. J. Oncol. 2011,38, 503511.
(58) Wu, C.; Cheng, Y.; Hsu, N.; Yeh, K.; Tsai, Y.; Chiang, C.; Wang,
W.; Tung, J. Mirna-221 Negatively Regulated Downstream P27kip1
Gene Expression Involvement in Pterygium Pathogenesis. Mol. Vision
2014,20, 10481056.
(59) Overwijk, W.; Restifo, N. B16 as a Mouse Model for Human
Melanoma. Current Protocals in Immunology 2001,1DOI: 10.1002/
0471142735.im2001s39.
(60) Gaudi, S.; Messina, J. Molecular Bases of Cutaneous and Uveal
Melanomas. Pathology Research International 2011,2011, 159421.
(61) Zhang, L.; Chen, Z.; Li, Y. Dual-Degradable Disulfide-
Containing Pei-Pluronic/DNA Polyplexes: Transfection Efficiency
and Balancing Protection and DNA Release. Int. J. Nanomed. 2013,8,
36893701.
(62) Intra, J.; Salem, A. Rational Design, Fabrication, Character-
ization and in Vitro Testing of Biodegradable Microparticles That
Generate Targeted and Sustained Transgene Expression in Hepg2
Liver Cells. J. Drug Targeting 2011,19, 393408.
(63) Chen, Z.; Zhang, L.; He, Y.; Li, Y. Sandwich-Type Au-Pei/
DNA/Pei-Dexa Nanocomplex for Nucleus-Targeted Gene Delivery in
Vitro and in Vivo. ACS Appl. Mater. Interfaces 2014,6, 1419614206.
(64) Lademann, J.; Otberg, N.; Richter, H.; Weigmann, H.;
Lindemann, U.; Schaefer, H.; Sterry, W. Investigation of Follicular
Penetration of Topically Applied Substances. Skin Pharmacol. Appl.
Skin Physiol. 2001,14,1722.
(65) Lademann, J.; Richter, H.; Schaefer, U.; Blume-Peytavi, U.;
Teichmann, A.; Otberg, N.; Sterry, W. Hair Follicles - a Long-Term
Reservoir for Drug Delivery. Skin Pharmacol. Physiol. 2006,19, 232
236.
(66) Boakye, C.; Patel, K.; Singh, M. Doxorubicin Liposomes as an
Investigative Model to Study the Skin Permeation of Nanocarriers. Int.
J. Pharm. 2015,489, 106116.
(67) Wang, B.; Zhang, L.; Bae, S.; Granick, S. Nanoparticle-Induced
Surface Reconstruction of Phospholipid Membranes. Proc. Natl. Acad.
Sci. U. S. A. 2008,105, 1817118175.
(68) Roiter, Y.; Ornatska, M.; Rammohan, A.; Balakrishnan, J.;
Heine, D.; Minko, S. Interaction of Nanoparticles with Lipid
Membrane. Nano Lett. 2008,8, 941944.
(69) Dawson, K.; Salvati, A.; Lynch, I. Nanotoxicology: Nanoparticles
Reconstruct Lipids. Nat. Nanotechnol. 2009,4,8485.
(70) Hartono, D.; Qin, W.; Yang, K.; Yung, L. Imaging the
Disruption of Phospholipid Monolayer by Protein-Coated Nano-
ACS Applied Materials & Interfaces Research Article
DOI: 10.1021/acsami.6b16378
ACS Appl. Mater. Interfaces XXXX, XXX, XXXXXX
M
particles Using Ordering Transitions of Liquid Crystals. Biomaterials
2009,30, 843849.
(71) Plasencia, I.; Norlen, L.; Bagatolli, L. Direct Visualization of
Lipid Domains in Human Skin Stratum Corneums Lipid Membranes:
Effect of Ph and Temperature. Biophys. J. 2007,93, 31423155.
(72) Ryman-Rasmussen, J.; Riviere, J.; Monteiro-Riviere, N.
