• Sonuç bulunamadı

Formation of the inclusion complex of water soluble fluorescent calix[4]arene and naringenin: solubility, cytotoxic effect and molecular modeling studies

N/A
N/A
Protected

Academic year: 2021

Share "Formation of the inclusion complex of water soluble fluorescent calix[4]arene and naringenin: solubility, cytotoxic effect and molecular modeling studies"

Copied!
14
0
0

Yükleniyor.... (view fulltext now)

Tam metin

(1)

Full Terms & Conditions of access and use can be found at

https://www.tandfonline.com/action/journalInformation?journalCode=tbsd20

Journal of Biomolecular Structure and Dynamics

ISSN: 0739-1102 (Print) 1538-0254 (Online) Journal homepage: https://www.tandfonline.com/loi/tbsd20

Formation of the inclusion complex of water

soluble fluorescent calix[4]arene and naringenin:

solubility, cytotoxic effect and molecular modeling

studies

Mehmet Oguz, Asif Ali Bhatti, Berna Dogan, Serdar Karakurt, Serdar Durdagi

& Mustafa Yilmaz

To cite this article: Mehmet Oguz, Asif Ali Bhatti, Berna Dogan, Serdar Karakurt, Serdar

Durdagi & Mustafa Yilmaz (2020) Formation of the inclusion complex of water soluble fluorescent calix[4]arene and naringenin: solubility, cytotoxic effect and molecular modeling studies, Journal of Biomolecular Structure and Dynamics, 38:13, 3801-3813, DOI: 10.1080/07391102.2019.1668301 To link to this article: https://doi.org/10.1080/07391102.2019.1668301

View supplementary material Published online: 11 Oct 2019.

Submit your article to this journal Article views: 367

View related articles View Crossmark data

(2)

Formation of the inclusion complex of water soluble fluorescent calix[4]arene

and naringenin: solubility, cytotoxic effect and molecular modeling studies

Mehmet Oguza,b , Asif Ali Bhattia,c , Berna Dogand , Serdar Karakurte , Serdar Durdagid and Mustafa Yilmaza

a

Department of Chemistry, Selcuk University, Konya, Turkey;bDepartment of Advanced Material and Nanotechnology, Selcuk University, Konya, Turkey;cDepartment of Chemistry, Government College University Hyderabad, Hyderabad, Pakistan;dComputational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahcesehir University, Istanbul, Turkey;eDepartment of Biochemistry, Selcuk University, Konya, Turkey

Communicated by Ramaswamy H. Sarma

ABSTRACT

Naringenin is considered as an important flavonoid in phytochemistry because of its important effect on cancer chemoprevention. Unfortunately its poor solubility has restricted its therapeutic applications. In this study, an efficient water-soluble fluorescent calix[4]arene (compound 5) was synthesized as host macromolecule to increase solubility and cytotoxicity in cancer cells of water-insoluble naringenin as well as to clarify localization of naringenin into the cells. Complex formed by host–guest interaction between compound 5 and naringenin was analyzed with UV–visible, fluorescence, FTIR spectroscopic techniques and molecular modeling studies. Stern–Volmer analysis showed binding constant value of Ksv3.5 107M1suggesting strong interaction between host and guest. Binding capacity shows 77%

of naringenin was loaded on compound 5. Anticarcinogenic effects of naringenin complex were eval-uated on human colorectal carcinoma cells (DLD-1) and it was found that 5-naringenin complex inhib-its proliferation of DLD-1 cells 3.4-fold more compared to free naringenin. Fluorescence imaging studies show 5-naringenin complex was accumulated into the cytoplasm instead of the nucleus. Increased solubility and cytotoxicity of naringenin with fluorescent calix[4]arene makes it one of the potential candidates as a therapeutic enhancer. For deep understanding of host–guest interaction mechanisms, complementary multiscale molecular modeling studies were also carried out.

ARTICLE HISTORY Received 10 July 2019 Accepted 28 August 2019 KEYWORDS Water soluble; calix[4]arenes; naringenin; inclusion complexes; fluorescence; solubilization; cytotoxicity; molecu-lar docking 1. Introduction

Flavonoids are secondary metabolites of plants, synthe-sized via Shikimic acid pathway following pentose

phos-phate pathway and glycolysis (Cao et al., 2019;

Crozier, Clifford, & Ashihara, 2008). They are known as

micronutrients that are widely identified in foods of plant origin. These are recognized as potent antioxidants, pos-sessing the bioactive potential to reduce cancer risk,

pre-vent cardiovascular disease and neurodegenerative

disorders (Lee et al., 2008; Tu, Liu, et al., 2015;

Williams, Spencer, & Rice-Evans, 2004; Yadav et al., 2016). One of the important subgroups of flavonoids is flavonone that possesses anticancer activity beyond antioxidation effect that arise due to its capability to scavenge free radi-cals. It can have effects on cell proliferation, inhibition of angiogenesis and subcellular signaling with stimulation of

DNA repair enzymes (Erlund,2004; Gao et al.,2006; Kumar,

Birundha, Kaveri, & Devi, 2015; Meshram et al., 2019;

Sakalli, Burkina, Pilipenko, Zlabek, & Zamaratskaia, 2018; Selvaraj, Krishnaswamy, Devashya, Sethuraman, & Krishnan,

2014). Among flavanone, naringenin is considered as an

phytoestrogen with weak estrogenic and antiestrogenic activities that inhibits proliferation of colon cancer cells and melanoma cells (Frydoonfar, McGrath, & Spigelman,

2003; Isobe et al., 2018; T€urkkan, €Ozy€urek, Bener, G€uc¸l€u, &

Apak, 2012). A number of properties has been evaluated

for this flavanone such as it assists in reducing rate of migration of hepatocarcinoma and pancreatic cancer cells and decreases metastasis of melanoma cells up to 63%

(Lentini, Forni, Provenzano, & Beninati, 2007; Tu, Wang,

et al., 2015). Furthermore, reports suggest that naringenin

inhibits cytochrome P450 activity which can change pharmacokinetics in human liver and decrease estrogen synthesis and also has chemoprevention effects on cancer and suppresses toxic effects of aflatoxin B1 in human liver

(Buening et al., 1981; Moon, Wang, & Morris, 2006).

Furthermore, naringenin significantly inhibits migration of bladder cancer cells through inhibition of AKT and NF-kB (Liao et al.,2014).

However, the poor water solubility of this flavanone makes it less bioactive. To tackle the problem of solubility of this

CONTACT Mustafa Yilmaz myilmaz42@yahoo.com Department of Chemistry, Selcuk University, Konya 42075, Turkey; Serdar Durdagi

serdar.durdagi@med.bau.edu.tr Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahcesehir University, Istanbul, Turkey.

Supplemental data for this article is available online athttps://doi.org/10.1080/07391102.2019.1668301.

ß 2019 Informa UK Limited, trading as Taylor & Francis Group

2020, VOL. 38, NO. 13, 3801–3813

(3)

compound, different methods are being used in pharmaceutical applications, such as reduction in particle size, modification of crystal structure by the formation of various polymorphic forms, ionization of the molecule, addition of solubilizing agents or improvement of the wettability of the powder, surfactant-stabi-lized nanosuspension formation are formed (Gholami &

Bordbar, 2014; Kanaze et al., 2010; Sahoo et al., 2011;

Semalty, Semalty, Singh, & Rawat,2010; Qian et al.,2019; Zhou, Yang, et al.,2018; Zhao et al.,2018). Moreover, in recent years, supramolecular chemistry has gained large attention in drug delivery system and enhancement of solubility of the water-insoluble drugs (Bono et al.,2018; Drakalska et al., 2014). Thus, different macromolecules have been used as drug carriers (Guo, Wang, Wang, & Liu, 2012; Semalty, Tanwar, & Semalty, 2014;

Tommasini, Calabro, Raneri, Ficarra, & Ficarra, 2004;

Tozuka, Kishi, & Takeuchi, 2010; Yang, 2011; Yilmaz & Sayin,

2016). Among them, calixarenes are well-established

macromo-lecules with excellent host properties, particularly water-soluble versions with sulfo functionality at upper rim facilitates the

water solubility of guest molecules (Guo & Liu, 2014;

Mareeswaran et al., 2014; Ran et al., 2018; Ukhatskaya et al.,

2010; Yang , Zhao, et al.,2016; Zhou, Li, & Yang,2015). Besides, enhancement of solubility of the drug, drug carrier with fluores-cence imaging strategies are expanding rapidly for tracking drug in targeted cells. Initially, fluorescence was mostly used from small organic dyes attached by means of antibodies to the protein of interest (Giepmans, Adams, Ellisman, & Tsien,

2006; Yang, Ran, et al.,2016; Yang, Xie, et al.,2016; Yang et al.,

2015). But in changing tactics, fluorescence technique can open new possibilities in safe and effective drug delivery systems along with synergistic advancements in fluorescent probes, tar-geting strategies, instrumentation and data analysis that enable high throughput screening, single-molecule (Das, Ghosh, Koley,

& Singha Roy,2018; Parveen, Misra, & Sahoo,2012; Zhou, Zhao, et al.,2018).

