1,590
Views
97
CrossRef citations to date
0
Altmetric
Research Article

Synthesis of some tetrahydropyrimidine-5-carboxylates, determination of their metal chelating effects and inhibition profiles against acetylcholinesterase, butyrylcholinesterase and carbonic anhydrase

, , , , , , & show all
Pages 1531-1539 | Received 22 Jan 2016, Accepted 08 Feb 2016, Published online: 28 Mar 2016

Abstract

2-(Methacryloyloxy)ethyl 6-methyl-2-oxo-4-phenyl-1,2,3,4-tetrahydropyrimidine-5-carboxylate, is a cyclic urea derivative synthesized from urea, 2-(methacryloyloxy) ethyl acetoacetate and substituted benzaldehyde, and tested in terms of the inhibition of two physiologically relevant carbonic anhydrase (CA) isozymes I and II. Acetylcholinesterase (AChE) is found in high concentrations in the red blood cells and brain. Butyrylcholinesterase (BChE) is another enzyme abundantly present in the liver and released into blood in a soluble form. Also, they were tested for the inhibition of AChE and BChE enzymes and demonstrated effective inhibition profiles with Ki values in the range of 429.24–530.80 nM against hCA I, 391.86–530.80 nM against hCA II, 68.48–97.19 nM against AChE and 104.70–214.15 nM against BChE. On the other hand, acetazolamide clinically used as CA inhibitor, showed Ki value of 281.33 nM against hCA I, and 202.70 nM against hCA II. Also, Tacrine inhibited AChE and BChE showed Ki values of 396.03 and 209.21 nM, respectively.

Introduction

The pyrimidinethiones display many pharmacological properties, as part of our interest in this kind of materials; we report here the synthesis and crystal structure determination of the title compoundCitation1. Our synthesis is based on the Bidjinelli reaction, which consists of a three-component condensation of an aldehyde, a methylene active compound and a urea derivative in acidic media. This procedure is the most simple and useful for the preparation of 3,4-dihydropyrimidene-2(1H) onesCitation2–4.

New cyclic ureas were obtained by continuing researches in the field of the synthesis of various classes of organic nitrogen compounds and the study of their transformations. So for the first time 2-(methacryloyloxy)ethyl 6-methyl-2-oxo-4-phenyl-1,2,3,4-tetrahydropyrimidine-5-carboxylate was obtained by us based on the trifluoroacetic acid (TFAA) catalyst.

Transition metal ions are key elements in various biological processes ranging from oxygen formation to hypoxia sensing. Their homeostasis is maintained within strict limits through tightly regulated mechanisms of uptake, storage and secretionCitation5–7. The breakdown of metal ion homeostasis can lead to an uncontrolled formation of reactive oxygen species (ROS) via Fenton reaction, which produces hydroxyl radicals (OH˙)Citation8–11.

ROS can cause oxidative damage to biological macromolecules such as DNA, carbohydrate, lipids and proteins. An imbalance between the formation of ROS and their elimination by antioxidant defence systems is termed as oxidative stressCitation12–14. Most vulnerable to free radical attack is the cell membrane, which may undergo enhanced lipid peroxidation, finally producing mutagenic and carcinogenic malondialdehyde (MDA), 4-hydroxynonenal and other exocyclic DNA adductsCitation15–17. Among the transition metal ions, iron is known as the most important lipid oxidation pro-oxidant due to its high reactivity. Iron is an essential mineral for normal physiology, but excess can result in cellular injuryCitation18–20. Ferrous iron (Fe2+) can facilitate the production of ROS within animal and human systems and the ability of substances to chelate iron can be valuable for antioxidant propertyCitation21–23. The effective Fe2+ chelating may also afford protection against oxidative damage by removing iron that may otherwise participate in HO˙ generating Fenton-type reactions. Iron, in nature, can be found as either Fe2+ or ferric ion (Fe3+), with the latter form predominant in foods. Fe3+ also produces radicals from peroxides although the rate is tenfold less than that of Fe2+Citation24–26.

