You are on page 1of 14

pubs.acs.

org/molecularpharmaceutics Article

Assessment of Amphiphilic Poly‑N‑vinylpyrrolidone Nanoparticles’


Biocompatibility with Endothelial Cells in Vitro and Delivery of an
Anti-Inflammatory Drug
Aikaterini Berdiaki, Emmanouela Perisynaki, Antonios Stratidakis, Pavel P. Kulikov, Andrey N. Kuskov,
Polychronis Stivaktakis, Petra Henrich-Noack, Anna L. Luss, Mikhail M. Shtilman, George N. Tzanakakis,
Aristidis Tsatsakis, and Dragana Nikitovic*
Cite This: Mol. Pharmaceutics 2020, 17, 4212−4225 Read Online
See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.
Downloaded via UNIV NACIONAL DE LA PLATA on November 29, 2022 at 21:09:07 (UTC).

ACCESS Metrics & More Article Recommendations *


sı Supporting Information

ABSTRACT: Nanoparticles (NPs) produced from amphiphilic


derivatives of poly-N-vinylpyrrolidone (Amph-PVP), composed of
various molecular weight polymeric hydrophilic fragments linked
into hydrophobic n-alkyl chains of varying lengths, were previously
shown to exert excellent biocompatibility. Although routes of
administration can be different, finally, most nanosystems enter the
blood circulation or lymphatic vessels, and by this, they establish
direct contact with endothelial cells. In this study, Amph-PVP NPs
and fluorescently labeled Amph-PVP-based NPs, namely “PVP”
NPs (Amph-PVP-NPs (6000 Da) unloaded) and “F”-NPs (Amph-
PVP-NPs (6000 Da) loaded with fluorescent FITC), were synthesized to study Amph-PVP NPs interactions with HMEC-1
endothelial cells. PVP NPs were readily uptaken by HMEC-1 cells in a concentration-dependent manner, as demonstrated by
immunofluorescence imaging. Upon uptake, the FITC dye was localized to the perinuclear region and cytoplasm of treated cells.
The generation of lipopolysaccharide (LPS)-induced activated endothelium model revealed an increased uptake of PVPNPs, as
shown by confocal microscopy. Both unloaded PVP NPs and F-NPs did not affect EC viability in the 0.01 to 0.066 mg/mL range.
Furthermore, we focused on the potential immunological activation of HMEC-1 endothelial cells upon PVPNPs treatment by
assessing the expression of their E-Selectin, ICAM-1, and VCAM-1 adhesion receptors. None of the adhesion molecules were
affected by NP treatments of both activated by LPS and nonactivated HMEC-1 cells, at the utilized concentrations (p = NS). In this
study, PVP (6000 Da) NPs were used to encapsulate indomethacin, a widely used anti-inflammatory drug. The synthesized drug
carrier complex did not affect HMEC-1 cell growth and expression of E-selectin, ICAM-1, and VCAM-1 adhesion receptors. In
summary, PVP-based NPs are safe for use on both basal and activated endothelium, which more accurately mimics pathological
conditions. Amph-PVP NPs are a promising drug delivery system.
KEYWORDS: poly-N-vinylpyrrolidone, amphiphilic polymer, nanoparticle, endothelial cells, assessment, viability,
immunological activation, fluorescent probes

1. INTRODUCTION plethora of hydrophobic drugs is accomplished. The formation


Nanoparticles (NPs) are ultrafine units with dimensions of drug-carrier complexes enhances their biological compati-
measured in nanometers. Based on their morphology, shapes, bility, stability, and, importantly, bioavailability in aqueous
or sizes, they are classified into different categories. The biological solutions.7,8 The preliminary examination of Amph-
application of NPs systems in biomedicine is increasingly PVPNPs, in vitro and in vivo, demonstrated no adverse effects
popular due to their ability to deliver various drugs to specific on cell viability and satisfactory biocompatibility verifying,
biological targets, resolving, thus, unmet medical and thus, their application potential.8−10 Noteworthy, the utiliza-
pharmaceutical needs.1−4 Our previous studies have focused tion of some poly-N-PVP-based carriers has been approved for
on synthesizing a novel drug delivery system based on NPs
produced from amphiphilic derivatives of poly-N-vinylpyrroli- Received: June 25, 2020
done (Amph-PVP) composed of various molecular weight Revised: September 27, 2020
polymeric hydrophilic fragments which were linked into Accepted: September 28, 2020
hydrophobic n-alkyl chains of varying lengths.5,6 Importantly, Published: September 28, 2020
we have also shown that upon self-assembly of such core−shell
polymeric NPs in aqueous media, an efficient entrapment of a

© 2020 American Chemical Society https://dx.doi.org/10.1021/acs.molpharmaceut.0c00667


4212 Mol. Pharmaceutics 2020, 17, 4212−4225
Molecular Pharmaceutics pubs.acs.org/molecularpharmaceutics Article

clinical studies.11 Importantly, nanocarrier-drug complexes are fered Saline (DPBS), ethyl acetate, methanol, ethanol,
delivered via various routes of administration, including dimethyl sulfoxide (DMSO), phosphate buffer saline (PBS),
inhalation, transdermal application, oral, and injection into (ethylene glycol-bis(β-aminoethyl ether)-N,N,N′,N′-tetraacetic
blood vessels.12 In all cases, the NPs are eventually taken up by acid) buffer (EGTA), indomethacin, and all other chemicals
blood or lymphatic vessels and enter the circulatory system. used in this study were purchased from Sigma-Aldrich (Sigma-
Thus, the endothelial cells lining blood vessels are directly Aldrich, St. Louis, MS, USA) unless otherwise specified and
exposed to NPs, which can result in possible deleterious used without further purification. Analytical grade preparations
consequences regarding these cells’ homeostasis and immuno- were used for all solvents and buffer solution components. The
logical response.13−16 Endothelial cells create a semiselective Millipore Milli-Q plus System (Merck KGaA, Darmstadt,
barrier that segregates the blood contents from the Germany) was used for the preparation of distilled−deionized
parenchyma of peripheral organs and tissues, exhibiting a water.
pivotal role in propagating vascular system homeostasis 2.2. Synthesis of Amphiphilic Poly-N-vinylpyrroli-
throughout the whole body.17 Primarily, the microvascular done-Based Polymeric Materials. Amphiphilic poly-N-
endothelial cell barrier regulates crucial functions of tissues/ vinylpyrrolidone consisting of a PVP hydrophilic fragment
organs and enables homeostasis.18 The endothelial cell layer is with a molecular weight of 6000 Da and an octadecyl
intimately associated with specific structures of the extrac- hydrophobic terminal fragment-namely, PVP (6000 Da), was
ellular matrix (ECM) denominated basement membranes, synthesized (Figure 1) and characterized, including TEM
which contribute to the barrier function.19,20 analysis, as described in the previous studies.8,10
The endothelium undergoes changes in function and
morphology, known as endothelial cell activation, to
participate in the inflammatory response.21,22 These changes
include the loss of vascular integrity, the expression of adhesion
molecules, cytokine production, and the upregulation of
human leukocyte antigen (HLA) molecules. The upregulation
of adhesion molecules, such as ICAM-1 (intercellular adhesion
molecule-1), VCAM-1 (vascular cell adhesion molecule-1),
and E-Selectin allows the adherence of monocytes on the Figure 1. Synthesis scheme of amphiphilic poly-N-vinylpyrrolidone.
endothelium and their enhanced transmigration to the
subendothelium.23 Also, endothelial cells increase their Briefly, 10.0 mL of N-vinyl-2-pyrrolidone was dissolved in 20
cytokine production, such as interleukins and tumor necrosis mL of 1,4-dioxane. Octadecyl mercaptan (0.286 g) and 0.107 g
factors, which further upregulate the immune response, of 2,2′-azobis(isobutyronitrile) (AIBN) were sequentially
including leukocyte trafficking.24 added, and the reaction mixture was stirred at 70 °C for 3 h.
Furthermore, platelets interact with an intact activated Afterward, 50 mL of distilled water was added, and the
endothelium, partially due to the attenuation of physiological resulting crude polymer mixture was purified by dialysis against
inhibitory mechanisms and partially due to de novo expression water (Slide-A-Lyzer Dialysis Flask, 2K MWCO, Thermo
of adhesion molecules on the surfaces of activated endothelial Scientific, USA) and was lyophilized (Alpha 1-4 LD plus,
cells and platelets. The above enhance the risk of thrombocytic Martin Christ, Germany) before storing. 13C NMR (Figure 1),
events.25 Importantly, patients with cardiovascular disease IR-spectroscopy analysis (Figure 2), and 1H NMR (Supple-
whose endothelium function is dysregulated per se are
especially sensitive to added disturbances of endothelial
function.26
Therefore, it is of utmost importance to evaluate the
interaction of NPs with the healthy and activated endothelial
barrier. In this study, the effects of Amph-PVP-NPs (6000 Da)
(PVP) on viability and immune response of basal state
endothelial cells and an LPS-challenged model of activated
endothelium were characterized for the first time. For this
purpose, unloaded PVP-NPs (6000 Da) (PVP) and PVP-NPs
(6000 Da) loaded with indomethacin were produced. The
uptake of PVP-NPs by resting and activated endothelial cells
was assessed by separately synthesized fluorescently labeled
PVP (6000 DA) (F-NPs).