Penetration of Intact Skin by Quantum Dots with Diverse
Physicochemical Properties. Toxicol. Sci. 2006,91, 159165.
(73) Baroli, B.; Ennas, M.; Loffredo, F.; Isola, M.; Pinna, R.; Arturo
Lopez-Quintela, M. Penetration of Metallic Nanoparticles in Human
Full-Thickness Skin. J. Invest. Dermatol. 2007,127, 17011712.
(74) Moritake, S.; Taira, S.; Ichiyanagi, Y.; Morone, N.; Song, S.;
Hatanaka, T.; Yuasa, S.; Setou, M. Functionalized Nano-Magnetic
Particles for an in Vivo Delivery System. J. Nanosci. Nanotechnol. 2007,
7, 937944.
(75) Peng, L.; Zhang, Y.; Han, L.; Zhang, C.; Wu, J.; Wang, X.; Gao,
J.; Mao, Z. Cell Membrane Capsules for Encapsulation of Chemo-
therapeutic and Cancer Cell Targeting in Vivo. ACS Appl. Mater.
Interfaces 2015,7, 1862818637.
(76) Wong, J.; Harris, J.; Rodriguez-Galindo, C.; Johnson, K.
Incidence of Childhood and Adolescent Melanoma in the United
States: 19732009. Pediatrics 2013,131, 846854.
(77) Yan, J.; Tingey, C.; Lyde, R.; Gorham, T.; Choo, D.;
Muthumani, A.; Myles, D.; Weiner, L.; Kraynyak, K.; Reuschel, E.;
Finkel, T.; Kim, J.; Sardesai, N.; Ugen, K.; Muthumani, K.; Weiner, D.
Novel and Enhanced Anti-Melanoma DNA Vaccine Targeting the
Tyrosinase Protein Inhibits Myeloid-Derived Suppressor Cells and
Tumor Growth in a Syngeneic Prophylactic and Therapeutic Murine
Model. Cancer Gene Ther. 2014,21, 507517.
(78) Tiwary, S.; Preziosi, M.; Rothberg, P.; Zeitouni, N.; Corson, N.;
Xu, L. Erbb3 Is Required for Metastasis Formation of Melanoma Cells.
Oncogenesis 2014,3, e110.
(79) Luo, C.; Tetteh, P.; Merz, P.; Dickes, E.; Abukiwan, A.; Hotz-
Wagenblatt, A.; Holland-Cunz, S.; Sinnberg, T.; Schittek, B.;
Schadendorf, D.; Diederichs, S.; Eichmuller, S. Mir-137 Inhibits the
Invasion of Melanoma Cells through Downregulation of Multiple
Oncogenic Target Genes. J. Invest. Dermatol. 2013,133, 768775.
(80) Liu, J.; Wu, X.; Liu, H.; Liang, Y.; Gao, X.; Cai, Z.; Wang, W.;
Zhang, H. Expression of Microrna-30a-5p in Drug-Resistant and Drug-
Sensitive Ovarian Cancer Cell Lines. Oncol. Lett. 2016,12, 2065
2070.
(81) Song, K.; Han, C.; Zhang, J.; Lu, D.; Dash, S.; Feitelson, M.;
Lim, K.; Wu, T. Epigenetic Regulation of Microrna-122 by Peroxisome
Proliferator Activated Receptor-Gamma and Hepatitis B Virus X
Protein in Hepatocellular Carcinoma Cells. Hepatology 2013,58,
16811692.
(82) Siu, K.; Chen, D.; Zheng, X.; Zhang, X.; Johnston, N.; Liu, Y.;
Yuan, K.; Koropatnick, J.; Gillies, E.; Min, W. Non-Covalently
Functionalized Single-Walled Carbon Nanotube for Topical Sirna
Delivery into Melanoma. Biomaterials 2014,35, 34353442.