In this study, we have synthesized a water-soluble cal-ix[4]arene containing fluorescent moiety at the lower rim (compound 5) and formed the water-soluble inclusion complex with naringenin. The complex was investigated for proliferation of human colon cancer cells and clarifica-tion of localizaclarifica-tion in living cells. Molecular modeling stud-ies were also applied to better understand the interaction

between host–guest complex formed by naringenin and

the calix[4]arene derivative.

2. Materials and methods 2.1. Materials

All the chemicals used in this work were of reagent grade or analytical grade and obtained from various commercial sources. All aqueous solutions were prepared with deionized water that was obtained via a Millipore milli-Q Plus water purification sys-tem and 100/ATR Sampling Accessory. pH was measured

through Orion 410Aþ pH meter. CEM-MDS 2000 closed vessel

microwave system was used in this study to prepare real and certified samples. IR spectra were performed on a Perkin Elmer spectrum 100 FT-IR spectrometer (ATR). UV/Vis spectra were

measured with Shimadzu 1700 UV–Vis spectrophotometer.

Bio-imaging studies were performed by using ZOE Fluorescent Cell Imager (Bio-Rad, CA, USA).

2.2. Synthesis

Figure 1 represents the schematic rout for the synthesis of

compound 5 according to the previously reported method.

Figure 1. The schematic route for the synthesis of dansyl p-sulfonatocalix[4]arene (compound 5). (i) BrCH2COOCH3/K2CO3/Acetone. (ii) Tris(2-aminoethyl)amine/

(4)

Synthesis of compound 5 was confirmed by1HNMR. % Yield: 71,1HNMR (D2O, 25 C) dH:2.16 (m, 2 H, CH2N), 2.39 (m, 4 H,

CH2N) 2.65 (s, 6 H, N(CH3)2), 3.01 (m, 8 H, NHCH2), 3.61 (m,

4 H, ArCH2Ar), 4.15 (m, 2 H, NHSO2),4.31 (m, 4 H, ArCH2Ar),

4.69 (s, 4 H, OCH2þH2O(solvent)) 7.21 (s, 4 H, Ar-H/Calix), 7.50

(s, 4 H, Ar-H/Calix), 7.67 (m, 2 H, ArHdans). 8.03 (m, 2 H,

ArHdans), 8.13 (m, 1 H, ArHdans), 8.28 (m, 1 H, ArHdans) (Collins

et al., 1991; Liu, He, Qing, Xu, & Qin, 2005; Oguz, Bhatti, Karakurt, Aktas, & Yilmaz,2017).

2.3. Preparation of inclusion complex

100 mg of compound 5 was taken in 50 mL Erlenmeyer flask and dissolved in 10 mL water with adjusting pH up to 8.0. In a separate Erlenmeyer flask, the equimolar amount of narin-genin was dissolved in 3 mL methanol and mixed with the solution containing compound 5 and stirred at room tem-perature for 4 hours at room temtem-perature. Methanol was removed using vacuum distillation and the water phase was filtered by using 0.45lm PTFE filter to get the water-soluble material. The filter was washed with methanol and remaining amount of naringenin was measured by the spectro-scopic method.

Binding capacity % (BC %) was calculated by applying the following formula:

BCð%Þ ¼Total mass of drugFree drug in supernatant Total mass of drug  100

(1)

2.4. Determination of binding constant and solubility constant

Stern–Volmer analysis was utilized to probe the nature of

the quenching or enhancement process in the complexation of the drug (Lakowicz & Weber,1973; Zhu, Sun, Wang, Xu, & Wang,2017).

Io

I ¼ 1 þ Ksv½ Q (2)

where Iois the fluorescence intensity for compound 5 in the

absence of naringenin, Io is the fluorescence intensity of

compound 5 in the presence of naringenin. Ksv static

quenching constant and Q is the quencher concentration.

Benesi–Hildebrand equation was used to determine the

binding constant for compound 5-naringenin

complex complex. 1 I Io¼ b a b 1 K M½  þ1   (3)

where I0 represents the fluorescence intensity of compound

5 and I is the fluorescence intensity of for 5-naringenin com-plex, [M] is the concentration of the naringenin and K is the binding constant for complex. From the plot of 1/I– I0vs. 1/

[M], value of b/a– b can be found.

The aqueous solubility of naringenin was determined with increasing concentrations of the compound 5 at pH 7.0, by using Higuchi and Conners method (Higuchi & Connors,

1965). 10 mg of naringenin was taken in each test tube

containing 10 mL aqueous solution (1 105 to 1 104).

The mixtures were equilibrated for 4 h and filtered through 0.45lM nylon filter. The concentrations of dissolved substan-ces in water were determined by absorption spectroscopy.

Ks¼

Slope So ð1  SlopeÞ

(4)

where So is the intrinsic solubility of naringenin and slope

value was obtained from the plot using Higuchi and Conners method.

2.5. Cell culture studies

The human colon cancer cell line, DLD1 was provided from ATCC (American Type Culture Collection, ATCC, Rockville,

MD, USA) and cultured in RPMI-1640 (ATTC-30-2001)

medium. All media were supplemented with 10% fetal

bovine serum, 1% L-glutamine and 1%

penicillin/strepto-mycin. This growth medium was also used for dilution of other chemicals. The cell lines were maintained at 37C in a

5% CO2atmosphere and 95% humidity.

2.6. Cell proliferation assays

DLD-1 cells were seeded into 96-well plates (1 104 cell/

well) as three replicates in order to determine the IC50value

of naringenin, compound 5 and 5-naringenin complex were incubated for 48 h at 37C in 5% CO2. The effect of

naringe-nin, compound 5 and 5-naringenin complex on the

prolifer-ation of DLD-1 cells and IC50 values were determined by

using various concentrations ranging from 0.1 to 200mM.

The differences in cell growth were monitored by Alamar blue method in which conversion of resazurin (blue color) to resorufin (pink color) measured by spectrophotometrically.

2.7. Cell imaging studies

Following the calculation of IC50 values, DLD-1 cells were

seeded on 18 mm glass coverslips and allowed to adhere for 24 h. Following to incubation, the medium was replaced with fresh one containing 5-naringenin complex and incubated

1 h at 37C in a 5% CO2atmosphere and 95% humidity. The

images were obtained by ZOE Fluorescent Cell Imager (Bio-Rad, California, USA).

2.8. Computational methods 2.8.1. Geometry optimizations

The crystal structure of tetrahydroxycalix[4]arene was downloaded from Cambridge Structural Database (CSD identifier WUVKON, Deposition number 184678) (Atwood,

Barbour, & Jerga, 2002) and it was modified to compound

5 using Maestro molecular modeling package (2015). The structure was first minimized using OPLS3 force field

(Harder et al., 2016) to remove steric clashes. The pKa of

ionizable groups such as amines, amides, hydroxy groups etc., were predicted by MarvinSketch program (v., 16.1.4.0,

(5)

compound found as –4. MacroModel module of Maestro was used to generate different conformers of compound 5 using OPLS3 force field force field and 115 conformers were generated. The structure of the conformer with the lowest energy was then optimized by quantum mechanical (QM) calculations at density functional theory (DFT) level.