2. MATERIALS AND METHODS Figure 2. 13C NMR spectra of amphiphilic poly(N-vinyl-2-


pyrrolidone) PVP-OD6000.
2.1. Materials. N-Vinyl-2-pyrrolidone (VP), 2,2′-azobis-
(isobutyronitrile) (AIBN), 1,4-dioxane, and 6-hexane diamine
were obtained from Acros Organics (Thermo Fisher Scientific,
Geel, Belgium). Acrylic acid (AA), 5(6)-carboxyfluorescein mentary Figure S1) determined the structure of the PVP-
diacetate N-succinimidyl ester (CFSE), succinimide, N,N′- OD6000 polymer (Figure 3). The average molecular weight of
dicyclohexylcarbodiimide (DCC, 1.0 M solution in methylene the polymer is 6 kDa, whereas the hydrophobic fragment is
chloride), octadecylmercaptan (ODM), 1,6-diphenyl-1,3,5- octadecyl (Figures 1 and 2).
hexatriene (DPHT), fluorescein 5-isothiocyanate (FITC), The amphiphilic polymer’s critical aggregation concentra-
and 3-chloroperoxybenzoic acid, Dulbecco’s Phosphate-Buf- tion (CAC) was determined by fluorescence spectroscopy
4213 https://dx.doi.org/10.1021/acs.molpharmaceut.0c00667
Mol. Pharmaceutics 2020, 17, 4212−4225
Molecular Pharmaceutics pubs.acs.org/molecularpharmaceutics Article

Figure 3. IR spectra of amphiphilic poly(N-vinyl-2-pyrrolidone) PVP-OD6000. The average molecular weight of the polymer is 6 kDa, whereas the
hydrophobic fragment is octadecyl.

(Hitachi 650-10S, Japan) by using 1,6-diphenyl-1,3,5-hexa- preparations. For further investigations, all NPs samples were
triene (DPHT) as the fluorescent probe. resuspended in water or PBS (pH 7.4). The particle size
The polymer NPs were prepared via the emulsification distribution and ζ-potential were determined by the dynamic
method with solvent evaporation. Briefly, Amph-PVP was light scattering (DLS) method (Malvern Zetasizer Nano ZS,
dissolved in a minute amount of ethyl acetate to obtain a UK).
solution which was then emulsified in an aqueous phase by 2.3. Preparation and Characterization of PVP-NPs
ultrasonic treatment (Sonoplus HD 2070, Bandelin, Ger- (6000 Da) Carrying Fluorophores. F-NPs were prepared by
many). The colloidal system resulting after removing the using an emulsification method. More specifically, appropriate
organic solvent was concentrated by evaporation using a rotary amounts of the PVP (6000 Da) polymer were dispersed in a
evaporator, Laborota 4010 (Heidolph, Germany). The specific volume of H2O, and specific amounts of FITC were
polymer NP solution was frozen and lyophilized using an dissolved in the organic solvent. In this case, chloroform,
Alpha I-4LD freeze-dryer system (Martin Christ GmbH, followed by an oil-in-water (O/W) emulsification, solvent
Germany) to obtain freeze-dried, solid NP. These NPs were evaporation, and freeze-drying.
then resuspended in PBS (pH 7.4). The particle size distribution and ζ-potential were
Indomethacin loaded PVP-NPs (6000 Da) were prepared determined by the dynamic light scattering (DLS) method
following the same method as for PVP-NPs (6000 Da), by (Malvern Zetasizer Nano ZS, UK).
adding the appropriate amount of indomethacin to the initial 2.4. Cell Culture. In this study, the HMEC-1 human
polymer solution in the organic solvent, in this case, methanol, dermal microvascular endothelial cell line was utilized. Cells
followed by an oil-in-water (O/W) emulsification, solvent were grown in MCDB 131 (Biochrom AG; F0455)
evaporation, freeze-drying, and resuspension in PBS. The supplemented with 10% fetal bovine serum (FBS; Invitrogen
amount of IMC loaded in the NPs was determined by 10500-064; heat-inactivated), glutamine (10 mM; Biosera
measuring the UV absorbance at 318 nm using a Unico 2802 XCT1715), hydrocortisone (1 μg/mL), gentamycin (Invitro-
spectrophotometer (Unico, USA). The IMC content intro- gen; 15710-049), and penicillin/streptomycin (100 units/mL;
duced into the hydrophobic core of the NPs was evaluated Biosera LMA4118). Before the addition of treatments, cells
from the amount of drug incorporated in nanoparticles and the were cultured in 2% medium for 24 h at 37 °C and 5% CO2.
total weight of drug-loaded NPs form using the following 2.5. Measurement of NP Uptake Utilizing Fluores-
equation: cence. Growing cells were harvested and seeded in black 96-
weight of IMC in nanoparticles well plates (Corning; 3603) at a density of 3,500 cells per well
IMC content (%) = × 100 in 200 μL of MCDB 131 (10% FBS) for 24 h, and then, the
total weight of IMC loaded nanoparticles
medium was replaced with 200 μL of MCDB 131 (2% FBS)
The Drug Loading Efficiency (DLE) was evaluated using the for 24 h. Treatments were added for the next 48 h at 37 °C and
following equation: 5% CO2 in 2% FBS medium. The cells were washed, and lysis
weight of entrapped IMC in nanoparticles buffer was added before fluorescence measurements were
DLE (%) =
initial weight of IMC used
× 100 performed using the CyQUANT fluorometric assay (Thermo
Scientific; C7026) according to the manufacturer’s instruc-
Differential scanning calorimetry (DSC) and thermogravimet- tions. A standard curve of NP serial dilutions was calculated
ric analysis (TGA), presented in Supplementary Figures S2 and was used to convert fluorescence units to mg/mL of NPs
and S3, confirmed the absence of solvent residues in the final in the cell lysates. Fluorescence was measured in a fluorometer
4214 https://dx.doi.org/10.1021/acs.molpharmaceut.0c00667
Mol. Pharmaceutics 2020, 17, 4212−4225
Molecular Pharmaceutics pubs.acs.org/molecularpharmaceutics Article

Table 1. Characteristics of PVP (6000 Da), Indomethacin-Loaded PVP (6000 Da), and F NPs (Mean ± SD, n = 3) in PBS at
37 °C
particle size zeta potential indomethacin indomethacin loading
NPs (nm) PDI (mV) content (%) efficiency (%)
hollow PVP (6000 Da) nanoparticles 178 ± 10 0.162 ± 0.036 −7.14 ± 0.39
indomethacin loaded PVP (6000 Da) nanoparticles 194 ± 11 0.168 ± 0.044 −6.35 ± 0.32 20 94.8
FITC loaded PVP (6000 Da) nanoparticles 180 ± 8 0.149 ± 0.041 +8.08 ± 1.29
(F nanoparticles)

Figure 4. Structure of F-NPs. Structure of F-NPs - FITC dye molecules is encapsulated into Amph-PVP-NPs (6000 Da).

