Sei sulla pagina 1di 10

Biomaterials 33 (2012) 3604e3613

Contents lists available at SciVerse ScienceDirect

Biomaterials
journal homepage: www.elsevier.com/locate/biomaterials

Nano-carrier for gene delivery and bioimaging based on carbon dots with
PEI-passivation enhanced fluorescence
Changjun Liu a, b, Peng Zhang a, Xinyun Zhai a, Feng Tian b, Wenchen Li a, Jianhai Yang a, Yuan Liu a,
Hongbo Wang a, Wei Wang a, Wenguang Liu a, *
a
School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin 300072, PR China
b
Institute of Medical Equipment, Academy of Military Medical Sciences, Tianjin 300161, PR China

a r t i c l e i n f o a b s t r a c t

Article history: Polyethylenimine (PEI) functionalized carbon dots (CD-PEI) were fabricated by one-step microwave
Received 16 January 2012 assisted pyrolysis of glycerol and branched PEI25k mixture where the formation of carbon nanoparticles
Accepted 29 January 2012 and the surface passivation were accomplished simultaneously. In this hybrid C-dot, PEI molecule played
Available online 16 February 2012
two key roles in the system  as a nitrogen-rich compound to passivate surface to enhance the fluo-
rescence and as a polyelectrolyte to condense DNA. This CD-PEI was shown to be water soluble and emit
Keywords:
stable bright multicolor fluorescence relying on excitation wavelength. The DNA condensation capability
Carbon dots
and cytotoxicity of CD-PEI could be regulated by pyrolysis time possibly due to the somewhat destruction
Microwave
Gene delivery
of PEI during the formation of carbon dots. CD-PEI obtained at an appropriate pyrolysis time exhibited
Photoluminescence lower toxicity, higher or comparable gene expression of plasmid DNA in COS-7 cells and HepG2 cells
Bioimaging relative to control PEI25k. Intriguingly, the CD-PEIs internalized into cells displayed tunable fluorescent
emission under varying excitation wavelength, suggesting the potential application of CD-PEI in gene
delivery and bioimaging.
Ó 2012 Elsevier Ltd. All rights reserved.

1. Introduction spectra, tunable emission wavelength, and excellent biocompati-


bility compared with conventional organic dyes and semiconductor
The fusion of nanotechnology and medicine has led to the quantum dots which have raised serious health and environmental
emergence of nanomedicine where a nanomaterial platform is used hazard concerns [17]. Routes to construct C-dots can be generally
for gene/drug therapy and diagnostic probe [1,2]. The central classified into two groups: top-down and bottom-up methods. The
challenge in this field is to design multifunctional nano-carriers top-down methods, by which C-dots are generally formed through
that combine both therapeutic and diagnostic capabilities. post-treating carbon particles broken from a larger carbon structure,
Recently, many kinds of inorganic nanomaterials, such as gold consist of arc discharge [18], laser ablation [19e21], electrochemical
nanoparticles [3,4], iron oxide nanoparticles [5,6], silica nano- oxidation [22e24]. The bottom-up approaches comprise combustion
particles [7,8], semiconductor quantum dots [9e11], carbon nano- [25,26], thermal carbonization [27], acid dehydration [28] and
tubes [12,13], nanodiamonds [14] and graphene [15], have been ultrasonic treatment [29], by which C-dots are transformed from
explored as nonviral vector for nucleic acid delivery by chemical suitable molecular precursors. The approaches mentioned above
modifications on purpose. always involve intricate processes, expensive original materials or
Lately, photoluminescent carbon dots (C-dots), as one of new great energy-consuming devices, and the yield of C-dots is very low.
members of carbon nanomaterials family, have drawn tremendous The synthesized C-dots, typically, are always oxidized by nitric acid
attention in nanotechnology field [16]. These surface-passivated and further surface-passivated by diamine-terminated organic
carbon nanoparticles, with diameters lower than 10 nm, have been molecule to gain obvious photoluminescence properties.
utilized for the application of biolabeling owing to their remarkable More recently, strong photoluminescent C-dots have been
advantages in stable photoluminescence (PL), broad excitation produced directly via microwave pyrolysis of carbohydrates solu-
tion in the presence of passivation agent, 4,7,10-trioxa-1,
13-tridecanediamine (TTDDA) using a domestic microwave oven in
* Corresponding author. Tel./fax: þ86 22 27404724. our lab [30]. The strong photoluminescent performance of C-dots,
E-mail address: wgliu@tju.edu.cn (W. Liu). as we found, has a close connection with the introduction of N

0142-9612/$ e see front matter Ó 2012 Elsevier Ltd. All rights reserved.
doi:10.1016/j.biomaterials.2012.01.052
C. Liu et al. / Biomaterials 33 (2012) 3604e3613 3605