The hybrid functional B3LYP (Becke, 1988; Lee, Yang, &

Parr, 1988) with basis set of 6-31G was used for first

optimization at gas phase with convergence criteria of

5.0 106 Hartree. The final structure obtained at this

basis set was then subjected to further optimization again

with B3LYP functional but using 6-31Gþþ basis set. The

atomic charges from electrostatic potential (ESP) fitting was also obtained by DFT calculations (at

B3LYP/6-31Gþþ level) to be used for further docking and

molecular dynamics (MD) simulations.

The 3D structure of naringenin was downloaded from PubChem database and LigPrep module of Maestro with OPLS3 force field was used to optimize the geometry. In

addition, Epik (Shelley et al., 2007) was used to generate

ionization and tautomeric states for molecules in aqueous solution at pH 7.4. As there was one chiral center, both R

and S isomers were generated to be used in host–guest

complex formation. MacroModel module was used to gen-erate different conformers for both isomers (14 different conformers for each isomer). The structures of all conform-ers were then optimized by DFT calculations at

B3LY3/6-31Gþþ level and ESP charges were also calculated for

each conformer.

2.8.2. Molecular docking

The initial host–guest interaction between compound 5 and

R/S naringenin was predicted by molecular docking methods. The Glide docking with extra precision (XP) option (Friesner et al.,2004) was used to predict the initial complex structure. In docking simulations, compound 5 was considered as receptor (host) while naringenin R/S were treated as ligand (guest). The center of the binding cavity of compound 5 (eg grid center) was determined according to following Cartesian

coordinates x ¼ –3.40 Å, y ¼ –2.50 Å and z ¼ 1.43 Å. The

inner box was set as 10 Å 10 Å  10 Å while the outer box

was set as 20 Å  20 Å  20 Å. To soften the potential for

the nonpolar parts of the host compound 5, the van der Waals radii of atoms with partial atomic charges less than 0.25 were scaled by a factor of 0.8. The conformers of R/S naringenin generated by MacroModel and further optimized by DFT calculations were used for ligand docking with Glide/ XP. For each isomer of naringenin, at most 100 docking poses were requested. The intramolecular hydrogen bonds

were asked to be rewarded and planarity of conjugated p

groups were enhanced. The potential of nonpolar part of ligands were also softened by scaling of van der Waals radii of atoms with partial atomic charges less than 0.15 by a fac-tor of 0.6. The previously calculated ESP charges of both host and guest molecules were used. Docking poses obtained were clustered based on the root mean square deviations (RMSD) of ligands, R/S naringenin, with a cutoff value of 2.0 Å.

2.8.3 Molecular Dynamics (MD) Simulations

A representative host–guest complex structure from each

cluster of docking poses as well as top docking poses for both isomers were subjected to MD simulations. For R iso-mer, six different complex structures were considered for MD simulations, whereas for S isomer, eight different

host–guest complexes were used. While ESP charges

obtained by QM calculations were used in host-guest com-plexes, OPLS3 force field was utilized to obtain other force

field parameters in Desmond. (Bowers et al.,2006), . Each

system was then solvated using TIP3P water model

(Jorgensen, Chandrasekhar, Madura, Impey, & Klein, 1983)

in orthorhombic boxes with the sizes of boxes calculated by buffer method and distances along all three directions were chosen as 25 Å. For each system, first the default relaxation protocol of Desmond was applied in which a series of minimizations and short MD simulations were performed to relax the model system before production MD simulations. The production MD simulations were per-formed under NPT (constant pressure and temperature) ensemble for 100 ns. During production simulations

Nose-Hoover thermostat (Nose-Hoover, 1985; Nose,1984) was utilized

while the initial temperature was set as 310 K. The pres-sure was set at 1.01325 bar and it was initially controlled by the Martyna-Tobias-Klein method (Martyna, Tobias, &

Klein, 1994) with isotopic coupling style and relaxation

time of 2 ps. The equation of motions in dynamics was cal-culated using the multi-step RESPA integrator with time steps of 2.0, 2.0 and 6.0 fs for bonded, near non-bonded and far non-bonded interactions, respectively. For the

short-range electrostatic and Lennard–Jones interactions

the cutoff value was set as 9.0 Å. Long-range interactions were estimated using Particle mesh Ewald (Ananchenko

et al., 2007) method (Darden, York, & Pedersen, 1993;

Ewald, 1921) along with periodic boundary conditions. The

trajectories obtained by MD simulations were also clus-tered based on the RMSD fluctuations of the ligand R/S naringenin. The MM/GBSA approach (Lyne, Lamb, & Saeh,

2006; Sirin et al., 2014) implemented in Prime module of

Maestro with solvent model of VSGB 2.0 (Li et al., 2011)

was used to calculate the binding free energies.

3. Result and discussion

Controlled drug delivery requires the understanding of drug design, stability and metabolism together with the complexities imposed by the biological systems such as drug-target interaction. Fluorescence technique provides a comprehensive tool for investigating many aspects of drug

delivery in single cells and whole tissue (White &

Errington, 2005). Many drugs are inherently

auto-fluores-cent and therefore can be tracked using microscopy tech-niques, while others need ancillary methodologies. In this scenario, by maintaining the drug stability, efficient carrier along with fluorescence probe is required (Johnson &

Spence, 2010). The purpose of this work is to utilize

water-soluble fluorescence calixarene as solubility enhancer for naringenin, as well as to increase the cytotoxicity of

(6)

naringenin in cancer cells.. Compound 5 was synthesized and characterized according to the previously published

method (Oguz et al.,2017). The compound 5 was selected

due to presence of different functionalities such as -SO3H

at upper rim that provide good environment to calixarene to accommodate guest species, while fluorescent dansyl moiety at lower rim help to identity the presence of com-plex in living cell. Therefore, compound 5 may act as an ideal derivative for this study (Figure 3).

3.1. FTIR analysis

Interaction of naringenin with compound 5 was confirmed with FTIR spectroscopy by comparing individual component with

complex. Figure 2(a) shows spectrum of compound 5 with

broad band at 3359 cm1 for –OH group, characteristic band

for –SO3 appeared at 1189 and 1041 cm1, respectively.

Characteristic band appeared at 1630 cm1 assigned to C-N

band for amide and at 792 cm1 for aromatic ring (Valand,

Patel, & Menon, 2015). Naringenin shows broad peak at

3287 cm1for -OH vibration, 1628 and 1588 cm1are assigned to the stretching vibration for C¼ O and C ¼ C aromatic group,

1309 and 1150 cm1 are correspond to the C–O–C vibration

and 1065 cm1correspond to C–H of the aromatic group

bend-ing vibration (Semalty et al., 2010). The complex shows quite different characteristics bands with broadening of OH stretching band at 3367 cm1due to interaction of phenolic–OH of

narin-genin with SO3 through hydrogen bonding. Presence of broad

band at 1600 cm1is due to merging of different bands (C¼ O and C¼ C), this can also be attributed to p-p stacking interac-tions of naringenin and calixarene cavity, that confirms forma-tion of complex (Figure 3).

3.2. UV–visible and fluorescence study

Confirmation of inclusion complex was carried out by

analyz-ing it through UV–Vis and fluorescence spectroscopy. As

evi-denced from the absorbance and emission spectra (Figure

4(a,b)), compound 5 and 5-naringenin complex have

differ-ent profiles. The blue shift in 5-naringenin complex from 289

Figure 2. FTIR spectra of compound 5 (a), naringenin (b) and 5-naringenin complex (c).