The carbonic anhydrases (CAs, EC 4.2.1.1) are the metalloenzymes containing zinc ions (Zn2+), which classically participate in the maintenance of pH homeostasis. CAs catalyse the reversible hydration of CO2 in two-step reaction to yield bicarbonate (HCO3) ion and H+Citation27,Citation28,Citation29. The interconversion of these chemical species is shown in following equation, which however is too slow to meet the physiological needs of most biochemical processesCitation30–32.

CAs have six genetically distinct enzyme families: the α-, β-, γ-, δ-, ζ- and η-CA. Mammals generally contain α-CAs, which is the best studied CA family. α-CAs exist in 16 isoforms identified in various tissues and organs and differ in their cellular localization (cytosol, mitochondria or cell membrane), sensibility to inhibitors and catalytic activityCitation33–35. Also, this CA family has molecular kinetic and different properties, oligomeric rearrangements and expression levels, as well as various abilities to respond to different inhibitory classesCitation36–38. According to the known cellular localization, some of them are cytosolic (CA I, CA II, CA III, CA VII and CA XIII), some other CA isoenzymes are membrane bound (CA IV, CA IX, CA XII and CA XIV), two of CAs are mitochondrial (CA VA and CA VB) and one of CAs is salivary (CA VI)Citation39–42. CA XV is not synthesized in humans and other primates and is abundantly found in rodents and other vertebrates as an isoform. Three acatalytic forms are also reported and named as CA related proteins (CARPs), CARP VIII, X and XI, which are found in the cytosolCitation43–45. The two important CA isozymes (CA I and CA II) are present at higher concentrations in the cytosol in erythrocytes. hCA I and II have various medical applications and shows optimal activity at physiological pH and temperaturesCitation46–48.

CAs are well established therapeutic targets for treatment of a wide range of disorders through the use of inhibitors or activators, as well as recognized tools for drug delivery purposesCitation49–51. CA inhibitors (CAIs) have many clinical usages of major diseases such as diuretics, antiglaucoma, gastroduodenal ulcers, antiobesity drugs, acid-base disequilibria and antiepileptic. CAIs are useful for the treatment of some neurological disorders such as idiopathic intracranial hypertensionCitation52–54. The design of CAIs as therapeutic agents is related to the large number of isoforms in humans, their rather diffuse localization in many tissues or organs and the lack of isoenzyme selectivity of the presently available inhibitorsCitation55–59.

Acetylcholinesterase (AChE, E.C.3.1.1.7) enzyme, a serine hydrolase, is responsible for the degradation of ACh in the synaptic cleft of cholinergic synapses and neuromuscular junctions into inactive metabolites such as choline and acetateCitation9,Citation60,Citation61. It has essential role in regulating many vital functions such as memory, learning, cortical organization of movement and cerebral blood flow control which demonstrates the high degree of importance of ACh as a neurotransmitter target for the study of cerebrovascular diseases associated with hypertensionCitation60,62–64. On the other hand, butyrylcholinesterase (BChE, E.C. 3.1.1.8) has a higher activity in liver, intestine, heart, lung and kidney. AChE and BChE share 65% amino acid sequence homology and have similar molecular forms and active sites despite being products of different genes on the human chromosomes. Both cholinesterases participate in cholinergic neurotransmission by hydrolysing ACh in the central and peripheral nervous system. Also, they play an important role in the development of Alzheimer’s disease (AD)Citation65. AD is a neurodegenerative disease of the central nervous system associated with progressive memory loss resulting in dementiaCitation66. Gradual loss of ACh has been demonstrated to impair memory, especially in the progression of ADCitation67. The increased AChE reactivity has been shown to be closely associated with neurofibrillary tangle pathology in AD. Also, AChE inhibitors are thought to be promising therapeutic drugs for the treatment of neurodegenerative disease characterized with ACh deficiency, such as senile dementia or ADCitation67,Citation68.