(83) Desai, P.; Marepally, S.; Patel, A.; Voshavar, C.; Chaudhuri, A.;
Singh, M. Topical Delivery of Anti-Tnfalpha Sirna and Capsaicin Via
Novel Lipid-Polymer Hybrid Nanoparticles Efficiently Inhibits Skin
Inflammation in Vivo.J. Controlled Release 2013,170,5163.
ACS Applied Materials & Interfaces Research Article
DOI: 10.1021/acsami.6b16378
ACS Appl. Mater. Interfaces XXXX, XXX, XXXXXX
N
... Moreover, GNP conjugated with quercetin have inhibited epithelialmesenchymal transition, angiogenesis, and invasiveness of MCF-7 and MDA-MB-231 breast cancer cell lines [70]. Modulation of several mediators, pathways, and genes such as MMP-9, NF-κB, AP-1, PI3K/Akt, genes Mi221, and HIC1 has contributed to the cellular migration retardation and anti-invasion effect of GNP conjugated with various ligands [71][72][73]. In vitro adhesion assay of prostate and breast cancer cell lines following treatment with GNP-BSA Adhesion assay was employed to investigate the effect of GNR-BSA and GNS-BSA on the adhesion ability of the three cell lines. ...
Article
Full-text available
Breast and prostate cancers are prevalent in women and men, respectively. The process of metastasis plays a crucial role in cancer advancement. Herein, two distinct forms of gold nanoparticles (GNP) were prepared and modified with bovine serum albumin (BSA) to create gold nanorods-BSA (GNR-BSA) and gold nanospheres-BSA (GNS-BSA). Various aspects of biological interactions of these nanoparticles with two prostate cancer cell lines (DU-145 and PC-3) and a breast cancer cell line (MDA-MB-231) have been investigated. The cell viability of DU-145 and PC-3 ranged from 17 to 95% across concentrations of 0.55 to 34.5 µg/mL and for MDA-MB-231 ranged from 17 to 85%. GNS-BSA exhibited no significant cytotoxicity against the cancer cell lines. Regarding cellular uptake, GNR-BSA demonstrated uptake rates of 10%, 14%, and 5% for DU-145, PC-3, and MDA-MB-231 cell lines, respectively, while GNS-BSA showed uptake of less than 0.4% for all the cell lines investigated. Notably, GNR-BSA significantly impeded the cellular migration of DU-145 and PC-3 cells over 48 h (hr) and MDA-MB-231 cells over 24 h compared to controls. GNS-BSA inhibited cell migration over 48 h (hr) for DU-145 and over 24 h for PC-3 and MDA-MB-231. Adhesion assay showed a moderate reduction of PC-3 adhesion ability (∼\documentclass[12pt]{minimal} \usepackage{amsmath} \usepackage{wasysym} \usepackage{amsfonts} \usepackage{amssymb} \usepackage{amsbsy} \usepackage{mathrsfs} \usepackage{upgreek} \setlength{\oddsidemargin}{-69pt} \begin{document}$$\sim$$\end{document} 20%) by GNS-BSA, while a minimum effect was observed on DU-145 (∼\documentclass[12pt]{minimal} \usepackage{amsmath} \usepackage{wasysym} \usepackage{amsfonts} \usepackage{amssymb} \usepackage{amsbsy} \usepackage{mathrsfs} \usepackage{upgreek} \setlength{\oddsidemargin}{-69pt} \begin{document}$$\sim$$\end{document} 5%). GNR-BSA has minimally affected the adhesion ability of both PC-3 (∼\documentclass[12pt]{minimal} \usepackage{amsmath} \usepackage{wasysym} \usepackage{amsfonts} \usepackage{amssymb} \usepackage{amsbsy} \usepackage{mathrsfs} \usepackage{upgreek} \setlength{\oddsidemargin}{-69pt} \begin{document}$$\sim$$\end{document} 8%) and DU-145 (∼\documentclass[12pt]{minimal} \usepackage{amsmath} \usepackage{wasysym} \usepackage{amsfonts} \usepackage{amssymb} \usepackage{amsbsy} \usepackage{mathrsfs} \usepackage{upgreek} \setlength{\oddsidemargin}{-69pt} \begin{document}$$\sim$$\end{document} 13%), and no adhesion ability reduction was observed on MDA-MB-231 by both GNR-BSA or GNS-BSA. This study suggests that GNP-BSA could be promising potential agents for combating cancer and inhibiting cellular invasion, and they could serve as promising platforms for drug delivery.