(7)

to 324 nm (Figure 4(a)) can be attributed to the interaction of phenolic moieties of naringenin with sulphate present at upper rim of calixarene that enforce hydrophilic interactions

(Valand et al.,2015). Compound 5 is highly fluorescent with

dansyl moiety at its lower rim. We can use fluorescent spec-troscopy to analyze the effect on intensity of compound 5 by titrating it with naringenin that is regarded as

non-fluor-escent.Figure 4(b)shows the increase in intensity at 505 nm

in fluorescence spectra attributed structural changes due to holding the naringenin in calix[4]arene cavity (Mareeswaran et al.,2014). This result clearly demonstrated that the entrap-ment of hydrophobic naringenin in the hydrophilic environ-ment created by the compound 5 is crucial for the solubilizing process.

3.3. Determination of Stoichiometric ratio, binding and solubility constant

Effect on fluorescence intensity with changing concentration of naringenin was observed. Data obtained was fitted in

modified Benesi–Hildebrand equation (Eq. 3). From the plot

of 1/I– I0 and 1/[M] (Figure 5(a)) binding constant K was

cal-culated for 5-naringenin complex as 1.2 107 M1. From

Stern–Volmer plot (Figure 5(b)), Ksv was determined as

3.5 107 M1. The Ksvvalue suggests the high binding

affin-ity of compound 5 for the naringenin in aqueous media. Binding of naringenin with compound 5 was also evident from binding constant

Figure 4. UV–visible spectra (a) and fluorescence response (b) of 5, naringenin and 5-naringenin complex.

Figure 5. Benesi–Hildebrand plot compound 5 (1.5  106M) and naringenin (1 108M to 3.3 108M) (a). Stern–Volmer plot (b).

Figure 6. Phase solubility diagram for 5-naringenin as per Higuchi Connors method.

Figure 7. Effects of naringenin and 5-naringenin complex on viability and pro-liferation of DLD-1 cells, the viability of DLD-1 cells after treatment with differ-ent concdiffer-entrations of naringenin and 5-naringenin measured by Alamar blue assay, IC50values were determined from the sigmoidal plot of compound

(8)

Solubility studies were carried out using phase solubility technique devised by Higuchi and Connors (1965) (Figure 6).

Many phenolic and flavonoid compounds have

anticarcinogenic and antiproliferative properties, however, the main problem is inadequate solubility which decreases the bioavailability of the compound. One of the main active

Figure 8. Fluorescence images and their corresponding bright-field transmission images: Bright-field (right panels, (aand dw), DLD-1 cells incubated with

5-narin-genin complex (10mM) (middle, (b and ew)) and overlap of images of bright field and fluorescence (left panels, (c and fw)), respectively. , 20 magnification andw, 40 magnification.

Figure 9. The optimized structure of compound 5 by B3LYP hybrid functional with 6-31Gþþ basis set. Gray, red, blue, yellow and white colored spheres repre-sent C, O, N, S and polar H atoms, respectively. Yellow dashed lines show the hydrogen bonds. (a and b) Side views and (c and d) bottom and top views, respectively.

(9)

compounds in grapefruit is naringenin which affects the metabolism of many xenobiotics including drugs and proto-xicants, metabolized by cytochrome P450 enzymes especially,

CYP3A4 (Edwards & Bernier, 1996). Hence, inhibition of

CYP3A4 has crucial importance to prevent cytotoxicity of those hazardous compounds. Naringenin was proved to inhibit the activity of this enzyme (Henderson, Miranda, Stevens, Deinzer, & Buhler,2000).

Increased bioavailability of naringenin may protect living systems against toxic compounds. Therefore, interaction of naringenin with water-soluble calix[4]arene increases the solubility in water, which protects toxic effects of organic sol-vents. Cytotoxicity studies showed that compound 5 has no toxic effect on proliferation of DLD-1 cells. When compared with free naringenin, cytotoxic effects of 5-naringenin

com-plex was elevated 3.43-fold (p< 0.001) as IC50 value was

decreased from 66mM (free naringenin) to 19.2 mM

(5-narin-genin complex), (Figure 7).

Following the cytotoxicity studies, the cells were

incu-bated with 10mM of 5-naringenin complex for 45 min to

clar-ify localization in living cells. Fluorescence imaging studies showed 5-naringenin complex was accumulated into the cytoplasm instead of the nucleus. Localization of compound 5-naringenin complex in cytoplasm may explain action of compound since one of the important death mechanisms, extrinsic apoptosis was observed to occur in cytoplasm (Figure 8).

3.4. Computational modeling of compound 5-naringenin (R/S)

The DFT optimized geometry of compound 5 at

B3LYP/6-31Gþþ level can be found in Figure 9. The calix[4]arene

part of compound 5 is found at so-called‘pinched cone’

con-formation (Podoprygorina, Bolte, & B€ohmer, 2009; Scheerder et al., 1996; Zuo, Wiest, & Wu, 2011) most likely due to the bridging of upper rim (narrow rim) and intramolecular hydro-gen bonds. In this conformation, the two aromatic rings that are connected via a dansyl group orientated in parallel, while the other two rings are strongly tilted outwards as shown in the single crystal structure of bridged compounds obtained by Podoprygorina et al. (2009). 1,3-alternate conformation of calix[4]arene of compound 5 is also considered and

opti-mized again at B3LYP/6-31Gþþ level, however ‘pinched

cone’ conformation is found to be the lower energy

con-former between the two structures. Moreover, NMR spectrum

of compound 5 also indicates a cone-like structure (Figure

S1b) hence ‘pinched cone’ conformer shown in Figure 9 is

used for further docking and MD studies.

The initial host–guest complex structures between

com-pound 5 and R/S naringenin has been determined by molecular docking studies. The poses generated by Glide/ XP are clustered based on RMSD fluctuations of ligand orientation and considering RMSD value of less than 2.0 Å resulted in five complex structures for R isomer and seven

Figure 10. The representative structures obtained after the MD simulations of compound 5/R-naringenin complex. The ligand naringenin is represented by green colored ball and sticks while compound 5 is represented in gray color. Atom coloring scheme is same asFigure 9and yellow dashed lines represent hydrogen bonds, whereas blue dashed lines representp-p stacking interactions. Ligand electrostatic potential is also displayed as mesh surfaces with blue to red colored areas representing negative to positive partial charges.

(10)

complex structures for S isomer with at least five members in each cluster. All docking poses could be found in

Figures S2 and S3. Both isomers of naringenin are partially

included in host compound 5 based on the docking poses and they could not cross completely the cavity of calix[4]-arene most likely due to the hydrogen bonding

interac-tions with SO3– groups at the lower rim of calix[4]arene

and/or the tightening of upper rim part of cavity due to the bringing of 1,3 aromatic rings by dansyl group. This is actually consistent with our previous study in which quer-cetin, another flavonoid, formed a complex with a calix[4]-arene derivative and in silico studies again indicated that it could only be partially included in the calix[4]arene cavity (Yilmaz et al.,2019).

The top-docking poses as well as representative structures from each cluster of docking poses for both isomers have been used as initial structures for MD simulations and 100-ns simulations were run for each complex structure. The trajec-tories obtained by MD are visualized to analyze the stability

of interactions between host and guest compounds.

Regardless of the initial structure of MD simulations, R-narin-genin, in most of the trajectories, is found as attached to the

host compound 5 especially for starting structure in Figure

S2(b). However, the detachment of guest molecule

R-narin-genin is also observed such that guest molecule was outside of the calix[4]arene cavity but still interacting with the host compound. After the clustering of all six 100 ns MD

simula-tions, representative host–guest complex structures for

R-Figure 11. The representative structures obtained after MD simulations complex of compound 5 and S-naringenin. The ligand naringenin is represented by green colored ball and sticks while compound 5 is represented in gray color. Atom coloring scheme is same asFigure 9and yellow dashed lines represent hydrogen bonds, whereas blue dashed lines representp-p stacking interactions. Ligand electrostatic potential is also displayed as mesh surfaces with blue to red colored areas representing negative to positive partial charges.

(11)

naringenin were identified (Figure 10). While complete

separ-ation of host–guest complex has also been witnessed in

some trajectories, such as the MD simulations started from initial pose inFigure S2(e) and (f), the re-attachment and/or re-interactions of host–guest are also observed in some cases

as shown inFigure 10(d). In the case of S-naringenin–

com-pound 5 complex, more detachment events are witnessed by analysis of all eight 100 ns MD simulation trajectories.