(Biotek, Winooski, Vermont, USA) using the proposed 2.8. Fluorescence Microscopy. HMEC-1 cells were
excitation (494 nm) and emission filters (518 nm). seeded in 24-well plates, at 150,000 cells/well, and incubated
2.6. ΜΤΤ Assay. Growing cells were harvested and seeded in complete medium for 24 h. The medium was replaced with
in 98-well plates (Corning; 3603) at a density of 15,000 cells 2% FBS medium for 24 h. Treatments were added, and the
per well in 200 μL of MCDB 131 (10% FBS) for 24 h, and cells were incubated for 48 h at 37 °C and 5% CO2. The
then, the medium was replaced with 200 μL of MCDB 131 medium was removed; the cells were washed twice with 2%
(2% FBS) for 24 h. Treatments were added for the next 48 h at FBS medium and replaced with a fresh one. Pictures were
37 °C and 5% CO2 in 2% FBS medium. The labeling of the obtained using a fluorescent microscope (Leica, DM IRE2).
cells and the measurements were performed according to the 2.9. Confocal Microscopy. HMEC-1 cells were seeded on
manufacturer’s instructions (ThermoFisher, Vybrant ΜΤΤ round coverslips placed in 24-well plates, at a concentration of
Assay). 150,000 cells/well, and incubated in complete medium for 24
2.7. Western Blot. Harvested cells were lysed with RIPA h. The media was replaced with 2% FBS medium for 24 h.
solution (50 mM Tris-HCl, 1% NP-40, 0.25% Na-deoxy- Treatments were added, and the cells were incubated for 48 h
cholate, 150 mM NaCl, 1 mM EDTA with protease and at 37 °C and 5% CO2. The cells were fixed with 5%
formaldehyde and 2% sucrose in PBS for 10 min at RT. After
phosphatase inhibitors). Equal amounts of protein were
three washes with PBS, the permeabilizing agent Triton X100
subjected to SDS-PAGE using 10% polyacrylamide gels
was added for 10 min. TO-PRO-3 iodide (Molecular Probes;
under reducing conditions. Separated protein bands were
T3605) diluted 1:500 in deionized H2O was applied for 40
transferred to nitrocellulose membranes in 10 mM (pH 11), min to stain nuclei. The coverslips were then mounted onto
containing 10% methanol. Membranes were blocked overnight slides using glycerol and visualized using confocal microscopy
at 4 °C with PBS containing 0.1% Tween-20 (PBS-Tween) (Leica, TCS SP2 SE).
and 5% (w/v) low-fat milk powder. The membranes were 2.10. Cell Growth Assay. The growth of human
incubated for 1 h at room temperature (RT) with the primary microvascular endothelial HMEC-1 cells was assessed, as
antibody in PBS containing 0.1% Tween-20 (PBS-Tween) and previously reported.10 In short, HMEC-1 growing cells from
1% (w/v) low-fat milk powder. The immune complexes were nonconfluent cultures were harvested and seeded in black 96-
detected after incubation with the appropriate peroxidase- well plates (Corning; 3603) at a density of 1.000 cells per well
conjugated secondary antibody diluted (1:3,000) in PBS- in 200 μL of DMEM (10% FBS). The cell density number was
Tween, 2% low-fat milk, using the LumiSensorChemilumines- chosen from optimization experiments. The cells were allowed
cent HRP substrate kit (Genscript; L00221 V500), according to rest overnight. Treatments were added for the next 48 h at
to the manufacturer’s instructions. Protein expression of Actin 37 °C and 5% CO2 in 0% FBS. The cells were then lysed, and
was used to correct for the amount of each sample analyzed. their number was calculated using the CyQUANT fluoro-
4215 https://dx.doi.org/10.1021/acs.molpharmaceut.0c00667
Mol. Pharmaceutics 2020, 17, 4212−4225
Molecular Pharmaceutics pubs.acs.org/molecularpharmaceutics Article

metric assay (Thermo Scientific; C7026) according to the fluorescence of cell lysate was measured. This approach
manufacturer’s instructions. Fluorescence was measured in a demonstrated that the uptake of F-NPs was concentration-
fluorometer (Biotek, Winooski, Vermont, USA) using the dependent (Figure 6B). In continuation, we wanted to
proposed excitation (485 nm) and emission (528 nm) filters. A examine the uptake of F-NPs by activated endothelial
separate standard curve was used to convert fluorescence units HMEC-1 cells. It is well established that LPS, when in contact
to cell numbers. All experiments were performed in triplicate. with endothelial cells, interacts with the TLR4 receptors and
2.11. Statistical Analysis. The statistical significance was induces their activation.27 A critical downstream mediator of
evaluated by the Student’s t test or one way-ANOVA analysis. TLR4 is nuclear factor κβ (NF-κβ). Binding of LPS to TLR4
triggers the activation of phosphokinases, which phosphorylate
3. RESULTS the cytoplasmic NF-κβ inducing its translocation to the
3.1. Synthesis and Characterization of Amph-PVP nucleus.28 HMEC-1 cells were challenged with LPS (100
NPs and Loaded with IMC Amph-PVP NPs. In this study, μg/mL) during 2 h to generate a model of activated
IMC-loaded NPs were prepared using the amphiphilic polymer endothelium. Western blot demonstrated a strong upregulation
PVP (6000 Da) and the solvent evaporation technique to of NF-κβ phosphorylation in LPS treated cells verifying their
examine the influence of drug loading on NP properties activation (Figure 6C). Basal state and immunologically
biocompatibility. The freeze-dried IMC-loaded or unloaded activated HMEC-1 cells were exposed to F-NPs during 48 h,
PVP (6000 Da) was resuspended in PBS for further at which point the cells were harvested and cell lysate was
characterization. The particle size distribution of the prepared collected. Measuring fluorescence of respective cell samples
NPs formed from the PVP (6000 Da) polymeric materials was demonstrated an increased uptake of F-NPs at 0.033 and 0.066
determined by the DLS method and is presented in Table 1. mg/mL compared to cells exposed to 0.01 mg/mL (p ≤ 0.01)
The drug loading percentage of the IMC-loaded NPs was 20% of both activated and nonactivated HMEC-1 cells (Figure 6D).
w/w, whereas indomethacin loading efficiency was 94.8%. An enhanced uptake of F-NPs was observed in cells challenged
3.2. Preparation and Characterization of PVP (6000 with LPS, compared to the nonactivated HMEC-1 cells (p ≤
Da) Carrying Fluorophores. For better evaluation of the 0.05) at all concentrations tested (Figure 6D). These results
interactions of PVP NPs with biological systems, fluorescently show not only that the uptake of PVP NPs is dependent on
labeled NPs were developed. Specifically, F-NPs were their concentration but also that the activation status of
developed by incorporating FITC molecules into the hydro- endothelial cells affects their uptake.
phobic core of PVP NPs (6000 Da), as shown in Figure 4. 3.4. Effect of PVP-NPs (6000 Da) on HMEC-1 Cell
The concentration of FITC encapsulated into the NPs was Viability at Basal and Activated States. We evaluated the
35 mg/g. The CAC of the synthesized PVP (6000 Da) effect of PVP-NPs (6000 Da) and F-NPs on HMEC-1 cells’
polymer, determined in the presence of DPHT, was 0.054 viability utilizing the MTT assay. Cells were seeded at various
mmol/L (Figure 5). The mean particle size of F-NPs was 180 concentrations and absorbance, corresponding to the number
nm (Table 1), while the ζ-potential measured in PBS at +30 of viable cells, measured after 48 h following the
°C was +8.08 ± 1.29 mV. manufacturer’s instructions (Figure 7A). This approach
revealed that the ratio of absorbance/cell number when
utilizing 8000 cells was most suitable for further studies and
was duly used in all experiments. Exposing the HMEC-1 cells
with increasing concentrations (0.01 to 0.066 mg/mL) of both
PVPNPs and F-NPs during 48 h did not affect their viability
(Figure 7B). These experiments demonstrate the biocompat-
ibility of PVP NPs with the endothelial cells even at high
concentrations (66 μg/mL). FITC dye was found not to affect
endothelial cell viability at concentrations utilized (data not
shown).
Under pathological conditions or upon mechanical damage,
the endothelial cells acquire the activated phenotype. Thus, we
generated a model of activated endothelium by exposing
HMEC-1 cells to LPS (100 μg/mL) during 2 h. The LPS
Figure 5. Fluorescence intensity of the solution of PVP-ODM with challenge is well established to induce many endothelial cell
DPHT at 25 °C. biological responses in a manner dependent on NF-kB
downstream signaling.29,30 Therefore, HMEC-1 cells chal-
lenged with LPS during 2 h were exposed to unloaded PVP
3.3. Uptake of F-NPs by Endothelial Cells. To evaluate NPs and F-NPs for 48 h after which their viability was
the uptake of PVP-NPs (6000 Da), we utilized F-NPs. evaluated. As shown in Figure 7C, neither unloaded PVP-NPs
Fluorescently labeled NPs were diluted in PBS/cell culture (6000 Da) or F-NPs affected the viability of basal and activated
medium, and the fluorescence of samples was measured. The HMEC-1 cells (p = NS).
sample fluorescence was converted to mg of NPs per ml of 3.5. Effect of Amph-PVP NPs on HMEC-1 Cells’
diluted sample (mg/mL) utilizing a concentration curve. The Adhesion Molecule Expression. To evaluate the immunos-
fluorescence of diluted NPs samples, denominated as arbitrary timulatory effect of unloaded PVPNPs (6000 Da) and F-NPs
units, was verified to be concentration-dependent, as shown in on basal state and activated endothelium, we treated HMEC-1
Figure 6A. To determine the uptake of F-NPs by HMEC-1 cells with NPs or with a combination of NPs and LPS at a
endothelial cells, we exposed these cells to respective NPs concentration verified to induce these cells’ activation (see
during 48 h, at which point the cells were harvested, and the Figure 8A). Western blot of treated cells’ extracts showed that
4216 https://dx.doi.org/10.1021/acs.molpharmaceut.0c00667
Mol. Pharmaceutics 2020, 17, 4212−4225
Molecular Pharmaceutics pubs.acs.org/molecularpharmaceutics Article

Figure 6. Uptake of PVP-NPs (6000 Da) by HMEC-1 endothelial cells. (A) Concentration-dependent fluorescence of F-NPs. (B) Uptake of F-
NPs by basal state endothelial cells, expressed as mg/mL of cells lysates. The cells were cultured in 96-well plates and treated with F-NPs for 48 h.
(C) Effect of LPS on HMEC-1 cells NF-κB activation. The position of the nearest respective protein marker band is depicted to the right. (D)
Uptake of F-NPs by activated endothelial cells expressed as mg/mL of NPs in the cells lysate; F+LPS treatments were compared to the respective F
treatments (same concentration). The cells were cultured in 96-well plates, challenged with LPS, and exposed to F-NPs for 48 h. The results
represent the average of three separate experiments. C = control; cells incubated with medium only. Statistical significance: * = p ≤ 0.05, ** = p ≤
0.01.