atoms onto the skeleton of carbon nanoparticles. Other studies also a standard sample to calculate the QYof test sample (i.e. C-dots) which was dissolved in
suggested that the N-containing diamine-terminated oligomeric ultrapure water at different concentrations. All the absorbance values of the solutions
at the excitation wavelength were measured with UVeVis spectrophotometer. Pho-
poly-(ethylene glycol) (PEG1500N) played an important role in the toluminescence (PL) emission spectra of all the sample solutions were recorded by
surface passivation of C-dots [20,28]. That was reminiscent of FLS920 Fluorometer at an excitation wavelength of 350 nm. The integrated fluores-
polyethylenimine (PEI), a commonly used high performance cence intensity is the area under the PL curve in the wavelength range from 380 to
nonviral vector, consisting of much more amino groups in its 700 nm. Then a graph was plotted using the integrated fluorescence intensity against
the absorbance and a trend line was added for each curve with intercept at zero.
molecular structure. So we hypothesize that, the use of PEI as the
Absolute values were calculated according to the following equation:
surface passivation agent will able to aid in generating the C-dots
  !
with both enhanced fluorescent properties and gene delivery GradX h2X
FX ¼ FST (1)
ability. However, the traditional surface functionalization process of GradST h2ST
nanoparticles usually involves time-consuming multiple steps.
Previous investigations have mostly focused on the synthesis Where the subscripts ST and X denote standard and test respectively, F is the
fluorescence quantum yield, Grad is the gradient from the plot of integrated fluo-
process, but the poor gene condensation capability has restricted
rescence intensity vs absorbance, and h is the refractive index of the solvent. In order
the use of C-dots in gene delivery itself. Herein, we report a hybrid to minimize re-absorption effects, absorbance in the 10 mm fluorescence cuvette
nano-carrier based on PEI-functionalized C-dots (CD-PEI) via one- should never exceed 0.1 at the excitation wavelength.
step microwave assisted pyrolysis of inexpensive glycerol in the
presence of branched PEI in a few minutes. The most favorable 2.5. Preparation of vectors/pDNA complexes
feature of this strategy is that the formation of carbon nanoparticles
CD-PEI and branched PEI25k were separately dissolved in ultrapure water to
and the surface passivation with PEI can be realized simultaneously form 2 mg/mL solutions which were subsequently sterilized by filtration through
in one pot. It is expected that the outer cationic polymer layer has a 0.22 mm filter. Vector/pDNA complexes at varied ratios were then formulated by
the ability to mediate plasmid DNA transfection, and the entrapped adding sterilized vector of prescribed concentrations to an equal volume of a defined
C-dots emitting discernible fluorescence can serve as bioimaging. pDNA solution, pipetting up and down to make the mixture homogeneous thor-
oughly. Then the mixtures were incubated at room temperature for 30 min to allow
complex formation completely. In this study, the complexing ratio was expressed as
2. Materials and methods
the weight ratio of vector/pDNA. Noted that the selected complexing ratios in the
following assay correspond to the maximum transfection efficiency of each vector
2.1. Materials and chemicals
unless otherwise statement.
Branched polyethylenimine (PEI) with molecular weight 25 kDa was purchased
from SigmaeAldrich (St Louis, MO, USA). Quinine sulfate (98%, suitable for fluo- 2.6. Agarose gel electrophoresis
rescence) was supplied by Fluka (New York, USA). Plasmid pGL3-control with SV40
promoter and enhancer sequences encoding luciferase (5262 bp) was obtained from The binding of pDNA with CD-PEI was evaluated by agarose gel electrophoresis.
Promega (Madison, WI, USA). 3-(4,5-dimethyl-2-thiazoyl)-2,5-diphenyl tetrazolium The vector/pDNA complexes at different weight ratios were prepared freshly as
bromide (MTT, 98%) was supplied by Alfa-Aesar (Beijing, China). The plasmids were described above. After incubation for 30 min at room temperature, 8 mL of complex-
amplified in Escherichia coli and purified with QIAGEN Plasmid Maxi Kit (Qiagen, containing solution was mixed with 2 mL loading buffer, and loaded into a 1% agarose
Hilden, Germany). Glycerol, NaH2PO4, Na2HPO4, and H2SO4 were obtained from gel containing ethidium bromide (0.5 mg/mL). The electrophoresis experiment was
Guangfu Fine Chemical (Tianjin, China). And all other chemicals were analytical carried out for 40 min in 1  TAE buffer at a constant voltage of 100 V. Then the
reagent grade and were used without further purification. pDNA bands were visualized under a UV transilluminator at a wavelength of 365 nm.