Some observed host–guest complex structures for

S-naringe-nin after clustering of all MD trajectories could be found in

Figure 11. Though guest S-naringenin leaves the cavity of

calix[4]arene, in most of the trajectories it again re-attaches to the cavity and/or comes close to interact with compound 5 after a time of complete detachment such as shown in

Figure 11(e). We have also witnessed some conformational

changes of complex as seen in Figures 10(c) and11(c) such

that not only the dansyl group which is quite mobile during MD trajectories but also one of the calix[4]arene aromatic

rings changed its conformation to so called ‘partial cone’

conformation (Zuo et al.,2011).

Additionally, MM/GBSA calculations are performed for all MD simulations of R/S naringenin-compound 5 complexes though this method could only give crude binding energies that could not be necessarily compared to experimental

val-ues (Genheden & Ryde, 2015; Godschalk, Genheden,

S€oderhjelm, & Ryde, 2013; Greenidge, Kramer, Mozziconacci, & Wolf, 2013). Nevertheless MM/GBSA energies could aid to understand the kind of interactions that drive the host–guest complex formation. Here we have also observed that there is a distinct difference between binding energy of complex when the guest compound is in cavity of calix[4]arene and when the guest leaves the cavity but continue to interact with the host molecule (as shown inFigures 10(d)and11(e)). For R-naringenin in complex with compound 5 the average

binding energy found as –21.61 ± 7.61 kcal/mol when the

guest is in cavity during all MD trajectories, whereas the binding energy is found as–12.23 ± 7.24 kcal/mol in case that R-naringenin leaves the cavity but still interacts with com-pound 5 during MD simulations. In both cases the binding energy is mostly driven by electrostatic, van der Waals and lipophilic interactions as can be seen from the binding

energy components list given inTable 1. The same

phenom-enon is true for S-isomer of naringenin where in the case that guest compound staying in the calix[4]arene cavity the

binding energy is found as –24.21 ± 4.30 kcal/mol, whereas

the value of binding energy decreases to –13.71 ± 9.10 kcal/

mol when the S-naringenin leaves the cavity but continue interacting with the host compound 5. The driving force for the complex formation again seems to be electrostatic, van der Waals and lipophilic interactions in both cases for S-iso-mer. As mentioned before, binding energy values for both R/

S-naringenin complex formation with compound 5 may not be directly compared to experimental binding energy value –10.70 kcal/mol (calculated from binding constant value of Ksv3.5 107 M1). In summary, it could be said that complex

formation between naringenin (R/S) and compound 5 are found to be favorable and the molecules are strongly inter-acting based on the low binding energies.

4. Conclusion

In this study, water-soluble fluorescent calix[4]arene (5) used as a host for water-insoluble naringenin to enhance its water

solubility and introduce fluorescent property. Host–guest

complex was formed at pH 8.0. Spectroscopic measurements and molecular modeling studies were conducted to broaden the host–guest interaction, which confirmed the efficiency of compound 5 as drug carrier. Moreover, it has been found that 5-naringenin complex possesses anti-proliferative effects

of human colon cancer cells with IC50 value of 19.2mM.

Besides, fluorescent property of compound 5 can track pres-ence of non-fluorescent naringenin in the cells. 5-Naringenin complex has been demonstrated to localize in cytoplasmic part of cell. Compound 5 could be versatile candidate as drug carrier in pharmaceutical industry.

Disclosure statement

No potential conflict of interest was reported by the authors.

Funding

This study was financially support by the Research Foundation of Selcuk University (Project No.: 19401060).

ORCID

Mehmet Oguz http://orcid.org/0000-0002-3999-620X

Asif Ali Bhatti http://orcid.org/0000-0001-5921-532X

Berna Dogan http://orcid.org/0000-0002-5650-5177

Serdar Karakurt http://orcid.org/0000-0002-4449-6103

Serdar Durdagi http://orcid.org/0000-0002-0426-0905

Mustafa Yilmaz http://orcid.org/0000-0003-2904-160X

References

Ananchenko, G. S., Udachin, K. A., Pojarova, M., Jebors, S., Coleman, A. W., & Ripmeester, J. A. (2007). A molecular turnstile in para-octa-noyl calix[4]arene nanocapsules. Chemical Communications, (7), 707–709. doi:10.1039/B613972H

Atwood, J. L., Barbour, L. J., & Jerga, A. (2002). Storage of methane and freon by ınterstitial van der Waals confinement. Science (New York, N.Y.), 296(5577), 2367–2369. doi:10.1126/science.1072252

Table 1. Contributions to binding energy for naringenin (R/S)– compound 5 complexes calculated using MM/GBSA method.

DEelectrostatic DEvdw DElipophilic DGsolv GB DEHbond corr DEpacking corr DG binding

R-naringenin-a –7.39 –15.41 –8.50 13.05 –0.60 –2.76 –21.61 ± 7.61 R-naringenin-b –2.80 –10.09 –3.37 5.82 –0.14 –1.64 –12.23 ± 7.24 S-naringenin-a –7.74 –17.54 –9.87 14.71 –0.57 –3.21 –24.21 ± 4.30 S-naringenin-b –3.99 –10.45 –4.28 7.19 –0.24 –1.94 –13.71 ± 9.10 All values are given in kcal/mol.

(12)

Becke, A. D. (1988). Density-functional exchange-energy approximation with correct asymptotic behavior. Physical Review A, 38(6), 3098–3100. doi:10.1103/PhysRevA.38.3098

Bono, N., Pennetta, C., Sganappa, A., Giupponi, E., Sansone, F., Volonterio, A., & Candiani, G. (2018). Design and synthesis of biologic-ally active cationic amphiphiles built on the calix [4] arene scaffold. International Journal of Pharmaceutics, 549(1–2), 436–445. doi:10.1016/ j.ijpharm.2018.08.020

Bowers, K. J., Chow, D. E., Xu, F., Dror, R. O., Eastwood, M. P.,Gregersen, B. A., … Shaw, D. E. (2006). Scalable algorithms for molecular dynam-ics simulations on commodity clusters. SC’06: Proceedings of the 2006 ACM/IEEE Conference on Supercomputing, IEEE,Tampa, Florida. Buening, M. K., Chang, R. L., Huang, M.-T., Fortner, J. G., Wood, A. W., &

Conney, A. H. (1981). Activation and inhibition of benzo(a)pyrene and aflatoxin B1 metabolism in human liver microsomes by naturally occurring flavonoids. Cancer Research, 41(1), 67–72.

Cao, X., He, Y., Kong, Y., Mei, X., Huo, Y., He, Y., & Liu, J. (2019). Elucidating the interaction mechanism of eriocitrin withb-casein by multi-spectroscopic and molecular simulation methods. Food Hydrocolloids, 94, 63–70. doi:10.1016/j.foodhyd.2019.03.006

Collins, E. M., McKervey, M. A., Madigan, E., Moran, M. B., Owens, M., Ferguson, G., & Harris, S. J. (1991). Chemically modified calix[4]arenes. Regioselective synthesis of 1,3-(distal) derivatives and related com-pounds. X-Ray crystal structure of a diphenol-dinitrile. Journal of the Chemical Society, Perkin Transactions, 1(12), 3137–3142. doi:10.1039/ p19910003137

Crozier, A., Clifford, M. N., & Ashihara, H. (2008). Plant secondary metabo-lites: Occurrence, structure and role in the human diet. New Jersey: John Wiley & Sons.