the exposure to PVP NPs (6000 Da) and F-NPs did not affect fluoresces with green. As shown in Figure 9, a concentration-
the expression of ICAM-1 and E-selectin adhesion molecules. dependent increase in the intracellular deposition of FITC
As expected, increased expression of ICAM-1 and E-selectin labeled PVP NPs (6000 Da) was determined. Furthermore, the
molecules upon LPS treatment was demonstrated, whereas intensity of the green stain was more intense in activated
coadministration of PVPNPs (6000 Da) or F-NPs did not endothelial cells, suggesting that upon activation, the uptake of
further affect the LPS-dependent increase of the particular PVP-NPs (6000 Da) is enhanced. These results confirm that
adhesion molecules (Figures 8B and 8C). Endothelial cells the ability of HMEC-1 endothelial cells to uptake PVP-NPs
have a crucial role in regulating blood vessel tone, monocytes (6000 Da) is dependent on their immunological status.
recruitment, and thrombogenicity so that the dysfunction of 3.7. Intracellular Localization of PVP-NPs (6000 Da).
endothelial cells is directly correlated to the pathogenesis of The cellular uptake not only is a key event for drug delivery but
various diseases.26,31 These data demonstrate the PVP NPs also is immediately correlated to the biological consequences
(6000 Da) do not exert synergistic effects on the dysregulation of NPs utilization.3,13 HMEC-1 cells were treated with a high
of activated endothelium. dose (0.066 mg/mL) of F-NPs and were subsequently
3.6. Uptake of PVP-NPs (6000 Da) by Living HMEC-1 investigated with confocal microscopy to observe the internal-
Cells. Fluorescence microscopy and F-NPs were utilized to ization and intracellular localization of PVP-NPs (6000 Da).
visualize the uptake of PVP NPs (6000 Da) by living HMEC-1 Most FITC staining was located to the perinuclear cytoplasmic
cells. The FITC dye, which has been incorporated into F-NPs, space, but diffuse staining to the cytoplasm was likewise
4217 https://dx.doi.org/10.1021/acs.molpharmaceut.0c00667
Mol. Pharmaceutics 2020, 17, 4212−4225
Molecular Pharmaceutics pubs.acs.org/molecularpharmaceutics Article

Figure 7. Effect of PVP-NPs (6000 Da) and F-NPs on the viability of HMEC-1 cells. (A) Optimization of the HMEC-1 cells’ seeding number. (B)
The effect of PVP-NPs (6000 Da) and F-NPs on HMEC-1 cells’ viability. Effect of F-NPs on the viability of LPS-activated HMEC-1 cells. The cells
were cultured in 96-well plates and treated with NPs for 48 h. C = control; cells incubated with medium only. The results represent the average of
three separate experiments. Statistical significance: NS = not significant.

evident (Figure 10A, B). Our data indicate that PVP-NPs selectin, ICAM-1, and VCAM-1 specific antibodies. As
(6000 Da) are compatible with uptake/transport through the presented in Figure 12, Western blot demonstrated that
endothelium barrier. none of the treatments induced significant changes in the
3.8. Effect of IMC-Loaded PVP-NPs (6000 Da) on expression of E-selectin, ICAM-1, and VCAM-1 as compared
HMEC-1 Cell Growth. In continuation, we wanted to evaluate to the control.
the effect of a drug-carrier complex on endothelial cell growth
and viability. The effect of IMC-loaded PVP-NPs (6000 Da) 4. DISCUSSION
and unloaded PVP-NPs (6000 Da) on the proliferative
capacity of HMEC-1 cells was studied. In the experiments Due to their capability to entrap hydrophobic drugs and
performed, three different concentrations of 0.01, 0.033, and increase their bioavailability and pharmaceutical efficiency,
0.066 mg/mL of indomethacin, unloaded PVP-NPs (6000 while decreasing side toxicity, polymeric NPs have been widely
Da), and PVP-NPs (6000 Da) encapsulating Indomethacin investigated and applied in biomedical areas, especially for the
(PVP-IMC) were administered for 48 h, respectively. The creation of novel drug delivery systems.9,10,33,34 Despite the
utilization of the CyQuant proliferative assay demonstrated global use of different nanoscaled polymeric drug carriers for
that the increased concentration of indomethacin inhibits research, more detailed information concerning their biological
endothelial cells’ growth at higher concentrations of 0.033 and effects, toxicity, and safety are still required.35−38 The vascular
0.066 mg/mL. The treatment of HMEC-1 cells with unloaded endothelium consists of a monolayer of endothelial cells, lines
PVP-NPs (6000 Da) and PVP-IMC did not lead to statistically the cavities of arteries, veins, and capillaries, and is in direct
significant changes as compared to control (p = NS). As shown contact with the blood components. The endothelium is not
in Figure 11, indomethacin exerts growth inhibitory and toxic only just a barrier between blood and tissues but also the
effects on endothelial cells at higher concentrations (p ≤ largest endocrine gland.32 Indeed, the endothelial cells are
0.001). Previously, indomethacin was shown to downregulate involved in the control of thrombosis and thrombolysis,
the migration of cancer cells due to dysregulation of the Ca determine the dilation and contraction of blood vessels, and
metabolism32 and to induce injury of endothelial cells by participate in the interactions of platelets and leukocytes with
disturbing mitochondria metabolism,33 well following the the vessel wall.39−41 Therefore, it is of utmost importance to
results of the present study. Treating the cells with NPs exclude possible deleterious effects of NPs on endothelial cells
loaded with indomethacin at the low concentration (0.01 mg/ homeostasis.
mL) likewise did not affect their growth. These results show In our previous study, we determined that the NPs
that the PVP (6000 Da) carrier, in combination with comprising amphiphilic-PVP NPs (2000, 4000, 6000, 8000,
indomethacin, did not affect HMEC-1 cell growth/viability and 12000 Da) did not significantly affect the growth of
suggesting that the Amp-PVP carrier can in a biocompatible HMEC-1 endothelial cells.10 In this study, we evaluated the
manner transport the drug to tissue parenchyma. effect of the amphiphilic-PVP (6000 Da) NPs on basal and
The effect of indomethacin, indomethacin-loaded PVP-NPs activated endothelial cells’ viability and immunological
(6000 Da) (PVP-IMC), and unloaded PVP-NPs (6000 Da) on activation. To examine the interaction of amphiphilic-PVP-
HMEC-1 adhesion molecules was in continuation studied. based polymeric materials with endothelial cells, we synthe-
These cells were exposed to respective treatments at a sized PVP (6000 Da), F-NPs where the FITC molecules were
concentration of 0.01 mg/mL during 48 h, at which point incorporated into the PVP (6000 Da) hydrophobic cores, and
the cells were harvested. Cell extracts were probed with E- PVP (6000 Da) loaded with the drug indomethacin.
4218 https://dx.doi.org/10.1021/acs.molpharmaceut.0c00667
Mol. Pharmaceutics 2020, 17, 4212−4225
Molecular Pharmaceutics pubs.acs.org/molecularpharmaceutics Article

Figure 8. Effect of PVP-NPs (6000 Da) and F-NPs on HMEC-1 cells’ adhesion molecule expression. (A) Activation of ICAM-1 expression by
increasing concentrations of LPS. (B) Effect of F- and PVP-NPs (6000 Da) on E-selectin expression of basal state and LPS-activated HMEC-1 cells.
(C) Effect of F- and PVP-NPs (6000 Da) on the expression of the ICAM-1 of basal state and LPS-activated HMEC-1 cells. C = control; cells
incubated with medium only. The position of the nearest respective protein marker band is depicted to the right. The results represent the average
of three separate experiments. Statistical significance: ** = p ≤ 0.01 compared to control.