... DMSO being as polar aprotic solvent helps in reducing the barrier of stratum corneum of skin and potentiates NPs to traverse easily that fights skin cancer (Menezes et al., 2016). Cationic gold NPs incorporated with plasmid DNA and conjugated with HIV-1 twinarginine translocation peptides (TAT) potentially targets the cutaneous melanoma (Niu et al., 2017). ...
Chapter
In current scenario, skin cancer is one of the most prevalent forms of cancer, and mortality and morbidity rates are still continuously rising in many other countries. Skin cancer is classified into two types: melanoma skin cancer and nonmelanoma skin cancer. Further, nonmelanoma skin cancer is subdivided into basal cell carcinoma and squamous cell carcinoma. Occurrence of the skin cancer is due to many reasons, and UV radiation is one of the major contributors. So majority of approaches are now a days designed to reduce exposure of UV radiations. Excision is still a gold standard treatment for localized skin cancer. But in very rare cases, cancerous cells spread to regional lymph nodes (LNs) as well as to distant sites (Lalan et al., 2021; Kaur and Kesarwani, 2021; Borgheti-Cardoso et al., 2020). For the treatment of metastasized skin cancers, nanocarriers are one of the effective drug delivery systems, which allow anticancer drugs to reach exactly to the cancer site and, thus, enhance therapeutic efficacy. It is a revolutionary approach that includes designing, preparation, evaluation, and application by controlling shape and size at the nanoscale. These biomimetic features of nanocarriers and the possibility of modifying their properties raised the interest in its theranostic applications. Targeted nanomedicine enhances permeability in cancerous cells, thus allowing the delivery of drugs to cancer cells specifically. Moreover, specific binding of nanocarriers to cancer cells improves the efficiency of the treatment of cancerous cells, without effecting healthy cells (Singh and Lillard, 2009; Sanvicens and Marco, 2008; Dianzani et al., 2014).
... Additionally, the AuPT is very effective at promoting the intracellular absorption and nuclear targeting of the pDNAs in cells, ensuring successful transfection. This study shows that penetrating peptide-attached cationic gold nanoparticles are an effective delivery system for pDNA transfection and skin penetration, showing tremendous promise for topical gene therapy [63]. ...
Article
Full-text available
Carcinoma is a condition that continues to pose a significant challenge, despite current medical advances. Skin carcinoma is the leading cause of cancer, and it has seen a massive increase all over the world. The challenges with current treatment are due to toxicity that leads to many more skin complications. Due to this to avoid such complications by designing diverse nanoparticles as delivery carriers, nanomedicine is employed as a hub for diagnostics and therapy. Liposomes, gold nanoparticles, transferases, nanofibers, etc., can all be used as delivery nanocarriers. These nanoparticles’ structures and characteristics protect the medicine from degradation and improve its stability. Surface modifying agents and procedures are employed to functionalize nanoparticles, resulting in smart delivery systems. The application of nanotechnology-based approaches systematically increases drug delivery to target cells. Skin cancer has several challenges, including a long time to diagnose early types of cancer and a slower growth rate. This review focuses on innovative skin cancer therapy techniques, focusing on nanotechnology and the challenges associated with current treatment of skin cancer. Graphical abstract
Article