Darden, T., York, D., & Pedersen, L. (1993). Particle mesh Ewald: An Nlog(N) method for Ewald sums in large systems. The Journal of Chemical Physics, 98(12), 10089–10092. doi:10.1063/1.464397

Das, S., Ghosh, P., Koley, S., & Singha Roy, A. (2018). Binding of naringin and naringenin with hen egg white lysozyme: A spectroscopic investi-gation and molecular docking study. Spectrochimica Acta Part A: Molecular and Biomolecular Spectroscopy, 192, 211–221. doi:10.1016/j. saa.2017.11.015

Drakalska, E., Momekova, D., Manolova, Y., Budurova, D., Momekov, G., Genova, M., … Rangelov, S. (2014). Hybrid liposomal PEGylated calix [4] arene systems as drug delivery platforms for curcumin. International Journal of Pharmaceutics, 472(1–2), 165–174. doi:10.1016/ j.ijpharm.2014.06.034

Edwards, D. J., & Bernier, S. M. (1996). Naringin and naringenin are not the primary CYP3A inhibitors in grapefruit juice. Life Sciences, 59(13), 1025–1030. doi:10.1016/0024-3205(96)00417-1

Erlund, I. (2004). Review of the flavonoids quercetin, hesperetin, and nar-ingenin. Dietary sources, bioactivities, bioavailability, and epidemi-ology. Nutrition Research, 24(10), 851–874. doi:10.1016/j.nutres.2004. 07.005

Ewald, P. P. (1921). Die Berechnung optischer und elektrostatischer Gitterpotentiale. Annalen Der Physik, 369(3), 253–287. doi:10.1002/ andp.19213690304

Friesner, R. A., Banks, J. L., Murphy, R. B., Halgren, T. A., Klicic, J. J., Mainz, D. T., … Perry, J. K. (2004). Glide: A new approach for rapid, accurate docking and scoring. 1. Method and assessment of docking accuracy. Journal of medicinal Chemistry, 47(7), 1739–1749. doi:10.1021/ jm0306430

Frydoonfar, H., McGrath, D., & Spigelman, A. (2003). The variable effect on proliferation of a colon cancer cell line by the citrus fruit flavonoid naringenin. Colorectal Disease : The Official Journal of the Association of Coloproctology of Great Britain and Ireland, 5(2), 149–152. doi:10. 1046/j.1463-1318.2003.00444.x

Gao, K., Henning, S. M., Niu, Y., Youssefian, A. A., Seeram, N. P., Xu, A., & Heber, D. (2006). The citrus flavonoid naringenin stimulates DNA repair in prostate cancer cells. The Journal of Nutritional Biochemistry, 17(2), 89–95. doi:10.1016/j.jnutbio.2005.05.009

Genheden, S., & Ryde, U. (2015). The MM/PBSA and MM/GBSA methods to estimate ligand-binding affinities. Expert Opinion on Drug Discovery, 10(5), 449–461. doi:10.1517/17460441.2015.1032936

Gholami, S., & Bordbar, A.-K. (2014). Exploring binding properties of nar-ingenin with bovine b-lactoglobulin: A fluorescence, molecular dock-ing and molecular dynamics simulation study. Biophysical Chemistry, 187–188, 33–42. doi:10.1016/j.bpc.2014.01.003

Giepmans, B. N. G., Adams, S. R., Ellisman, M. H., & Tsien, R. Y. (2006). The Fluorescent Toolbox for assessing protein location and function. Science, 312(5771), 217–224. doi:10.1126/science.1124618

Godschalk, F., Genheden, S., S€oderhjelm, P., & Ryde, U. (2013). Comparison of MM/GBSA calculations based on explicit and implicit solvent simulations. Physical Chemistry Chemical Physics, 15(20), 7731–7739. doi:10.1039/c3cp00116d

Greenidge, P. A., Kramer, C., Mozziconacci, J.-C., & Wolf, R. M. (2013). MM/GBSA binding energy prediction on the PDBbind data set: Successes, failures, and directions for furtherımprovement. Journal of Chemical Information and Modeling, 53(1), 201–209. doi:10.1021/ ci300425v

Guo, D.-S., & Liu, Y. (2014). Supramolecular chemistry of p-sulfonatoca-lix[n]arenes and its biological applications . Accounts of Chemical Research, 47(7), 1925–1934. doi:10.1021/ar500009g

Guo, D.-S., Wang, K., Wang, Y.-X., & Liu, Y. (2012). Cholinesterase-respon-sive supramolecular vesicle. Journal of the American Chemical Society, 134(24), 10244–10250. doi:10.1021/ja303280r

Harder, E., Damm, W., Maple, J., Wu, C., Reboul, M., Xiang, J. Y., … Knight, J. L. (2016). OPLS3: A force field providing broad coverage of drug-like small molecules and proteins. Journal of Chemical Theory and Computation, 12(1), 281–296. doi:10.1021/acs.jctc.5b00864

Henderson, M., Miranda, C., Stevens, J., Deinzer, M., & Buhler, D. (2000). In vitro inhibition of human P450 enzymes by prenylated flavonoids from hops, Humulus lupulus. Xenobiotica; The Fate of Foreign Compounds in Biological Systems, 30(3), 235–251. doi:10.1080/ 004982500237631

Higuchi, T., & Connors, K. (1965). Advances in analytical chemistry and instrumentation (Vol. 4, pp. 117–212). New York: Interscience. Hoover, W. G. (1985). Canonical dynamics: Equilibrium phase-space

distri-butions. Physical Review A, 31(3), 1695. doi:10.1103/PhysRevA.31.1695

Isobe, T., Ohkawara, S., Ochi, S., Tanaka-Kagawa, T., Jinno, H., & Hanioka, N. (2018). Naringenin glucuronidation in liver and intestine micro-somes of humans, monkeys, rats, and mice. Food and Chemical Toxicology, 111, 417–422. doi:10.1016/j.fct.2017.11.057

Johnson, I., & Spence, M. (2010). A guide to fluorescent probes and labeling technologies. Life Technologies, 11th Edition, California. Jorgensen, W. L., Chandrasekhar, J., Madura, J. D., Impey, R. W., & Klein,

M. L. (1983). Comparison of simple potential functions for simulating liquid water. The Journal of Chemical Physics, 79(2), 926–935. doi:10. 1063/1.445869

Kanaze, F., Kokkalou, E., Niopas, I., Barmpalexis, P., Georgarakis, E., & Bikiaris, D. (2010). Dissolution rate and stability study of flavanone aglycones, naringenin and hesperetin, by drug delivery systems based on polyvinylpyrrolidone (PVP) nanodispersions. Drug Development and Industrial Pharmacy, 36(3), 292–301. doi:10.1080/03639040903140589

Kumar, S. P., Birundha, K., Kaveri, K., & Devi, K. (2015). Antioxidant studies of chitosan nanoparticles containing naringenin and their cytotoxicity effects in lung cancer cells. International Journal of Biological Macromolecules, 78, 87–95. doi:10.1016/j.ijbiomac.2015.03.045

Lakowicz, J. R., & Weber, G. (1973). Quenching of fluorescence by oxy-gen. Probe for structural fluctuations in macromolecules. Biochemistry, 12(21), 4161–4170. doi:10.1021/bi00745a020

Lee, C., Yang, W., & Parr, R. G. (1988). Development of the Colle-Salvetti correlation-energy formula into a functional of the electron density. Physical Review B, 37(2), 785–789. doi:10.1103/PhysRevB.37.785

Lee, E. R., Kang, Y. J., Kim, H. J., Choi, H. Y., Kang, G. H., Kim, J. H., … Cho, S. G. (2008). Regulation of apoptosis by modified naringenin derivatives in human colorectal carcinoma RKO cells. Journal of cellu-lar biochemistry, 104(1), 259–273. doi:10.1002/jcb.21622

Lentini, A., Forni, C., Provenzano, B., & Beninati, S. (2007). Enhancement of transglutaminase activity and polyamine depletion in B16-F10 mel-anoma cells by flavonoids naringenin and hesperitin correlate to reduction of the in vivo metastatic potential. Amino Acids, 32(1), 95–100. doi:10.1007/s00726-006-0304-3

(13)

Li, J., Abel, R., Zhu, K., Cao, Y., Zhao, S., & Friesner, R. A. (2011). The VSGB 2.0 model: A next generation energy model for high resolution pro-tein structure modeling. Proteins: Structure, Function, and Bioinformatics, 79(10), 2794–2812. doi:10.1002/prot.23106