Notably, some NPs exert cytotoxic effects on endothelial micelles caused low or modest toxic effects on human
cells. Thus, metal-based NPs, including ZnO or TiO2 NPs42−44 umbilical vein endothelial cells (HUVEC).48
and silica-based NPs, were shown to cause oxidative stress and One of the critical applications of nanomaterials is the
endothelial dysfunction, in vitro, partly through the activation targeted transfer of drugs. Therefore, the possible effects of the
of the MAPK/Nrf2 pathway and NF-κβ signaling.43,45 drug-carrier complex on endothelial cell viability need to be
Intravenous administration of silver NPs, in in vivo models, taken into consideration. In previous studies, we showed that
induced organ toxicity attributed to the disturbance of drugs of high hydrophobicity, such as indomethacin, could be
interendothelial junctions due to the NP-dependent increase efficiently encapsulated, during the self-assembly process, to
the hydrophobic core of amphiphilic polymeric NPs.5
of radical oxygen species production.46 In a previous study, we
Indomethacin is a nonsteroidal anti-inflammatory drug, a
showed that Amph-PVP NPs did not affect HMEC-1
derivative of indole, which also has analgesic and antipyretic
endothelial cell growth.10 In the present study, we verified properties. Its anti-inflammatory activity is due to the
their safety, demonstrating that PVP (OD6000) NPs do not inhibition of prostaglandin synthesis and possibly other
cause toxic effects up to 66 μg/mL. Previous studies likewise actions, such as phosphodiesterase inhibition and leukocyte
showed that micelle-based NPs exhibit low toxicity concerning migration.49,50 In this study to PVP-NPs (6000 Da), the model
endothelial cells. Indeed, polymeric micelles, such as methoxy drug, indomethacin, was incorporated, and the effects of the
poly(ethylene glycol)-poly(D,L-lactide) (MPEG-PLA)-based complex were examined. No adverse effect of the complex on
micelles, exerted no toxic effects on endothelial cells viability endothelial cell viability was observed. Previously, Cu(II)
up to concentrations of 200 μg/mL.47,48 Diglutamic acid-based complex-loaded solid lipid NPs were shown to exert in vitro
4219 https://dx.doi.org/10.1021/acs.molpharmaceut.0c00667
Mol. Pharmaceutics 2020, 17, 4212−4225
Molecular Pharmaceutics pubs.acs.org/molecularpharmaceutics Article

Figure 9. Detection of PVP-NPs (6000 Da) uptake by living HMEC-1 cells. (A) The uptake of F-NPs by living HMEC-1 cells was assessed using a
fluorescence microscope (green; FITC). (B) The uptake of F-NPs was evaluated after challenging HMEC-1 cells with LPS for 2 h (green; FITC).
C = control; cells incubated with medium only. Representative images are presented - magnification ×10.

cytotoxic effects on MCF-7 breast cancer cells and had toxic Specifically, the overexpression of adhesion molecules,
effects on HUVEC control cells.51 including E-selectin, ICAM-1, and VCAM-1, initiates the
Also, endothelium plays a prominent role in the inflamma- recruitment of monocytes, which are, under these conditions,
tory response. The phenomenon through which the able to attach steadily onto the activated endothelial cells.26,31
endothelium undergoes modifications upon challenge is A cascade is generated where monocytes differentiate into
denominated as endothelial activation.52 In anaphylaxis or macrophages and transform into macrophage foam cells upon
sepsis, there is a disintegration of intercellular contacts in engulfing excessive lipids from the plasma.56 The resulting
postcapillary venules under the conditions of an inflammatory agglomeration of macrophage foam cells into the arterial wall is
response, resulting in intercellular gap formation edema.53,54 a hallmark of atherosclerosis.57 The exposure of endothelial
Endothelial cells have a crucial role in the regulation of blood cells to different classes of NPs, e.g., metal-based,58,44
vessel tone, monocytes recruitment, and thrombogenicity so carbonaceous NPs,59,60 and silica NPs,43 enhanced the release
that dysfunction of endothelial cells is directly correlated to the of inflammatory factors and increased the expression of
pathogenesis of cardiovascular diseases including atheroscle- adhesion molecules (e.g., ICAM-1, VCAM-1, and selectins).
rosis.26 The expression of specific adhesion molecules, e.g., In the present study, PVP OD6000 NPs did not affect the
VCAM-1, ICAM-1, and selectins, is increased upon endothe- expression of adhesion molecules, including E-selectin, ICAM-
lium activation facilitating, thus, the binding and the 1, and VCAM-1, up to the high concentration of 66 μg/mL.
penetration of endothelium barrier by immune cells as well Likewise, no adverse effects on the expression of adhesion
as their subsequent transmigration to tissue parenchyma.55 receptors were evident when endothelial cells were treated
4220 https://dx.doi.org/10.1021/acs.molpharmaceut.0c00667
Mol. Pharmaceutics 2020, 17, 4212−4225
Molecular Pharmaceutics pubs.acs.org/molecularpharmaceutics Article

Figure 10. Intracellular localization of PVP-NPs (6000 Da). (A, Β) HMEC-1 cell treated with 0.066 mg/mL of F-NPs (FITC; green stain). (C)
Cells stained with TOPRO, without NP treatments. Representative images are presented - magnification ×60 and zoom 4.

mimic pathological states.61,62 It is well established that LPS,


when in contact with endothelial cells, interacts with the TLR4
receptors and induces their activation.27 A critical downstream
mediator of TLR4 is NF-κβ. Binding of LPS to TLR4 triggers
the activation of phosphokinases, which phosphorylate the
cytoplasmic NF-κβ. Upon activation, NF-κβ translocates to the
nucleus, where it exerts transcriptional regulation of inflam-
matory mediators.28 Therefore, the ratio p-NF-κβ/total-NF-κβ
can be used as a reliable indicator of cellular immune
activation.63−65
Generating a model of activated endothelium demonstrated
that PVP NPs (6000 Da), F-NPs, and loaded with
indomethacin PVP-NPs (6000 Da) did not exert adverse
Figure 11. Effect of indomethacin, indomethacin-loaded PVP (6000 effects on activated HMEC-1 cell viability. Furthermore,
Da), and unloaded PVP-NPs (6000 Da) on the proliferative ability of neither PVP NPs nor the drug-carrier complex affected the
HMEC-1 cells. Cells were cultured in 96-well plates and treated with
respective agents 48 h. C = control; cells incubated with medium only. expression of adhesion receptors, including E-selectin, ICAM-
The results represent the average of three separate experiments. 1, and VCAM-1. In previous studies, it was shown that ZnO-
Statistical significance: *** = p ≤ 0.001 compared to control. based NPs facilitated the TNFα-dependent increase in ICAM-
1 expression in HUVECs66 but did not induce synergistic
effects in the presence of LPS of palmitate and NPs on
with PVP OD6000 NPs loaded with indomethacin, demon-
strating an absence of synergistic action. endothelial cell inflammatory responses.67 Moreover, both Ag
Under various pathological conditions, the activation of the and silica NPs induced cytotoxicity and inflammatory response
endothelium is imminent; indeed, dysfunctional endothelium of HUVEC cells under shear stress.68,69 Furthermore, repeated
is a hallmark of pathologies such as diabetes or cardiovascular exposure to rutile TiO2 NPs induced enhancement in plaque
disease.26,31 Therefore, to anticipate the endothelial cell progression in atherosclerotic mice, correlated with inflamma-
response in vivo, it is vital to evaluate the biocompatibility of tion and oxidative stress.70 NPs based on chitosan derivatives
various nanomaterials with endothelium under conditions that (Ch-der) or poly(lactic-co-glycolic acid) (PLGA), encapsulated
4221 https://dx.doi.org/10.1021/acs.molpharmaceut.0c00667
Mol. Pharmaceutics 2020, 17, 4212−4225
Molecular Pharmaceutics pubs.acs.org/molecularpharmaceutics Article

Figure 12. Effect of indomethacin (IMC), indomethacin-loaded PVP-NPs (PVP-IMC), and unloaded PVP-NPs (6000 Da) (PVP) on protein
expression of HMEC-1 cells’ E-selectin, ICAM-1, and VCAM-1 adhesion molecules. The cells were treated with different agents for 48 h. (A) Effect
of indomethacin, PVP-IMC NPs, and hollow PVP-NPs (6000 Da) on E-selectin protein expression in HMEC-1 cells. (B) Effect of indomethacin,
PVP-IMC NPs, and hollow PVP-NPs (6000 Da) on the expression of ICM-1 protein in HMEC-1 cells. (C) Effect of indomethacin, PVP-IMC
NPs, and hollow PVP-NPs (6000 Da) on the expression of VCAM-1 protein in HMEC-1 cells. The results were normalized to Actin. The position
of the nearest respective protein marker band is depicted to the right. C = control; cells incubated with medium only. The results represent the
average of three separate experiments. Statistical significance: NS = not significant.