Because of their wide variety of biological effects, bioactive peptides (BAPs) have recently attracted a lot of attention. BAPs have been observed to be safe, thanks to their widely acknowledged safety status by the United States Food and Drug Administration (USFDA). This has led to their widespread use in various industries, such as food and nutrition, pharmaceuticals, and therapeutics. A considerable amount of research has been devoted to developing cutting‐edge nanomaterials derived from BAPs, which have been utilized in a wide range of industries. In the realm of scientific research, the remarkable ability of BAPs to self‐assemble has been harnessed to develop a wide range of nanoassemblies. These nanoassemblies hold immense potential for the advancement of biomaterials in the future. Research interest continues to focus on the study of the detection of BAPs using artificial intelligence (AI). Over the past few years, there has been a surge of interest in utilizing bio‐inspired strategies to explore new possibilities in the development of advanced energy devices and storage solutions. However, these strategies require scientific attention. This extensive review offers a broad perspective on the applications of BAPs in nanotechnology as well as pharmaceuticals and therapeutics. Moreover, in silico analysis of BAPs, coupled with ‐omics techniques, has been discussed. In the bargain, the next‐generation approaches in BAP research comprising BAP‐based energy devices, as well as BAPs in AI, have been catalogued. There is a considerable emphasis on developing more eco‐friendly energy‐storage technologies that draw inspiration from nature and use BAPs.
Article
Cancer is among the leading causes of mortality and morbidity in the world. Metallic nanoparticles, especially gold nanoparticles (AuNPs) have emerged to be attractive systems to circumvent the associated adverse effects. By the virtue of their unique properties of tunable size, shape, composition, optical properties, biocompatibility, minimal toxicity, multivalency, fluorescence-luminescence property and surface plasmon resonance; AuNPs have the potential to be used as drug delivery systems. It is vital to ensure that the drug reaches the target site of action for selective kill of cancer cells without harm to healthy cells. These AuNPs can be easily functionalized with a wide array of ligands like peptides, oligonucleotides, polymers, carbohydrates for active targeting to ensure site specific delivery and reduced systemic effects. AuNPs have been in-vestigated as carriers for gene delivery, drug delivery with or without photothermal therapy, in diagnosis based on radiation or spectroscopy. They have emerged as attractive theranostic approach in the overall management of cancer with superior benefit to risk features. In this review, we have discussed synthesis of different AuNPs (nanorods, spherical nanoparticles, and hollow AuNPs), their functionalization strategies and their applications in biomedical domain. Various research studies and clinical trials on application of AuNPs in diagnosis and therapeutics are highlighted.
Preprint
Full-text available
Breast and prostate cancers are prevalent in women and men, respectively. The process of metastasis plays a crucial role in cancer advancement. Herein, two distinct forms of gold nanoparticles (GNP) were prepared and modified with bovine serum albumin (BSA) to create gold nanorods-BSA (GNR-BSA) and gold nanospheres-BSA (GNS-BSA). Various aspects of biological interactions of these nanoparticles with two prostate cancer cell lines (DU-145 and PC-3) and a breast cancer cell line (MDA-MB-231) have been investigated. The cell viability of DU-145 and PC-3 ranged from 17% to 95% across concentrations of 0.55 to 34.5 µg/mL, and for MDA-MB-231 ranged from 17% to 85%. GNS-BSA exhibited no significant cytotoxicity against the cancer cell lines. Regarding cellular uptake, GNR-BSA demonstrated uptake rates of 10%, 14%, and 5% for DU-145, PC-3, and MDA-MB-231 cell lines, respectively, while GNS-BSA showed uptake of less than 0.4% for all the cell lines investigated. Notably, GNR-BSA significantly impeded the cellular migration of DU-145 and PC-3 cells over 48 hr and MDA-MB-231 cells over 24 hr compared to controls. GNS-BSA inhibited cell migration over 48 hours for DU-145 and over 24 hours for PC-3 and MDA-MB-231. Adhesion assay showed a moderate reduction of PC-3 adhesion ability (~ 20%) by GNS-BSA, while a minimum effect was observed on DU-145 (~5%). GNR-BSA have minimally affected the adhesion ability of both PC-3 (~8%), and DU-145 (~13%) and no adhesion ability reduction was observed on MDA-MB-231 by both GNP-BSA. This study suggests that GNP-BSA could be promising potential agents for combating cancer and inhibiting cellular invasion, and they could serve as promising platforms for drug delivery.