2.2. Synthesis of CD-PEI 2.7. Cell culture

The photoluminescent C-dots passivated with branched PEI was synthesized by COS-7 cells (African green monkey kidney cells) and HepG2 cells (human
a facile green route of microwave assisted pyrolysis method. Briefly, 20 ml glycerol hepatocellular liver carcinoma line) were obtained from Peking Union Medical
and 6 ml 10 mM pH7.4 phosphate solution were mixed with 0.5 g PEI25k under College (Beijing, China). COS-7 cells and HepG2 cells were separately cultured in
vigorous stirring to form a homogeneous solution in a common 100 mL beaker. Then Dulbecco’s Modified Eagle Medium (DMEM, HyClone) with high glucose, containing
the beaker containing clear transparent solution was placed at the center of the 10% fetal bovine serum (FBS), 100 U/mL penicillin and 100 mg/mL streptomycin at
rotation plate of a domestic microwave oven (700 W) and heated for different time 37  C in 5% CO2 humidified atmosphere.
intervals. When cooled down to room temperature, the color-changed solution was
then diluted and dialyzed against pure water for 4 days. Finally, a clear, light yellow- 2.8. Cell viability assay
brown aqueous solution containing CD-PEI was lyophilized to collect dry CD-PEI. In
this experiment, three CD-PEIs were designed by varying microwave irradiation time, The cytotoxicity of PEI-passivated C-dots was assessed through MTT assay. COS-
5 min, 10 min and 15 min, denoted as CD-PEI-A, CD-PEI-B and CD-PEI-C respectively. 7 cells and HepG2 cells were seeded in a 96-well plate, at a density of 2  104 cells/
well and incubated overnight, respectively. The following morning, the culture
2.3. Instrumentation and characterizations medium in each well was replaced by 180 mL fresh DMEM/FBS, and 20 mL CD-PEI/
pDNA complexes solutions with various weight ratios (DNA concentration was
Fourier Transform Infrared Spectroscopy (FT-IR) spectra were conducted on 2 mg/mL) were then added to each well. After incubation for 24 h, the medium
a Nicolet 380 spectrometer (Thermo, America). The elementary compositions were containing CD-PEI/pDNA complexes was removed, and replaced with 200 mL fresh
further confirmed by elemental analysis with Vanio-EL (Elementar Analysensysteme medium containing 20 mL MTT (5 mg/ml in PBS) and incubated for another 4 h.
GmbH, Germany). X-Ray diffraction (XRD) patterns were recorded on a Rigaku-D/ Finally all medium was removed and 150 mL/well DMSO was added, followed by
MAX 2500 diffractometer (Rigaku, Japan) equipped with graphite mono- shaking for 15 min. The absorbance of each well was measured at 490 nm using
chromatized CuKa (l ¼ 0.15405 nm) radiation at a scanning speed of 4 /min in the a Synergy HT Multi-Mode Microplate Reader (BioTek, USA) with pure DMSO as
range from 5 to 60 . The morphologies of the CD-PEI were examined by high- a blank. Non-treated cell (in DMEM) was used as a control and the relative cell
resolution transmission electron microscopy (HRTEM) on a Philips Tecnai G2F20 viability (mean%  SD, n ¼ 3) was expressed as Abssample/Abscontrol  100%.
microscope (Philips, Netherlands) with an accelerating voltage of 200 kV. UVeVis
absorption was characterized by TU-1810 UVeVis Spectrophotometer (Pgeneral, 2.9. In vitro transfection and luciferase assay
China). Photoluminescence (PL) emission measurements were performed using
FLS920 Fluorometer (Edinburgh Instruments, Britain). The vector/pDNA complexes In vitro transfection of CD-PEI-A, CD-PEI-B and CD-PEI-C were assayed in COS-7
were observed under JEM-100CX II TEM (Jeol, Japan) after negatively stained with cells and HepG2 cells, respectively. The cells were seeded at a density of 5  104 cells/
1.5 wt% phosphotungstic acid (pH 6.7). Zeta potential of vector/pDNA complexes was well in 24-well plates and incubated for 24 h at 37  C, in 5% CO2 humidified
measured by using a Zetasizer Nano Z (Malvern Instruments, Malvern, UK). atmosphere. Just prior to transfection, the medium in each well was aspirated and
replaced with 450 mL of serum-free or 10% serum-containing DMEM. CD-PEI/pDNA
2.4. Measurement of fluorescence quantum yields complexes (50 mL, containing 1 mg DNA) at various weight ratios were then added to
each well (n ¼ 3 for each ratio). After incubating at 37  C in 5% CO2 for 3 h, the CD-
The quantum yield of the C-dots was determined by a comparative method. PEI/pDNA complexes that were not internalized were removed by aspirating the
Quinine sulfate in 0.1 M H2SO4 (literature quantum yield: 54%) was selected as medium and replacing with fresh medium. The transfected cells were re-incubated
3606 C. Liu et al. / Biomaterials 33 (2012) 3604e3613

Fig. 1. (a) CD-PEI aqueous solutions under UV light illumination (365 nm, from left to right: CD-PEI-A, CD-PEI-B and CD-PEI-C); (b) PL spectra of CD-PEI-A, CD-PEI-B and CD-PEI-C
aqueous solutions at the same concentration of 0.5 mg/ml (excitation wavelength: 350 nm, maxima emission wavelength: 470 nm); (c) HRTEM image of CD-PEI (scale bar is 50 nm;
the inset shows size distribution histogram); (d) XRD patterns for CD-PEI.

Fig. 2. FT-IR spectra of raw PEI and PEI-passivated C-dots.