with natural cherry extract, exhibited a protective effect on well established that the activated endothelium undergoes
LPS-stressed HUVEC cells.71 structural and functional changes, which, among others, result
The F-type (FITC included to the NP core) particles in the leakage of intravascular components.72,73 PVP NPs were
demonstrated efficient uptake and no deleterious effects on safe for use in an activated endothelium model, as no further
HMEC-1 cell viability and immunological activation. Fluo- changes in the upregulation of adhesion molecules were
rescent NPs are especially suitable for living cells and may be
evident. However, activated HMEC-1 endothelial cells
utilized for imaging of cellular uptake and intracellular
localization as well as tracking of diffusion processes in the exhibited an increased uptake of PVP NPs. Previous studies
extracellular space. The synthesis of fluorescent NPs have shown that conditions of shear stress can enhance the
demonstrated that FITC-labeled PVP NPs are nontoxic and HUVECs uptake of silica NPs.74 Moreover, the uptake of
noninflammatory and that PVP NPs (6000 Da) are uptaken by poly[acrylonitrile-co-(N-vinylpyrrolidone)] [P(AN-co-NVP)]
endothelial cells in a concentration-dependent manner. It is NPs by HUVECs was facilitated by IL-1β treatment. In
4222 https://dx.doi.org/10.1021/acs.molpharmaceut.0c00667
Mol. Pharmaceutics 2020, 17, 4212−4225
Molecular Pharmaceutics pubs.acs.org/molecularpharmaceutics Article

contrast, a modest increase in Au NPs uptake was shown by Mikhail M. Shtilman − Department of Biomaterials, D.
endothelial cells treated with TNFα.75 Mendeleev University of Chemical Technology of Russia,
Moscow 125047, Russian Federation
5. CONCLUSIONS George N. Tzanakakis − Laboratory of Histology-Embryology,
In this study, we demonstrated for the first time that PVP-NPs School of Medicine and Laboratory of Anatomy, School of
do not affect the viability and the immune response of the Medicine, University of Crete, 71003 Heraklion, Greece
basal state and the challenged LPS endothelial cells. The Aristidis Tsatsakis − Laboratory of Toxicology, School of
utilization of fluorescently labeled PVP-NPs showed that the Medicine, University of Crete, 71003 Heraklion, Greece;
uptake of PVP-NPs is increased by activated HMEK-1 Department of Biomaterials, D. Mendeleev University of
endothelial cells. In summary, PVP-NPs do not exert adverse Chemical Technology of Russia, Moscow 125047, Russian
effects of either basal state or challenged endothelial cells. The Federation
interactions of NP with the endothelium should be considered Complete contact information is available at:
when designing nanomaterials. In vitro assays are useful to https://pubs.acs.org/10.1021/acs.molpharmaceut.0c00667
predict the better response of the endothelium to NPs in vivo.
Amph-PVP NPs are a promising drug delivery system and Notes
could be utilized for specialized targeting of the desired tissues The authors declare no competing financial interest.
while reducing the side effects in patients.

■ ASSOCIATED CONTENT
■ ACKNOWLEDGMENTS
This work was financially supported by the Ministry of Science
*
sı Supporting Information and Higher Education of the Russian Federation within the
The Supporting Information is available free of charge at framework of the state assignment under the project FSSM-
h t t p s : / / p u b s . a c s . o r g / d o i / 10 . 1 02 1 / a c s .m o l p h a rm a - 2020-0004 and by a grant from Era Net.RUS. Plus, initiative
ceut.0c00667. (“NABUCO”, ID#169). The work of A.N.K. was supported by
D. Mendeleev University of Chemical Technology of Russia
Supplementary Figure S1, 1H NMR spectra; Supple- (Project Number K-2020-018).


mentary Figure S2, differential scanning calorimetry
(DSC); and Supplementary Figure S3, thermogravimet-
ABBREVIATIONS
ric analysis (TGA) (PDF)


NPs, nanoparticles; PVP, poly-N-vinylpyrrolidone; Amph-
PVP, amphiphilic poly-N-vinylpyrrolidone derivative consisting
AUTHOR INFORMATION of water-soluble polymer fragment with molecular weight 6000
Corresponding Author Da and one terminal n-octadecyl hydrophobic group; F, PVP-
Dragana Nikitovic − Laboratory of Histology-Embryology, NPs (6000 Da) linked with fluorescent FITC; ICAM-1,
School of Medicine, University of Crete, 71003 Heraklion, intercellular adhesion molecule-1; VCAM-1, vascular cell
Greece; orcid.org/0000-0003-3882-7399; adhesion molecule-1; ECM, extracellular matrix; LPS, lip-
Email: nikitovic@uoc.gr opolysaccharides

Authors
Aikaterini Berdiaki − Laboratory of Histology-Embryology,
■ REFERENCES
(1) Su, S.; Kang, P. M. Systemic Review of Biodegradable
School of Medicine, University of Crete, 71003 Heraklion, Nanomaterials in Nanomedicine. Nanomaterials 2020, 10 (4), 656.
Greece (2) De Matteis, V.; Rizzello, L. Noble Metals and Soft Bio-Inspired
Emmanouela Perisynaki − Laboratory of Histology- Nanoparticles in Retinal Diseases Treatment: A Perspective. Cells
Embryology, School of Medicine, University of Crete, 71003 2020, 9 (3), 679.
Heraklion, Greece (3) Henrich-Noack, P.; Nikitovic, D.; Neagu, M.; Docea, A. O.;
Antonios Stratidakis − Institute for Advanced Study (IUSS), Engin, A. B.; Gelperina, S.; Shtilman, M.; Mitsias, P.; Tzanakakis, G.;
Environmental Health Engineering, 27100 Pavia, Italy; Gozes, I.; Tsatsakis, A. The blood-brain barrier and beyond: Nano-
Laboratory of Toxicology, School of Medicine, University of based neuropharmacology and the role of extracellular matrix.
Nanomedicine 2019, 17, 359−379.
Crete, 71003 Heraklion, Greece (4) Taghizadehghalehjoughi, A.; Hacimuftuoglu, A.; Cetin, M.;
Pavel P. Kulikov − Department of Biomaterials, D. Mendeleev Ugur, A. B.; Galateanu, B.; Mezhuev, Y.; Okkay, U.; Taspinar, N.;
University of Chemical Technology of Russia, Moscow 125047, Taspinar, M.; Uyanik, A.; Gundogdu, B.; Mohammadzadeh, M.;
Russian Federation; Centre for Strategic Planning of FMBA of Nalci, K. A.; Stivaktakis, P.; Tsatsakis, A.; Jung, T. W.; Jeong, J. H.;
Russia, Moscow 119121, Russia Abd El-Aty, A. M. Effect of metformin/irinotecan-loaded poly-lactic-
Andrey N. Kuskov − Department of Biomaterials, D. Mendeleev co-glycolic acid nanoparticles on glioblastoma: in vitro and in vivo
University of Chemical Technology of Russia, Moscow 125047, studies. Nanomedicine (London, U. K.) 2018, 13 (13), 1595−1606.
Russian Federation; orcid.org/0000-0001-8140-2754 (5) Kuskov, A. N.; Kulikov, P. P.; Goryachaya, A. V.; Tzatzarakis, M.
Polychronis Stivaktakis − Laboratory of Toxicology, School of N.; Tsatsakis, A. M.; Velonia, K.; Shtilman, M. I. Self-assembled
Medicine, University of Crete, 71003 Heraklion, Greece amphiphilic poly-N-vinylpyrrolidone nanoparticles as carriers for
hydrophobic drugs: Stability aspects. J. Appl. Polym. Sci. 2018, 135,
Petra Henrich-Noack − Clinic of Neurology with Institute of 45637.
Translational Neurology, University Clinic Muenster, Muenster, (6) Basyreva, L. Y.; Voinova, E. V.; Gusev, A. A.; Mikhalchik, E. V.;
Germany Kuskov, A. N.; Goryachaya, A. V.; Gusev, S. A.; Shtilman, M. I.;
Anna L. Luss − Department of Biomaterials, D. Mendeleev Velonia, K.; Tsatsakis, A. M. Fluorouracil neutrophil extracellular
University of Chemical Technology of Russia, Moscow 125047, traps formation inhibited by polymer nanoparticle shielding. Mater.
Russian Federation Sci. Eng., C 2020, 108, 110382.

4223 https://dx.doi.org/10.1021/acs.molpharmaceut.0c00667
Mol. Pharmaceutics 2020, 17, 4212−4225
Molecular Pharmaceutics pubs.acs.org/molecularpharmaceutics Article