Liao, A. C. H., Kuo, C. C., Huang, Y. C., Yeh, C. W., Hseu, Y. C., Liu, J. Y., & Hsu, L. S. (2014). Naringenin inhibits migration of bladder cancer cells through downregulation of AKT and MMP-2. Molecular Medicine Reports, 10(3), 1531–1536. doi:10.3892/mmr.2014.2375

Liu, S.-y., He, Y-B., Qing, G.-Y., Xu, K.-X., & Qin, H.-J. (2005). Fluorescent sensors for amino acid anions based on calix[4]arenes bearing two dansyl groups. Tetrahedron: Asymmetry, 16(8), 1527–1534. doi:10.1016/ j.tetasy.2005.02.032

Lyne, P. D., Lamb, M. L., & Saeh, J. C. (2006). Accurate prediction of the relative potencies of members of a series of kinaseınhibitors using molecular docking and MM-GBSA scoring. Journal of Medicinal Chemistry, 49(16), 4805–4808. doi:10.1021/jm060522a

Mareeswaran, P. M., Babu, E., Sathish, V., Kim, B., Woo, S. I., & Rajagopal, S. (2014). p-Sulfonatocalix [4] arene as a carrier for curcumin. New Journal of Chemistry, 38(3), 1336–1345. doi:10.1039/c3nj00935a

Martyna, G. J., Tobias, D. J., & Klein, M. L. (1994). Constant pressure molecular dynamics algorithms. The Journal of Chemical Physics, 101(5), 4177–4189.

MarvinSketch, 16.1.4.0 (2016). ChemAxon (http://www.chemaxon.com). Meshram, R. J., Bagul, K. T., Pawnikar, S. P., Barage, S. H., Kolte, B. S., &

Gacche, R. N. (2019). Known compounds and new lessons: Structural and electronic basis of flavonoid-based bioactivities. Journal of Biomolecular Structure and Dynamics, 1–17 (In Press). doi:10.1080/ 07391102.2019.1597770

Moon, Y. J., Wang, X., & Morris, M. E. (2006). Dietary flavonoids: Effects on xenobiotic and carcinogen metabolism. Toxicology In Vitro, 20(2), 187–210. doi:10.1016/j.tiv.2005.06.048

Nose, S. (1984). A unified formulation of the constant temperature molecular dynamics methods. The Journal of chemical Physics, 81(1), 511–519. doi:10.1063/1.447334

Oguz, M., Bhatti, A. A., Karakurt, S., Aktas, M., & Yilmaz, M. (2017). New water soluble Hg2 þ selective fluorescent calix[4]arenes: Synthesis and application in living cells imaging. Spectrochimica Acta Part A: Molecular and Biomolecular Spectroscopy, 171, 340–345. doi:10.1016/j. saa.2016.08.042

Parveen, S., Misra, R., & Sahoo, S. K. (2012). Nanoparticles: A boon to drug delivery, therapeutics, diagnostics and imaging. Nanomedicine: Nanotechnology, Biology and Medicine, 8(2), 147–166. doi:10.1016/j. nano.2011.05.016

Podoprygorina, G., Bolte, M., & B€ohmer, V. (2009). Tetra-urea calix[4]ar-enes 1,3-bridged at the narrow rim. Organic & Biomolecular Chemistry, 7(8), 1592–1598. doi:10.1039/b819710e

Qian, X., Zhou, X., Ran, X., Ni, H., Li, Z., Qu, Q., … Yang, L. (2019). Facile and clean synthesis of dihydroxylatopillar[5]arene-stabilized gold nanoparticles integrated Pd/MnO2 nanocomposites for robust and ultrasensitive detection of cardiac troponin I. Biosensors & Bioelectronics, 130, 214–224. doi:10.1016/j.bios.2019.01.041

Ran, X., Qu, Q., Qian, X., Xie, W., Li, S., Li, L., & Yang, L. (2018). Water-sol-uble pillar [6] arene functionalized nitrogen-doped carbon quantum dots with excellent supramolecular recognition capability and super-ior electrochemical sensing performance towards TNT. Sensors and Actuators B: Chemical, 257, 362–371. doi:10.1016/j.snb.2017.10.185

Sahoo, N., Kakran, M., Shaal, L., Li, L., M€uller, R., Pal, M., & Tan, L. (2011). Preparation and characterization of quercetin nanocrystals. Journal of Pharmaceutical Sciences, 100(6), 2379–2390. doi:10.1002/jps.22446

Sakalli, S., Burkina, V., Pilipenko, N., Zlabek, V., & Zamaratskaia, G. (2018). In vitro effects of diosmin, naringenin, quercetin and indole-3-carbinol on fish hepatic CYP1A1 in the presence of clotrimazole and dexa-methasone. Chemosphere, 192, 105–112. doi:10.1016/j.chemosphere. 2017.10.106

Scheerder, J., Vreekamp, R. H., Engbersen, J. F. J., Verboom, W., van Duynhoven, J. P. M., & Reinhoudt, D. N. (1996). The pinched cone conformation of calix[4]arenes: Noncovalent rigidification of the cal-ix[4]arene skeleton. The Journal of Organic Chemistry, 61(10), 3476–3481. doi:10.1021/jo9600262

Maestro molecular modeling package. (2015). Schr€odinger, LLC, New York, NY.

Selvaraj, S., Krishnaswamy, S., Devashya, V., Sethuraman, S., & Krishnan, U. M. (2014). Investigations on the membrane interactions of naringin and its complexes with copper and iron: _Implications for their cyto-toxicity. RSC Advances, 4(87), 46407–46417. doi:10.1039/C4RA08157A

Semalty, A., Semalty, M., Singh, D., & Rawat, M. (2010). Preparation and characterization of phospholipid complexes of naringenin for effective drug delivery. Journal of Inclusion Phenomena and Macrocyclic Chemistry, 67(3–4), 253–260. doi:10.1007/s10847-009-9705-8

Semalty, A., Tanwar, Y. S., & Semalty, M. (2014). Preparation and charac-terization of cyclodextrin inclusion complex of naringenin and critical comparison with phospholipid complexation for improving solubility and dissolution. Journal of Thermal Analysis and Calorimetry, 115(3), 2471–2478. doi:10.1007/s10973-013-3463-y

Shelley, J. C., Cholleti, A., Frye, L. L., Greenwood, J. R., Timlin, M. R., & Uchimaya, M. (2007). Epik: A software program for pK a prediction and protonation state generation for drug-like molecules. Journal of Computer-Aided Molecular Design, 21(12), 681–691. doi:10.1007/ s10822-007-9133-z

Sirin, S., Kumar, R., Martinez, C., Karmilowicz, M. J., Ghosh, P., Abramov, Y. A., … Sherman, W. (2014). A computational approach to enzyme design: Predictingx-aminotransferase catalytic activity using docking and MM-GBSA scoring. Journal of Chemical Information and Modeling, 54(8), 2334–2346. doi:10.1021/ci5002185

Tommasini, S., Calabro, M. L., Raneri, D., Ficarra, P., & Ficarra, R. (2004). Combined effect of pH and polysorbates with cyclodextrins on solu-bilization of naringenin. Journal of Pharmaceutical and Biomedical Analysis, 36(2), 327–333. doi:10.1016/j.jpba.2004.06.013

Tozuka, Y., Kishi, J., & Takeuchi, H. (2010). Anomalous dissolution prop-erty enhancement of naringenin from spray-dried particles with a-glu-cosylhesperidin. Advanced Powder Technology, 21(3), 305–309. doi:10. 1016/j.apt.2009.12.013

Tu, B., Liu, Z.-J., Chen, Z.-F., Ouyang, Y., & Hu, Y.-J. (2015). Understanding the structure-activity relationship between quercetin and naringenin: _In vitro [10.1039/C5RA22551E]. RSC Advances, 5(128), 106171–106181. doi:10.1039/C5RA22551E

Tu, B., Wang, Y., Mi, R., Ouyang, Y., & Hu, Y.-J. (2015). Evaluation of the interaction between naringenin and human serum albumin: Insights from fluorescence spectroscopy, electrochemical measurement and molecular docking. Spectrochimica Acta Part A: Molecular and Biomolecular Spectroscopy, 149, 536–543. doi:10.1016/j.saa.2015.04.087