C. Liu et al. / Biomaterials 33 (2012) 3604e3613 3607

for additional 48 h. After incubation, the culture medium was removed and the cells all the samples used for characterization were prepared under the
were washed with PBS twice. The cells in each well were treated for 15 min with conditions of 20 ml glycerol, 6 ml 10 mM phosphate, 0.5 g PEI25kD
150 mL of reporter lysis buffer (RLB, Promega) followed by freezeepumpethaw
cycles to ensure complete lysis. The lysate was centrifuged for 4 min at
and microwave treatment for 10 min (CD-PEI-B) unless specifically
13,000 rpm and the supernatant was collected for luminescence measurements. The mentioned. As shown in Fig. 1a, the prepared C-dots passivated
luminescence of each sample was measured by 1420 Multilabel counter (Wallac, with PEI exhibited excellent water-soluble properties and emitted
USA) using Bright-GloÔ luciferase assay system (Promega, USA) according to the blue luminescence under UV light (365 nm). The full width at a half
manufacturer’s protocol. The results were expressed as relative light units (RLU) per
maximum (FWHM) at excitation of 350 nm is about 90 nm (Fig. 1b)
milligram of cell protein, and the protein concentration of each well was measured
by a BCA protein assay (Pierce, Rockford, IL, USA). Branched polyethylenimine indicating a narrow size distribution of C-dots. Quinine sulfate
25 kDa (PEI25k) served as a positive control. PEI25k/pDNA complex with weight (quantum yield 54%) was selected as a standard sample to calculate
ratio of 2/1 was prepared as suggested protocol. the quantum yield of C-dots and the results (Fig. S1) are CD-PEI-A
9.4%, CD-PEI-B 15.3% and CD-PEI-C 7.0%, respectively. There is an
2.10. Confocal microscopy
interesting phenomenon that only a proper microwave pyrolysis
For confocal microscopy observation, COS-7 cells were seeded over a glass time results in high performance photoluminescence. To under-
coverslip in 6-well plate for 24 h and then transfected with sterilized CD-PEI-B/ stand this, we assume that there exist three steps in the whole
pDNA at weight ratio of 16:1 for 3 h at 37  C. The cells were then washed with pyrolysis process, that is, (i) glycerol dehydration and nanoparticle
isotonic PBS (pH 7.4) three times, and fixed with 4% paraformaldehyde solution in formation, (ii) surface passivation of nanoparticle and (iii) growth
PBS at 4  C overnight. The samples were observed under a Leica laser scanning
confocal microscope (Mannheim, Germany) equipped with a UV laser (351/364 nm),
of carbon dots. The three steps have no well-defined boundary and
an Ar laser (457/488/514 nm) and a HeNe laser (543/633 nm). Non-transfected COS- might occur synchronously. Therefore, the C-dots made by 5 min
7 cells served as a negative control. and 15 min pyrolysis showing weak photoluminescence may be
ascribed to the incomplete surface passivation before 10 min and
3. Results and discussions large particle formation after 10 min [20]. Although the origins of
photoluminescence are not yet entirely understood in C-dots, there
3.1. Characterization of CD-PEI is mounting evidence that emission arises from the radioactive
recombination of excitations located at surface energy traps [16].
In this work, we presented three PEI-passivated C-dots with The luminescence lifetime of PEI-passivated C-dots was also
different microwave irradiation time. In the following discussion, measured, and the computed results are CD-PEI-A 5.38 ns, CD-PEI-

Fig. 3. (a) Fluorescent microscopy images (all scale bars: 500 mm) of diluted aqueous solution of CD-PEI observed under different excitation wavelengths from left to right:
ultraviolet, blue and green; the pictures were taken by an Olympus BX-51 optical system microscope (Tokyo, Japan). (b) Photoluminescence emission spectra of CD-PEI
(progressively longer excitation wavelengths from 340 nm to 500 nm with a 20 nm increment) and the emission spectral intensities are normalized in the inset. (For interpre-
tation of the references to colour in this figure legend, the reader is referred to the web version of this article.)
3608 C. Liu et al. / Biomaterials 33 (2012) 3604e3613

B 5.74 ns and CD-PEI-C 5.09 ns (Fig. S2). Additionally, the influence The CD-PEI could emit multicolor luminescence under different
of pH value of CD-PEI solution on the PL intensity was also exam- excitation illuminations, which was observed through fluorescent
ined (Fig. S3). The results suggest that the PL intensity of CD-PEI is microscopy. Typically, a tiny drop of aqueous solution containing
very stable in the pH range of 5e12 with a slight increase in the the CD-PEI was placed on a cover glass. Fig. 3a shows that the drop
acidic conditions. The high-resolution transmission electron exhibits blue, green and red luminescence under ultraviolet
microscopy (HRTEM) image, as shown in Fig. 1c, indicates that the (330e385 nm), blue (460e495 nm) and green (530e550 nm) light
formed carbon nanoparticles have a diameter distribution of excitation, respectively. The preeminent multicolor photo-
4e12 nm and the average value is about 7 nm. X-ray diffraction luminescent feature has great potential application in bioimaging,
(XRD) pattern (Fig. 1d) displays a broad diffraction peak at which will be discussed in the next section. To further investigate
2q ¼ 18.3 , suggesting an amorphous nature. The composition of C- the wavelength-dependence feature of C-dots, the PL spectra of CD-
dots passivated with PEI was revealed by Elemental analysis and PEI solution was recorded by changing the excitation wavelengths.
the results are as follows: CD-PEI-A C 47.53%, N 24.71%, H 9.82% and When the excitation wavelengths changes from 340 nm to 500 nm,
O (calculated) 17.94%; CD-PEI-B C 54.13%, N 22.93%, H 9.41% and O the emission peak position, as shown in Fig. 3b, is red-shifted from
(calculated) 13.53%; and CD-PEI-C C 57.60%, N 18.27%, H8.75% and O 450 (blue) to 550 nm (green), and the PL intensity decreased
(calculated) 15.38%. The result implies that more aminos in PEI remarkably, indicating a strong dependence on the excitation
were kept with short time microwave treatment. wavelengths. Although, up to date, the origins of PL in C-dots are
FT-IR spectra were recorded to identify the organic functional not very clear, we reason that this wavelength-dependent
groups on PEI-passivated C-dots. As shown in Fig. 2, the new peak phenomena (or multicolor PL) may come from the different
at 1645 cm1, the strong peaks of 1570 cm1, 1472 cm1 and distribution of emissive energy traps on the surface of C-dots, as
1309 cm1 are attributed to amide I C]O, NeH, CH2 and CeN suggested in the literature [13,22].
respectively. The broad bands centered at 3360 cm1 suggests the The formation of surface passivation, just as reported [20], plays
existence of eOH and NeH. A noticeable difference between raw a key role in the strong fluorescence of C-dots. To reconfirm the
PEI and PEI-functionalized carbon dots lies in the new absorption estimate, we also constructed a parallel control experiment. Sample
band located at 1645 cm1, and the absorption of NeH located at X, Y and Z were microwave-assisted pyrolysis products from
1570 cm1. As the time of microwave treatment increases, the PEI25k, glycerol and the mixture of glycerol and PEI respectively. As
absorption of C]O becomes stronger and stronger, while the NeH shown in Fig. S4, sample X has no fluorescence and sample Y shows
peak shows a declining trend. This suggests more amide groups weak fluorescence when excited at 350 nm. The quantum yield of
formed through longer pyrolysis endurance time and the PEI sample Y is only 4.63%, the same as reported in our previous work
molecules were chemically grafted onto the surface of carbon dots [30], much lower than that of sample Z. When PEI was used as
in situ. In the glycerol pyrolysis process, the carboxylic groups a surface passivation agent, sample Z exhibits a very strong fluo-
converted into amide groups immediately after they have formed rescent emission owing to the successful introduction of nitrogen-
on the surface of nanoparticle. The grafted PEI molecules not only containing organic molecules onto the surface of C-dots. It is worth
render C-dots water soluble, but also passivate the surface of CD to mentioning that the PEI, after treated by microwave irradiation
enhance the photoluminescence. 10 min, was nonemissive in the UVeVis range. Therefore the