(7) Villemson, A. L.; Kuskov, A. N.; Shtilman, M. I.; Galebskaya, L. (27) Opal, S. M.; van der Poll, T. Endothelial barrier dysfunction in
V.; Ryumina, E. V.; Larionova, N. I. Interaction of polymer aggregates septic shock. J. Intern. Med. 2015, 277 (3), 277−293.
based on stearoyl-poly-N-vinylpyrrolidone with blood components. (28) Hoesel, B.; Schmid, J. A. The complexity of NF-kappaB
Biochemistry 2004, 69 (6), 621−8. signaling in inflammation and cancer. Mol. Cancer 2013, 12, 86.
(8) Luss, A. L.; Kulikov, P. P.; Romme, S. B.; Andersen, C. L.; (29) Deng, H. F.; Wang, S.; Li, L.; Zhou, Q.; Guo, W. B.; Wang, X.
Pennisi, C. P.; Docea, A. O.; Kuskov, A. N.; Velonia, K.; Mezhuev, Y. L.; Liu, M. D.; Liu, K.; Xiao, X. Z. Puerarin prevents vascular
O.; Shtilman, M. I.; Tsatsakis, A. M.; Gurevich, L. Nanosized carriers endothelial injury through suppression of NF-kappaB activation in
based on amphiphilic poly-N-vinyl-2-pyrrolidone for intranuclear drug LPS-challenged human umbilical vein endothelial cells. Biomed.
delivery. Nanomedicine (London, U. K.) 2018, 13 (7), 703−715. Pharmacother. 2018, 104, 261−267.
(9) Kuskov, A. N.; Kulikov, P. P.; Shtilman, M. I.; Rakitskii, V. N.; (30) Yu, S.; Chen, X.; Xiu, M.; He, F.; Xing, J.; Min, D.; Guo, F. The
Tsatsakis, A. M. Amphiphilic poly-N-vynilpyrrolidone nanoparticles: regulation of Jmjd3 upon the expression of NF-kappaB downstream
Cytotoxicity and acute toxicity study. Food Chem. Toxicol. 2016, 96, inflammatory genes in LPS activated vascular endothelial cells.
273−9. Biochem. Biophys. Res. Commun. 2017, 485 (1), 62−68.
(10) Tsatsakis, A.; Stratidakis, A. K.; Goryachaya, A. V.; Tzatzarakis, (31) Gimbrone, M. A., Jr.; Garcia-Cardena, G. Endothelial Cell
M. N.; Stivaktakis, P. D.; Docea, A. O.; Berdiaki, A.; Nikitovic, D.; Dysfunction and the Pathobiology of Atherosclerosis. Circ. Res. 2016,
Velonia, K.; Shtilman, M. I.; Rizos, A. K.; Kuskov, A. N. In vitro blood 118 (4), 620−36.
compatibility and in vitro cytotoxicity of amphiphilic poly-N- (32) Guo, Y. C.; Chang, C. M.; Hsu, W. L.; Chiu, S. J.; Tsai, Y. T.;
vinylpyrrolidone nanoparticles. Food Chem. Toxicol. 2019, 127, 42− Chou, Y. H.; Hou, M. F.; Wang, J. Y.; Lee, M. H.; Tsai, K. L.; Chang,
52. W. C. Indomethacin inhibits cancer cell migration via attenuation of
(11) Giodini, L.; Re, F. L.; Campagnol, D.; Marangon, E.; Posocco, cellular calcium mobilization. Molecules 2013, 18 (6), 6584−96.
B.; Dreussi, E.; Toffoli, G. Nanocarriers in cancer clinical practice: a (33) Chan, J. M.; Valencia, P. M.; Zhang, L.; Langer, R.; Farokhzad,
pharmacokinetic issue. Nanomedicine 2017, 13 (2), 583−599. O. C. Polymeric nanoparticles for drug delivery. Methods Mol. Biol.
(12) Li, M.; Zou, P.; Tyner, K.; Lee, S. Physiologically Based 2010, 624, 163−75.
Pharmacokinetic (PBPK) Modeling of Pharmaceutical Nanoparticles. (34) Mioc, M.; Pavel, I. Z.; Ghiulai, R.; Coricovac, D. E.; Farcas, C.;
AAPS J. 2017, 19 (1), 26−42. Mihali, C. V.; Oprean, C.; Serafim, V.; Popovici, R. A.; Dehelean, C.
(13) Engin, A. B.; Nikitovic, D.; Neagu, M.; Henrich-Noack, P.; A.; Shtilman, M. I.; Tsatsakis, A. M.; Soica, C. The Cytotoxic Effects
Docea, A. O.; Shtilman, M. I.; Golokhvast, K.; Tsatsakis, A. M. of Betulin-Conjugated Gold Nanoparticles as Stable Formulations in
Mechanistic understanding of nanoparticles’ interactions with Normal and Melanoma Cells. Front. Pharmacol. 2018, 9, 429.
extracellular matrix: the cell and immune system. Part. Fibre Toxicol. (35) Gulati, N. M.; Stewart, P. L.; Steinmetz, N. F. Bioinspired
2017, 14 (1), 22. Shielding Strategies for Nanoparticle Drug Delivery Applications. Mol.
(14) Furuse, M. Molecular basis of the core structure of tight Pharmaceutics 2018, 15 (8), 2900−2909.
junctions. Cold Spring Harbor Perspect. Biol. 2010, 2 (1), No. a002907. (36) Sztandera, K.; Gorzkiewicz, M.; Klajnert-Maculewicz, B. Gold
(15) Bostan, H. B.; Rezaee, R.; Valokala, M. G.; Tsarouhas, K.;
Nanoparticles in Cancer Treatment. Mol. Pharmaceutics 2019, 16 (1),
Golokhvast, K.; Tsatsakis, A. M.; Karimi, G. Cardiotoxicity of nano-
1−23.
particles. Life Sci. 2016, 165, 91−99.
(37) Dhawan, A.; Sharma, V. Toxicity assessment of nanomaterials:
(16) Neagu, M.; Piperigkou, Z.; Karamanou, K.; Engin, A. B.; Docea,
methods and challenges. Anal. Bioanal. Chem. 2010, 398 (2), 589−
A. O.; Constantin, C.; Negrei, C.; Nikitovic, D.; Tsatsakis, A. Protein
605.
bio-corona: critical issue in immune nanotoxicology. Arch. Toxicol.
(38) Jones, C. F.; Grainger, D. W. In vitro assessments of
2017, 91 (3), 1031−1048.
nanomaterial toxicity. Adv. Drug Delivery Rev. 2009, 61 (6), 438−56.
(17) Chiba, H.; Osanai, M.; Murata, M.; Kojima, T.; Sawada, N.
(39) Kruger-Genge, A.; Blocki, A.; Franke, R. P.; Jung, F. Vascular
Transmembrane proteins of tight junctions. Biochim. Biophys. Acta,
Biomembr. 2008, 1778 (3), 588−600. Endothelial Cell Biology: An Update. Int. J. Mol. Sci. 2019, 20 (18),
(18) Mehta, D.; Malik, A. B. Signaling mechanisms regulating 4411.
endothelial permeability. Physiol. Rev. 2006, 86 (1), 279−367. (40) Laroia, S. T.; Ganti, A. K.; Laroia, A. T.; Tendulkar, K. K.
(19) Paulsson, M. Basement membrane proteins: structure, Endothelium and the lipid metabolism: the current understanding.
assembly, and cellular interactions. Crit. Rev. Biochem. Mol. Biol. Int. J. Cardiol. 2003, 88 (1), 1−9.
1992, 27 (1−2), 93−127. (41) Dora, K. A. Endothelial-smooth muscle cell interactions in the
(20) Yurchenco, P. D. Basement membranes: cell scaffoldings and regulation of vascular tone in skeletal muscle. Microcirculation 2016,
signaling platforms. Cold Spring Harbor Perspect. Biol. 2011, 3 (2), 23 (8), 626−630.
a004911. (42) Gu, Y.; Cheng, S.; Chen, G.; Shen, Y.; Li, X.; Jiang, Q.; Li, J.;
(21) Gimbrone, M. A., Jr.; Nagel, T.; Topper, J. N. Biomechanical Cao, Y. The effects of endoplasmic reticulum stress inducer
activation: an emerging paradigm in endothelial adhesion biology. J. thapsigargin on the toxicity of ZnO or TiO2 nanoparticles to
Clin. Invest. 1997, 99 (8), 1809−1813. human endothelial cells. Toxicol. Mech. Methods 2017, 27 (3), 191−
(22) Gimbrone, M. A., Jr.; Nagel, T.; Topper, J. N. Biomechanical 200.
activation: an emerging paradigm in endothelial adhesion biology. J. (43) Guo, C.; Xia, Y.; Niu, P.; Jiang, L.; Duan, J.; Yu, Y.; Zhou, X.;
Clin. Invest. 1997, 99 (8), 1809−13. Li, Y.; Sun, Z. Silica nanoparticles induce oxidative stress,
(23) Piga, R.; Naito, Y.; Kokura, S.; Handa, O.; Yoshikawa, T. Short- inflammation, and endothelial dysfunction in vitro via activation of
term high glucose exposure induces monocyte-endothelial cells the MAPK/Nrf2 pathway and nuclear factor-kappaB signaling. Int. J.
adhesion and transmigration by increasing VCAM-1 and MCP-1 Nanomed. 2015, 10, 1463−77.
expression in human aortic endothelial cells. Atherosclerosis 2007, 193 (44) Danielsen, P. H.; Cao, Y.; Roursgaard, M.; Møller, P.; Loft, S.
(2), 328−34. Endothelial cell activation, oxidative stress and inflammation induced
(24) Timmerman, I.; Daniel, A. E.; Kroon, J.; van Buul, J. D. by a panel of metal-based nanomaterials. Nanotoxicology 2015, 9 (7),
Leukocytes Crossing the Endothelium: A Matter of Communication. 813−24.
Int. Rev. Cell Mol. Biol. 2016, 322, 281−329. (45) Guo, H.; Zhang, J.; Boudreau, M.; Meng, J.; Yin, J. J.; Liu, J.;
(25) Chen, J.; Lopez, J. A. Interactions of platelets with Xu, H. Intravenous administration of silver nanoparticles causes organ
subendothelium and endothelium. Microcirculation 2005, 12 (3), toxicity through intracellular ROS-related loss of inter-endothelial
235−46. junction. Part. Fibre Toxicol. 2015, 13, 21.
(26) Libby, P. Inflammation in atherosclerosis. Arterioscler., Thromb., (46) Guo, Y. Y.; Zhang, J.; Zheng, Y. F.; Yang, J.; Zhu, X. Q.
Vasc. Biol. 2012, 32 (9), 2045−51. Cytotoxic and genotoxic effects of multi-wall carbon nanotubes on