Article
Full-text available
Targeted therapies with MAPK inhibitors (MAPKi) are faced with severe problems of resistance in BRAF-mutant melanoma. In parallel to the acquisition of genetic mutations, melanoma cells may also adapt to the drugs through phenotype switching. The ZEB1 transcription factor, a known inducer of EMT and invasiveness, is now considered as a genuine oncogenic factor required for tumor initiation, cancer cell plasticity, and drug resistance in carcinomas. Here, we show that high levels of ZEB1 expression are associated with inherent resistance to MAPKi in BRAF(V) (600)-mutated cell lines and tumors. ZEB1 levels are also elevated in melanoma cells with acquired resistance and in biopsies from patients relapsing while under treatment. ZEB1 overexpression is sufficient to drive the emergence of resistance to MAPKi by promoting a reversible transition toward a MITF(low)/p75(high) stem-like and tumorigenic phenotype. ZEB1 inhibition promotes cell differentiation, prevents tumorigenic growth in vivo, sensitizes naive melanoma cells to MAPKi, and induces cell death in resistant cells. Overall, our results demonstrate that ZEB1 is a major driver of melanoma cell plasticity, driving drug adaptation and phenotypic resistance to MAPKi.
Article
Full-text available
The present study aimed to explore the expression of microRNA (miRNA or miR) in drug-resistant and drug-sensitive ovarian cancer cell lines, and to seek the potential therapeutic target of ovarian cancer drug-resistant mechanism in order to improve drug resistance by altering miRNA levels. The drug-resistant characteristics of SKOV3/DDP, SKOV3, COC1/DDP and COC1 cell lines were studied. The miRNAs that were differentially expressed between cisplatin-resistant cells and its parental cells in ovarian cancer were screened with a miRNA chip. The effect of miRNAs was detected, and their drug-resistant mechanism was investigated by transfection and 3-(4,5-dimethylthiazol-2-yl) -2,5-diphenyltetrazolium bromide methods. Among the expression screening of miRNAs, 41 mRNAs, including Homo sapiens (hsa)-miR-30a-5p and hsa-miR-34c-5p, were highly expressed in the drug-resistant cells, whereas 44 miRNAs, including hsa-miR-96-5p and hsa-miR-200c-3p, were lowly expressed. The expression levels of hsa-miR-30a-5p in two types of ovarian cancer chemotherapy-resistant cell lines were significantly higher than those in chemotherapy-sensitive cell lines, which was associated with ovarian cancer chemotherapy resistance. In conclusion, high expression of miRNA-30a-5p was able to promote cell growth and colony forming ability, and enhance cell migration and invasion. Thus, miRNA-30a-5p is expected to become a meaningful novel target for ovarian cancer resistant treatment.
Article
Full-text available
Figure 2. (B) Top: schematic of the structure of membrane-targeted SEpHluorin/mCherry chimaera used to measure pH sm. Bottom: confocal images of SEpHluorin (left) and mCherry fluorescence (right) in A431 cells. Bar, 10 µm.