Scheme 1. Depiction of the formation of CD-PEI and CD-PEI/pDNA complex.


C. Liu et al. / Biomaterials 33 (2012) 3604e3613 3609

observed bright and colorful photoluminescence should arise from gel electrophoresis assay. Fig. 4 shows that the intensity of DNA
the PEI-passivated carbon nanoparticles. migration bands in the agarose gel decreases gradually with the
The influence of pyrolysis time and PEI25k dosage on the fluo- increase of weight ratio. For raw PEI25k, CD-PEI-A, CD-PEI-B and
rescence of PEI-passivated C-dots was also investigated thoroughly CD-PEI-C, the electrophoretic mobility of pDNA was thoroughly
(Fig. S5 and Fig. S6). As shown in Fig. S5, the PL intensity reaches confined at or above the weight ratio of 0.2, 0.3, 0.4 and 0.5,
a maximum when the microwave treating time is 10 min and then respectively, suggesting all the CD-PEI samples are capable of
a downward trend appears with time extension. Originally, condensing DNA at a very low weight ratio, though the condensing
a certain amount of water in the raw solution was necessary to form ability decreases for longer pyrolysis time.
a transparent homogenous solution with an appropriate viscosity Surface charge of the complexes also reflects the ability of
for carbonization uniformity. With heating, the water was evapo- cationic vectors to condense DNA. Zeta potential of various CD-PEI/
rated before the glycerol dehydration, which was the reason why pDNA complexes was measured to determine their surface charges.
no obvious PL was observed before 2 min. The longer the irradiation Fig. 5a shows the zeta potential data of CD-PEI-A/pDNA, CD-PEI-B/
time is, the stronger the PL spectrum emits. But when time is longer pDNA and CD-PEI-C/pDNA complexes at different weight ratios,
enough to exceed 20 min, the sample shows a relative weak PL respectively. The zeta potential of vector/pDNA complexes exhibits
spectrum, suggesting the change in the diameter size or surface an obvious rising trend along with an increase in weight ratio from
structure of C-dots. From Fig. S6, one can find that a proper dosage 2 to 24. Moreover, over the selected range of complexing ratio, the
of surface-passivation reagent is necessary for strong phtolumi- zeta potential of three complexes is in the descending order:
nescent C-dots as reported in our previous work [30]. As shown in CD-PEI-A/DNA > CD-PEI-B/DNA > CD-PEI-C/DNA. This trend also
the figure, the optimal dosage of PEI25k for the preparation of CD- reflects the order of DNA condensation ability, and the result is
PEI with highest QY is 0.5 g. consistent with that obtained by electrophoresis. Microwave irra-
diation can generate a fast heating effect in the original mixture,
3.2. Formation of CD-PEI/pDNA complexes causing glycerol dehydration, polymerization and carbonization. In
this process, the amino groups of PEI molecules are also consumed
As depicted in Scheme 1, the branched PEI-functionalized C-dots by taking part in the reactions; the longer irradiation time could
condense DNA by electrostatic interactions between positively
charged PEI on the surface of CD-PEIs and negatively charged
phosphate backbone of pDNA. The ability of PEI-passivated C-dots
to bind DNA at different complex ratios was investigated by agarose

Fig. 4. Agarose gel electrophoresis patterns of complexes: (a) PEI25k/DNA, (b) CD-PEI- Fig. 5. Zeta potential data (a) and particle size (b) of vector/DNA complexes at different
A/DNA, (c) CD-PEI-B/DNA and (d) CD-PEI-C/DNA. weight ratio.
3610 C. Liu et al. / Biomaterials 33 (2012) 3604e3613

Fig. 6. TEM images of negatively stained vector/pDNA complexes: CD-PEI-A/pDNA at 6:1 wt/wt (a), CD-PEI-B/pDNA at 16:1 wt/wt (b) and CD-PEI-C/pDNA at 24:1 wt/wt (c). The
scale bars in the all images are 50 nm.