T€urkkan, B., €Ozy€urek, M., Bener, M., G€uc¸l€u, K., & Apak, R. (2012). Synthesis, characterization and antioxidant capacity of naringenin-oxime. Spectrochimica Acta Part A: Molecular and Biomolecular Spectroscopy, 85(1), 235–240. doi:10.1016/j.saa.2011.09.066

Ukhatskaya, E. V., Kurkov, S. V., Matthews, S. E., El Fagui, A., Amiel, C., Dalmas, F., & Loftsson, T. (2010). Evaluation of a cationic calix [4] arene: Solubilization and self-aggregation ability. International Journal of Pharmaceutics, 402(1–2), 10–19. doi:10.1016/j.ijpharm.2010.09.011

Valand, N. N., Patel, M. B., & Menon, S. K. (2015). Curcumin-p-sulfonatoca-lix [4] resorcinarene (p-SC [4] R) interaction: Thermo-physico chemis-try, stability and biological evaluation. RSC Advances, 5(12), 8739–8752. doi:10.1039/C4RA12047G

White, N. S., & Errington, R. J. (2005). Fluorescence techniques for drug delivery research: Theory and practice. Advanced Drug Delivery Reviews, 57(1), 17–42. doi:10.1016/j.addr.2004.08.003

Williams, R. J., Spencer, J. P., & Rice-Evans, C. (2004). Flavonoids: Antioxidants or signalling molecules? Free Radical Biology & Medicine, 36(7), 838–849. doi:10.1016/j.freeradbiomed.2004.01.001

Yadav, A., Kumar, R., Sunkaria, A., Singhal, N., Kumar, M., & Sandhir, R. (2016). Evaluation of potential flavonoid inhibitors of glyoxalase-I based on virtual screening and in vitro studies. Journal of Biomolecular Structure & Dynamics, 34(5), 993–1007. doi:10.1080/ 07391102.2015.1064830

Yang, L., Ran, X., Cai, L., Li, Y., Zhao, H., & Li, C.-P. (2016). Calix[8]arene functionalized single-walled carbon nanohorns for dual-signalling electrochemical sensing of aconitine based on competitive host-guest recognition. Biosensors & Bioelectronics, 83, 347–352. doi:10.1016/j. bios.2016.04.079

(14)

Yang, L., Xie, X., Cai, L., Ran, X., Li, Y., Yin, T., … Li, C.-P. (2016). p-sulfo-nated calix[8]arene functionalized graphene as a“turn on” fluorescent sensing platform for aconitine determination. Biosensors & Bioelectronics, 82, 146–154. doi:10.1016/j.bios.2016.04.005

Yang, L., Zhao, H., Li, Y., Ran, X., Deng, G., Xie, X., & Li, C.-P. (2015). Fluorescent detection of tadalafil based on competitive host-guest ınteraction using p-sulfonated calix[6]arene functionalized graphene. ACS Applied Materials & Interfaces, 7(48), 26557–26565. doi:10.1021/ acsami.5b07833

Yang, L., Zhao, H., Li, Y., Ran, X., Deng, G., Zhang, Y., … Li, C.-P. (2016). Indicator displacement assay for cholesterol electrochemical sensing using a calix[6]arene functionalized graphene-modified electrode. The Analyst, 141(1), 270–278. doi:10.1039/c5an01843a

Yang, Y.-W. (2011). Towards biocompatible nanovalves based on meso-porous silica nanoparticles. MedChemComm, 2(11), 1033–1049. doi:10. 1039/c1md00158b

Yilmaz, M., Karanastasis, A. A., Chatziathanasiadou, M. V., Oguz, M., Kougioumtzi, A., Clemente, N., … Mavromoustakos, T. (2019). Inclusion of quercetin in gold nanoparticles decorated with supra-molecular hosts amplifiesıts tumor targeting properties. ACS Applied Bio Materials, 2(7), 2715. doi:10.1021/acsabm.8b00748

Yilmaz, M., & Sayin, S. (2016). Calixarenes in organo and biomimetic catalysis. Calixarenes and beyond (pp. 719–742). Switzerland: Springer.

Zhao, G., Ran, X., Zhou, X., Tan, X., Lei, H., Xie, X., … Du, G. (2018). Green synthesis of hydroxylatopillar[5]arene-modified gold nanopar-ticles and their self-assembly, sensing, and catalysis applications. ACS Sustainable Chemistry & Engineering, 6(3), 3938–3947. doi: 10.1021/acs-suschemeng.7b04292

Zhou, X., Yang, L., Tan, X., Zhao, G., Xie, X., & Du, G. (2018). A robust electrochemical immunosensor based on hydroxyl pillar[5]arene@AuNPs@g-C3N4 hybrid nanomaterial for ultrasensitive detection of prostate specific antigen. Biosensors & Bioelectronics, 112, 31–39. doi:10.1016/j.bios.2018.04.036

Zhou, X., Zhao, G., Chen, M., Gao, W., Zhou, X., Xie, X., … Du, G. (2018). Facile and green approach to prepare nanostructured Au@ MnO2 and its applications for catalysis and fluorescence sensing of glutathi-one in human blood. ACS Sustainable Chemistry & Engineering, 6(3), 3948–3956. doi:10.1021/acssuschemeng.7b04313

Zhou, Y., Li, H., & Yang, Y.-W. (2015). Controlled drug delivery systems based on calixarenes. Chinese Chemical Letters, 26(7), 825–828. doi:10. 1016/j.cclet.2015.01.038

Zhu, J., Sun, X., Wang, S., Xu, Y., & Wang, D. (2017). Formation of nano-complexes comprising whey proteins and fucoxanthin: Characterization, spectroscopic analysis, and molecular docking. Food Hydrocolloids, 63, 391–403. doi:10.1016/j.foodhyd.2016.09.027

Zuo, C.-S., Wiest, O., & Wu, Y.-D. (2011). Structures and conformations of heteroatom-bridged calixarenes. Journal of Physical Organic Chemistry, 24(12), 1157–1165. doi:10.1002/poc.1840

Şekil

Figure 1 represents the schematic rout for the synthesis of compound 5 according to the previously reported method.
Figure 2. FTIR spectra of compound 5 (a), naringenin (b) and 5-naringenin complex (c).
Figure 4. UV –visible spectra (a) and fluorescence response (b) of 5, naringenin and 5-naringenin complex.
Figure 8. Fluorescence images and their corresponding bright-field transmission images: Bright-field (right panels, (aand d w ), DLD-1 cells incubated with 5-narin- 5-narin-genin complex (10 mM) (middle, (b and e w )) and overlap of images of bright fiel
+4

Referanslar

Benzer Belgeler

In spite of attempts to use the ionization nonlinearity for spectral broadening of pulses at a multi-mJ energy level [7,8], SPM in gases is still the most controllable and reliable

No Arcobacter was isolated when the de Boer method was used for minced meat samples but the same five meat samples were found positive for arcobacters when CAT-supplemented media

Recently, Gurler (1996) studied a non- parametric estimator for the bivariate distribution function when a component is subject to left truncation; this study..

Given a directed graph G(V, E) with vertex set V and directed edge set E representing the users and the connections, respectively, of the social network, and the number of

First, a generic Cramer- Rao lower bound (CRLB) expression is obtained in the case of unknown channel coefficients and carrier-frequency offsets (CFOs) for cognitive radio systems

The study included 60 consecutive patients with unilateral middle one-third tibial fractures treated by anatomic reduc- tion and interlocking intramedullary nail fixation and who

Newton’un da (2001) dikkat çektiği üzere, güvenin çeşitli formlarının bulunması ve güven araştırmalarında, toplumun güven düzeyinin sadece genel güven

olarak el parmaklarının hareketinde kayıp oluşan hastaların rehabilitasyonu için eksik tahrikli bir parmak eksoskeletonu (dış iskeleti) tasarlanarak