4224 https://dx.doi.org/10.1021/acs.molpharmaceut.0c00667
Mol. Pharmaceutics 2020, 17, 4212−4225
Molecular Pharmaceutics pubs.acs.org/molecularpharmaceutics Article

human umbilical vein endothelial cells in vitro. Mutat. Res., Genet. cells: interaction with palmitate or lipopolysaccharide. J. Appl. Toxicol.
Toxicol. Environ. Mutagen. 2011, 721 (2), 184−91. 2017, 37 (8), 895−901.
(47) Liu, F.; Huang, H.; Gong, Y.; Li, J.; Zhang, X.; Cao, Y. (66) Ucciferri, N.; Collnot, E. M.; Gaiser, B. K.; Tirella, A.; Stone,
Evaluation of in vitro toxicity of polymeric micelles to human V.; Domenici, C.; Lehr, C. M.; Ahluwalia, A. In vitro toxicological
endothelial cells under different conditions. Chem.-Biol. Interact. 2017, screening of nanoparticles on primary human endothelial cells and the
263, 46−54. role of flow in modulating cell response. Nanotoxicology 2014, 8 (6),
(48) Menard, N.; Tsapis, N.; Poirier, C.; Arnauld, T.; Moine, L.; 697−708.
Gignoux, C.; Lefoulon, F.; Pean, J. M.; Fattal, E. Novel surfactants (67) Kim, D.; Lin, Y. S.; Haynes, C. L. On-chip evaluation of shear
with diglutamic acid polar head group: drug solubilization and toxicity stress effect on cytotoxicity of mesoporous silica nanoparticles. Anal.
studies. Pharm. Res. 2012, 29 (7), 1882−96. Chem. 2011, 83 (22), 8377−82.
(49) Van Kolfschoten, A. A.; Hagelen, F.; Van Noordwijk, J. (68) Mikkelsen, L.; Sheykhzade, M.; Jensen, K. A.; Saber, A. T.;
Indomethacin and paracetamol: interaction with prostaglandin Jacobsen, N. R.; Vogel, U.; Wallin, H.; Loft, S.; Møller, P. Modest
synthesis in the rat stomach. Eur. J. Pharmacol. 1982, 84 (1−2), effect on plaque progression and vasodilatory function in athero-
123−5. sclerosis-prone mice exposed to nanosizedTiO(2). Part. Fibre Toxicol.
(50) Laddha, S. S.; Bhatnagar, S. P. A new therapeutic approach in 2011, 8, 32.
Parkinson’s disease: some novel quinazoline derivatives as dual (69) Beconcini, D.; Felice, F.; Zambito, Y.; Fabiano, A.; Piras, A. M.;
selective phosphodiesterase 1 inhibitors and anti-inflammatory agents. Macedo, M. H.; Sarmento, B.; Di Stefano, R. Anti-Inflammatory
Bioorg. Med. Chem. 2009, 17 (19), 6796−802. Effect of Cherry Extract Loaded in Polymeric Nanoparticles:
(51) Kani, I.; Dikmen, G.; Eskiler, G. G.; Cecener, G.; Tunca, B.; Relevance of Particle Internalization in Endothelial Cells. Pharma-
Egeli, U. The Anticancer Activity of Complex [Cu2(mu-(C6H5)- ceutics 2019, 11 (10), 500.
2CHCOO)3(bipy)2)] (ClO4) -Solid Lipid Nanoparticles on MCF-7 (70) Aird, W. C. The role of the endothelium in severe sepsis and
Cells. Curr. Drug Delivery 2016, 13 (8), 1339−1350. multiple organ dysfunction syndrome. Blood 2003, 101 (10), 3765−
(52) Radeva, M. Y.; Waschke, J. Mind the gap: mechanisms 77.
regulating the endothelial barrier. Acta physiologica 2018, 222 (1), (71) Ince, C.; Mayeux, P. R.; Nguyen, T.; Gomez, H.; Kellum, J. A.;
e12860. Ospina-Tascon, G. A.; Hernandez, G.; Murray, P.; De Backer, D.;
(53) Nadar, S.; Blann, A. D.; Lip, G. Y. Endothelial dysfunction: Workgroup, A. X. The Endothelium in Sepsis. Shock 2016, 45 (3),
methods of assessment and application to hypertension. Curr. Pharm. 259−70.
Des. 2004, 10 (29), 3591−605. (72) Samuel, S. P.; Jain, N.; O’Dowd, F.; Paul, T.; Kashanin, D.;
(54) Filippi, M. D. Mechanism of Diapedesis: Importance of the Gerard, V. A.; Gun’ko, Y. K.; Prina-Mello, A.; Volkov, Y.
Transcellular Route. Adv. Immunol. 2016, 129, 25−53. Multifactorial determinants that govern nanoparticle uptake by
(55) Tuttolomondo, A.; Di Raimondo, D.; Pecoraro, R.; Arnao, V.; human endothelial cells under flow. Int. J. Nanomed. 2012, 7,
Pinto, A.; Licata, G. Atherosclerosis as an inflammatory disease. Curr. 2943−56.
Pharm. Des. 2012, 18 (28), 4266−88. (73) Freese, C.; Gibson, M. I.; Klok, H. A.; Unger, R. E.; Kirkpatrick,
(56) Moore, K. J.; Sheedy, F. J.; Fisher, E. A. Macrophages in C. J. Size- and coating-dependent uptake of polymer-coated gold
atherosclerosis: a dynamic balance. Nat. Rev. Immunol. 2013, 13 (10), nanoparticles in primary human dermal microvascular endothelial
709−21. cells. Biomacromolecules 2012, 13 (5), 1533−43.
(57) Tabas, I.; Bornfeldt, K. E. Macrophage Phenotype and Function
in Different Stages of Atherosclerosis. Circ. Res. 2016, 118 (4), 653−
67.
(58) Suzuki, Y.; Tada-Oikawa, S.; Ichihara, G.; Yabata, M.; Izuoka,
K.; Suzuki, M.; Sakai, K.; Ichihara, S. Zinc oxide nanoparticles induce
migration and adhesion of monocytes to endothelial cells and
accelerate foam cell formation. Toxicol. Appl. Pharmacol. 2014, 278
(1), 16−25.
(59) Cao, Y.; Roursgaard, M.; Danielsen, P. H.; Møller, P.; Loft, S.
Carbon black nanoparticles promote endothelial activation and lipid
accumulation in macrophages independently of intracellular ROS
production. PLoS One 2014, 9 (9), No. e106711.
(60) Cao, Y.; Roursgaard, M.; Jacobsen, N. R.; Møller, P.; Loft, S.
Monocyte adhesion induced by multi-walled carbon nanotubes and
palmitic acid in endothelial cells and alveolar-endothelial co-cultures.
Nanotoxicology 2016, 10 (2), 235−44.
(61) Setyawati, M. I.; Tay, C. Y.; Docter, D.; Stauber, R. H.; Leong,
D. T. Understanding and exploiting nanoparticles’ intimacy with the
blood vessel and blood. Chem. Soc. Rev. 2015, 44 (22), 8174−99.
(62) Cao, Y.; Gong, Y.; Liu, L.; Zhou, Y.; Fang, X.; Zhang, C.; Li, Y.;
Li, J. The use of human umbilical vein endothelial cells (HUVECs) as
an in vitro model to assess the toxicity of nanoparticles to
endothelium: a review. J. Appl. Toxicol. 2017, 37 (12), 1359−1369.
(63) Zhang, J. L.; Xu, Y.; Shen, J. Cordycepin inhibits lip-
opolysaccharide (LPS)-induced tumor necrosis factor (TNF)-alpha
production via activating amp-activated protein kinase (AMPK)
signaling. Int. J. Mol. Sci. 2014, 15 (7), 12119−34.
(64) Tsou, T. C.; Yeh, S. C.; Tsai, F. Y.; Lin, H. J.; Cheng, T. J.;
Chao, H. R.; Tai, L. A. Zinc oxide particles induce inflammatory
responses in vascular endothelial cells via NF-kappaB signaling. J.
Hazard. Mater. 2010, 183 (1−3), 182−8.
(65) Gong, Y.; Ji, Y.; Liu, F.; Li, J.; Cao, Y. Cytotoxicity, oxidative
stress and inflammation induced by ZnO nanoparticles in endothelial

4225 https://dx.doi.org/10.1021/acs.molpharmaceut.0c00667
Mol. Pharmaceutics 2020, 17, 4212−4225

You might also like