lead to more consumption of amino groups. Since zeta potential is The morphology and average size of CD-PEI/pDNA complexes
determined mainly by the surface charge of vector/pDNA were further inspected by both transmission electron microscope
complexes, CD-PEI-C/DNA displays the lowest potential while CD- (TEM) and dynamic light scattering (DLS). As shown in Fig. 6, at the
PEI-A/DNA shows the highest. selected vector/pDNA complex weight ratios, CD-PEI-A, CD-PEI-B

Fig. 7. Cytotoxicity testing results of vectors/pDNA complexes with a constant DNA concentration of 2 mg/mL against (a) COS-7 cells and (b) HepG2 cells from an MTT assay. The
values represent percentage cell viability (means  SD, n ¼ 3).
C. Liu et al. / Biomaterials 33 (2012) 3604e3613 3611

and CD-PEI-C are all able to condense DNA into nano-sized particles PEI-A/DNA at weight ratio 6:1, CD-PEI-B/DNA at 8:1 and CD-PEI-C/
(below 50 nm), which is favorable for the efficient cellular delivery. DNA at 20:1, respectively. Similar cytotoxicity results are obtained
The corresponding hydrodynamic diameters of complexes (Fig. 5b) for HepG2 cells (Fig. 7b), and about 80% cells keep viable for CD-PEI-
are larger than TEM results due to the hydration effect in aqueous A/DNA at weight ratio 4:1, CD-PEI-B/DNA at 10:1 and CD-PEI-C/DNA
solution. Among all three nano-carriers, CD-PEI-A forms the at 24:1, respectively. As the ratios of vector to DNA are raised up to
smallest nanoplexes with pDNA, which is attributed to its highest 18:1, the cytotoxicity of CD-PEI-A/DNA and PEI25k/DNA against both
surface positive charge. COS-7 and HepG2 cells is enhanced remarkably, but that of CD-PEI-B
and CD-PEI-C obtained at longer microwave treatment time show
3.3. Cytotoxicity evaluation of the CD-PEI a relative mild increase at the identical complexing ratio, possibly
due to the destruction of more aminos in PEI.
With integration of transfection ability and effectual bioimaging
in one single C-dot, CD-PEI nanovector is required to have not only 3.4. In vitro transfection of CD-PEI/pDNA complexes
optical merits, high transfection efficiency but also low cytotoxicity.
To evaluate the cytotoxicity of three CD-PEI vectors, the relative In order to evaluate the transfection ability of PEI-passivated C-
viabilities of COS-7 cells and HepG2 cells exposed to CD-PEI or dots, we transfected COS-7 and HepG2 cells with CD-PEI nano-
complexes were measured by using the MTT assay method with vectors both in serum-free and serum-containing conditions, and
branched PEI25k as a control(Fig. 7S and Fig. 7). All the samples the results are shown in Fig. 8. When COS-7 cells were transfected
exhibit a dose-dependent cytotoxicity. It is well known that the in medium without serum (Fig. 8a), the luciferase expression of
cytotoxicity of PEI25k is attributed to its abundant positive charges pGL-3 (RLU/mg protein) mediated by CD-PEI-A at weight ratio of
which can cause serious cell membrane damage [31]. In comparison 6:1 and CD-PEI-B at 16:1 are higher than or comparable to that of
to the PEI25k and PEI25k/DNA controls, CD-PEI and CD-PEI/DNA PEI25k. When the complex ratio further increases, the gene
complexes exhibit remarkably less toxicity in COS-7 cells and expression shows a decrease trend, which is probably due to the
HepG2 cells at the same concentrations or complexing ratios, increased cytotoxicity effect at high dose of CD-PEI. For the trans-
respectively. It is probably because large amount of amino groups of fection of COS-7 cells in serum-containing medium (Fig. 8b), CD-
PEI molecules were destroyed in the passivation process or PEI-A and CD-PEI-B can also achieve a gene expression level
embedded in the coreeshell structure of the nanoparticles. As superior to PEI25k, despite slightly lower than serum-free case. In
shown in Fig. 7a, there are more than 80% COS-7 cells viable for CD- the presence of the negatively charged serum, the optimal complex

Fig. 8. In vitro gene transfection efficiency of the vectors (CD-PEI-A, CD-PEI-B, and CD-PEI-C)/pDNA complexes in comparison with those of PEI25k and naked DNA (ND) at various
weight ratios in COS-7 cells in the absence (a) and in the presence of serum (b); HepG2 cells in the absence (c) and in the presence of serum (d). Results are presented as the
mean  SD in triplicate.
3612 C. Liu et al. / Biomaterials 33 (2012) 3604e3613

Fig. 9. Laser scanning confocal microscopy images of non-transfected cells as negative controls (a) and transfected cells (b). The samples were observed under bright field and
excited at 405 nm, 488 nm and 543 nm (All scale bars: 20 mm).