Article
Full-text available
Because of the aggressive and recurrent nature of cancers, repeated and multimodal treatments are often necessary. Traditional cancer therapies have a risk of serious toxicity and side effects. Hence, it is crucial to develop an alternative treatment modality that is minimally invasive, effectively treats cancers with low toxicity, and can be repeated as required. We developed a light-activatable microneedle (MN) system that can repeatedly and simultaneously provide photothermal therapy and chemotherapy to superficial tumors and exert synergistic anticancer effects. This system consists of embeddable polycaprolactone MNs containing a photosensitive nanomaterial (lanthanum hexaboride) and an anticancer drug (doxorubicin; DOX), and a dissolvable polyvinyl alcohol/polyvinyl pyrrolidone supporting array patch. Because of this supporting array, the MNs can be completely inserted into the skin and embedded within the target tissue for locoregional cancer treatment. When exposed to near-infrared light, the embedded MN array uniformly heats the target tissue to induce a large thermal ablation area and then melts at 50 °C to release DOX in a broad area, thus destroying tumors. This light-activated heating and releasing behavior can be precisely controlled and switched on and off on demand for several cycles. We demonstrated that the MN-mediated synergistic therapy completely eradicated 4T1 tumors within 1 week after a single application of the MN and three cycles of laser treatment. No tumor recurrence and no significant body weight loss of mice were observed. Thus, the developed light-activatable MN with a unique embeddable feature offers an effective, user-friendly, and low-toxicity option for patients requiring long-term and multiple cancer treatments.
Article
Full-text available
We demonstrate a new method to reversibly cross-link DNA-nanoparticle dimers, trimers, and tetramers using light as an external stimulus. A DNA interstrand photo-cross-linking reaction is possible via ligation of a cyano-vinyl carbazole nucleoside with an opposite thymine when irradiated at 365 nm. This reaction results in nanoparticle assemblies that are not susceptible to DNA dehybridization conditions. The chemical bond between the two complementary DNA strands can be reversibly broken upon light irradiation at 312 nm. This is the first example of reversible ligation in DNA-nanoparticle assemblies using light and enables new developments in the field of programmed nanoparticle organization.
Article
The aim of our study was to evaluate the clinical and morphological features of primary malignant melanomas of the urinary bladder. We obtained information on five such cases from three different institutions. These were three men and two women between 52 and 76 years of age. Three tumors presented with hematuria, one with dysuria, and one was discovered incidentally on imaging studies. All were invasive to muscularis propria on transuretral resections performed for diagnosis. Neoplastic cells showed variable patterns (large cell epithelioid, small cell diffuse, storiform, or mixed) in different tumors. Pigmentation was prominent in all except one case. Each case was labeled diffusely for S-100, HMB-45, and Melan-A. Pan-cytokeratin showed a perinuclear dot-like reaction in two tumors. Three cases showed the BRAF mutation in molecular studies. Two patients were already metastatic at the time of diagnosis. Two patients died, one is alive with disease after 15 months, and two patients are disease free at 1 and 5 years of surveillance.
Article
The 2 articles by Shousatri et al and Algazi et al in this issue of Cancer report the efficacy of programmed cell death receptor 1 (PD‐1) pathway blockade in metastatic acral/mucosal melanoma and uveal melanoma, respectively. The results point to the disparate activity of PD‐1 and PD‐1 ligand pathway blockade in these subtypes of melanoma and highlight the need to understand differences in the underlying biology of immune evasion in these and other malignancies as well as the need to identify mechanisms of success and failure of PD‐1 blockade. See also pages 3344‐53 and 3354‐62.
Article
Conventional cancer therapies involve the systemic delivery of anticancer agents that neither discriminate between cancer and normal cells nor eliminate the risk of cancer recurrence. Here, we demonstrate that the combination of gene, drug and phototherapy delivered through a prophylactic hydrogel patch leads, in a colon cancer mouse model, to complete tumour remission when applied to non-resected tumours and to the absence of tumour recurrence when applied following tumour resection. The adhesive hydrogel patch enhanced the stability and provided local delivery of embedded nanoparticles. Spherical gold nanoparticles were used as a first wave of treatment to deliver siRNAs against Kras, a key oncogene driver, and rod-shaped gold nanoparticles mediated the conversion of near-infrared radiation into heat, causing the release of a chemotherapeutic as well as thermally induced cell damage. This local, triple-combination therapy can be adapted to other cancer cell types and to molecular targets associated with disease progression.