ratios of CD-PEI-A and CD-PEI-B are increased to 8:1 and 18:1, untreated cells remain dark, suggesting large amount of nano-
respectively. Similar results of luciferase express are obtained in particles have been internalized into the cells. After 3-h trans-
HepG2 cells (Fig. 8c,d). It is noted that CD-PEI-C remains low fection, most nanoparticles uptaken appear in endosomal
transfection efficiency over the whole complexing ratio range, compartments; while some complexes have already successfully
though generally it exhibits an increasing trend. From the above escaped from endosome in such short time. Consistent with fluo-
results, we can draw the conclusion that with longer microwave rescence microscopy assay results (Fig. 3), CD-PEI shows blue, green
pyrolysis time, the transfection ability of C-dots first increases and and red color under 405 nm, 488 nm and 543 nm laser excitation
then decreases. As discussed above, during the formation of CD-PEI, wavelength. This multicolor emission is a great advantage of the C-
the amino groups in PEI are involved in reaction. As for CD-PEI-A, dots over other labeling agents, because it gives us much freedom
which was only treated under microwave for 5 min, a small to choose the wavelength for observation. It is noteworthy that,
amount of the amino groups were consumed. Nonetheless, such when the excitation wavelength was red-shifted enough, the C-
slight destruction of PEI molecule does not affect its DNA protection dots could even exhibit near-infrared emissions; though relatively
ability and buffer effect. Conversely, a slight loss of the positive weak, it may also have the potential to be used in the fluorescent
charged molecule could improve the biocompatibility of the tracking in vivo.
cationic carrier; on the other hand, the formation of coreeshell
structured CD-PEI may be the reason for the enhanced trans- 4. Conclusion
fection level. When the microwave treatment was further increased
to 10 min, an increased amount of PEI molecules may be damaged. In summary, we successfully constructed a high-efficient dual
Under this condition, high transfection efficiency can still be ob- functional nano gene vector based on PEI-passivated C-dots by one-
tained by simply increasing the complexing ratio. However, when step microwave assisted pyrolysis of glycerol in the presence of
the pyrolysis time lasts more than 15 min, too much amino groups branched PEI25k. In this hybrid nano-carrier system, PEI played
could have been destroyed. As a result, the positive charge of the two crucial roles: surface passivation to endow the C-dots with
obtained C-dots may be too weak to effectively protect the DNA strong photoluminescence, DNA condensation for gene trans-
from degradation, and meanwhile the reduced buffer capability fection. The elaborately fabricated CD-PEI exhibited excellent water
cannot lead to efficient escape of complexes from endosome either. solubility and bright multicolor fluorescence with great photo-
stability. CD-PEI could mediate gene transfection in COS-7 and
3.5. Confocal microscopy HepG2 cells with higher or comparable efficiency as well as lower
cytotoxicity relative to pristine PEI25k. To optimize the perfor-
CD-PEI fabricated in this study shows multicolor fluorescence mance of CD-PEI vector, proper pyrolysis time was essential to
under different laser excitation. This remarkable fluorescent feature balance between formation of C-dots and destruction of PEI. The
endows CD-PEI with an intrinsic ability to combine dual functions multicolor fluorescence of the C-dot/DNA complexes could be
of gene delivery and bioimaging. To further confirm the multi- clearly observed in cytosol, demonstrating the great potential of
functions of the C-dots, COS-7 cells were selected for the trans- CD-PEI in bioimaging and biolabeling.
fection and labeling by CD-PEI-B/pDNA complexes. Fig. 9 displays
photographs of the transfected COS-7 cells and negative controls Acknowledgement
captured by a laser scanning confocal microscope. We can see that
the transfected COS-7 cells become quite bright owing to the strong The authors gratefully acknowledge the support for this
fluorescence emitting from CD-PEI distributed in cytosol, while the work from the National Natural Science Foundation of China
C. Liu et al. / Biomaterials 33 (2012) 3604e3613 3613

(Grant 50973082) and National Science and Technology Major [13] Liu M, Chen B, Huang J, Zhang L, Zhang Z. Polyamidoamine-grafted multi-
walled carbon nanotubes for gene delivery: synthesis, transfection and
Project of China (Grant 2012ZX10004801-003-007).
intracellular trafficking. Bioconjug Chem 2011;22:2237e43.
[14] Zhang P, Yang J, Li W, Wang W, Liu C, Griffith M, et al. Cationic polymer brush
grafted nanodiamond via atom transfer radical polymerization for enhanced
Appendix. Supplementary material gene delivery and bioimaging. J Mater Chem 2011;21:7755e64.
[15] Kim H, Namgung R, Singha K, Oh IIK, KIM WJ. Graphene oxide-
Supplementary material associated with this article can be found, polyethylenimine nanoconstruct as a gene delivery vector and bioimaging
tool. Bioconjug Chem; 2011. doi:10.1021/bc200397j.
in the online version, at doi:10.1016/j.biomaterials.2012.01.052. [16] Baker SN, Baker GA. Luminescent carbon nanodots: emergent nanolights.
Angew Chem Int Ed 2010;49(38):6726e44.
[17] Hardman R. A toxicologic review of quantum dots: toxicity depends on
References physicochemical and environmental factors. Environ Health Perspect 2006;
114(2):165e72.
[1] Roy I, Ohulchanskyy TY, Bharali DJ, Pudavar HE, Mistretta RA, Kaur N, et al. [18] Xu XY, Ray R, Gu YL, Ploehn HJ, Gearheart L, Raker K, et al. Electrophoretic
Optical tracking of organically modified silica nanoparticles as DNA carriers: analysis and purification of fluorescent single-walled carbon nanotube frag-
a nonviral, nanomedicine approach for gene delivery. PNAS 2005;102(2): ments. J Am Chem Soc 2004;126(40):12736e7.
279e84. [19] Cao L, Wang X, Meziani MJ, Lu F, Wang HF, Luo PG, et al. Carbon dots for
[2] Barreto JA, O’Malley W, Kubeil M, Graham B. Stephan Holger, Spiccia L. multiphoton bioimaging. J Am Chem Soc 2007;129:11318e9.
Nanomaterials: applications in cancer imaging and therapy. Adv Mater 2011; [20] Sun YP, Zhou B, Lin Y, Wang W, Fernando KAS, Pathak P, et al. Quantum-sized
23:H18e40. carbon dots for bright and colorful photoluminescence. J Am Chem Soc 2006;
[3] Guo S, Huang Y, Jiang Q, Sun Y, Deng L, Liang Z, et al. Enhanced gene delivery 128(24):7756e7.
and siRNA silencing by gold nanoparticles coated with charge-reversal poly- [21] Wang X, Cao L, Lu FS, Meziani MJ, Li H, Qi G, et al. Photoinduced electron
electrolyte. ACS Nano 2010;4(9):5505e11. transfers with carbon dots. Chem Commun 2009;25:3774e6.
[4] Lee Y, Lee SH, Kim JS, Maruyama A, Chen X, Park TG. Controlled synthesis of [22] Zhao QL, Zhang ZL, Huang BH, Peng J, Zhang M, Pang DW. Facile preparation of
PEI-coated gold nanoparticles using reductive catechol chemistry for siRNA low cytotoxicity fluorescent carbon nanocrystals by electrooxidation of
delivery. J Control Release 2011;155(1):3e10. graphite. Chem Commun 2008;41:5116e8.
[5] Veiseh O, Kievit FM, Mok H, Ayesh J, Clark C, Fang C, et al. Cell transcytosing [23] Zheng LY, Chi YW, Dong YQ, Lin JP, Wang BB. Electrochemiluminescence of
poly-arginine coated magnetic nanovector for safe and effective siRNA water-soluble carbon nanocrystals released electrochemically from graphite.
delivery. Biomaterials 2011;32:5717e25. J Am Chem Soc 2009;131:4564e5.
[6] Namgung R, Singha K, Yu MK, Jon S, Kim YS, Ahn Y, et al. Hybrid super- [24] Zhou J, Booker C, Li R, Zhou X, Sham TK, Sun X, et al. An electrochemical
paramagnetic iron oxide nanoparticle-branched polyethylenimine magneto- avenue to blue luminescent nanocrystals from multiwalled carbon nanotubes
plexes for gene transfection of vascular endothelial cells. Biomaterials 2010; (MWCNTs). J Am Chem Soc 2007;129:744e5.
31:4204e13. [25] Liu H, Ye T, Mao C. Fluorescent carbon nanoparticles derived from candle soot.
[7] Radu DR, Lai CY, Jeftinija K, Rowe EW, Jeftinija S, Lin VS. A polyamidoamine Angew Chem Int Ed 2007;46:6473e5.
dendrimer-capped mesoporous silica nanosphere-based gene transfection [26] Tian L, Ghosh D, Chen W, Pradhan S, Chang X, Chen S. Nanosized carbon
reagent. J Am Chem Soc 2004;126:13216e7. particles from natural gas soot. Chem Mater 2009;21:2803e9.
[8] He WT, Xue YN, Peng N, Liu WM, Zhuo RX, Huang SW. One-pot preparation of [27] Yang ZC, Wang M, Yong AM, Wong SY, Zhang XH, Tan H, et al. Intrinsically
polyethylenimine-silica nanoparticles as serum-resistant gene delivery fluorescent carbon dots with tunable emission derived from hydrothermal
vectors: intracellular trafficking and transfection. J Mater Chem 2011;21: treatment of glucose in the presence of monopotassium phosphate. Chem
10496e503. Commun 2011;47:11615e7.
[9] Lee H, Kim IK, Park TG. Intracellular trafficking and unpacking of siRNA/quantum [28] Peng H, Travas-Sejdic J. Simple aqueous solution route to luminescent car-
dot-PEI complexes modified with and without cell penetrating peptide: bogenic dots from carbohydrates. Chem Mater 2009;21:5563e5.
confocal and flow cytometric FRET analysis. Bioconjug Chem 2010;21:289e95. [29] Li H, He X, Liu Y, Huang H, Lian S, Lee ST, et al. One-step ultrasonic synthesis of
[10] Zhang P, Liu W. ZnO QD@PMAA-co-PDMAEMA nonviral vector for plasmid water-soluble carbon nanoparticles with excellent photoluminescent prop-
DNA delivery and bioimaging. Biomaterials 2010;31:3087e94. erties. Carbon 2011;49:605e9.
[11] Bardi G, Malvindi MA, Gherardini L, Costa M, Pompa PP, Cingolani R, et al. The [30] Liu CJ, Zhang P, Tian F, Li WC, Li F, Liu WG. One-step synthesis of surface
biocompatibility of amino functionalized CdSe/ZnS quantum-dot-doped SiO2 passivated carbon nanodots by microwave assisted pyrolysis for enhanced
nanoparticles with primary neural cells and their gene carrying performance. multicolor photoluminescence and bioimaging. J Mater Chem 2011;21:
Biomaterials 2010;31(25):6555e66. 13163e71.
[12] Pantarotto D, Singh R, McCarthy D, Erhardt M, Briand JP, Prato M, et al. [31] Moghimi SM, Symonds P, Murray JC, Hunter AC, Debska G, Szewczyk A.
Functionalized carbon nanotubes for plasmid DNA gene delivery. Angew A two-stage poly(ethylenimine)-mediated cytotoxicity: implications for gene
Chem Int Ed 2004;43(39):5242e6. transfer/therapy. Mol Ther 2005;11:990e5.

Potrebbero piacerti anche