The aim of this study is to design and synthesize some novel tetrahydropyrimidine-5-carboxylates (13) and to investigate their metal chelating, inhibition potential of CA I and II isoenzymes, and to find out the most potent and favourable AChE and BChE inhibition properties of the compounds to give directions to further studies.

Experimental

Chemistry

Synthesis of 2-(methacryloyloxy)ethyl 6-methyl-2-oxo-4-phenyl-1,2,3,4-tetrahydropyrimidine-5-carboxylate (1)

Urea (1.20 g, 0.02 mol) is dissolved in acetylacetone and ethyl alcohol (3:1 mL:mL). Then, 2-(methacryloyloxy)ethyl acetoacetate (3.82 mL, 0.02 mol) is added on it drop by drop. After being dissolved in magnetic stirrer for 5 min, benzaldehyde (2.03 mL, 0.02 mol) is added. After determining that the reaction has been fully completed, the solvent is evaporated. Processing of the reaction mixture was carried out by washing the reaction mixture with ice water, the precipitate was filtered, washed with 0.5 L of water. Finally, it was dried and recrystallized from ethanol (75 mL). The yield is 2.4 g, mp. 211 °C. Eluent – ethanol:hexane (5:2). 1H NMR (300 MHz, DMSO-d6) 5 1.35 (s, 3H, CH3), 6.8–7.1 (m, H, Ar), 7.4 (m, H, Ar), 9.35 (s, 1H, NH). 13C NMR (75 MHz, DMSO-d6) 5 24, 29, 37, 51, 86, 117, 122, 129, 132, 141, 151, 205 (C=O).

Synthesis of allyl 1-(3-choloro-2-hydroxypropyl)-4-methyl-6-phenyl-2-thioxo-1,2,3,6-tetrahydropyrimidine-5-carboxylate (2)

Allyl 6-methyl-4-phenyl-2-thioxo-1,2,3,4-tetrahydropyrimidine-5-carboxylate (0.908 g, 3.3 mmol) is dissolved in 2:1 ratio of acetylacetone and ethyl alcohol (10 mL:5 mL). Then, epichlorohydrin (0.26 mL, 3.3 mmol) is added on it drop by drop. After being dissolved in the stirrer for 25 min, 0.03 g AlCl3 catalyst is added to it and mixed by heating at 65–70 °C. The progress of the reaction is controlled by Sulifol UV 254 plate. After determining the full completion of reaction, solution is evaporated and is cleansed in ethyl alcohol solution. The white crystalline having melting temperature of 168 °C is obtained. Eluent – ethanol:hexane (5:2). 1H NMR (300 MHz, DMSO-d6) 2.30 (s, 3H, CH3), 7.06–7.25 (m, H, Ar), 4.81 (H, OH), 9.84 (s, 1H, NH), 3.40, 3.65 (2H, CH2). 13C NMR (75 MHz, DMSO-d6) 15.5, 49.5, 56.3, 67.8, 71.2, 74.5, 106, 116.4, 127.1, 128.6, 133.5, 138, 158.8, 167.2, 178.4.

Synthesis of ethyl 1-(2-hydroxybutyl)-4-methyl-6-phenyl-2-thioxo-1,2,3,6-tetrahydropyrimidine-5-carboxylate (3)

Ethyl 6-methyl-2-thioxo-4-(p-tolyl)-1,2,3,4-tetrahydropyrimidine-5-carboxylate (1.52 g, 0.02 mol) is dissolved in 2:1 ratio of acetylacetone and ethyl alcohol (12 mL:5 mL) and 1,2-epoxobutane (2.03 mL, 0.02 mol) is added on it drop by drop. After being dissolved in the stirrer for 30 min, 0.02 g AlCl3 catalyst is added on it and mixed by heating at 60–65 °C. The progress of the reaction is controlled by Sulifol UV 254 plate. After determining the full completion of reaction, solution is evaporated and is cleansed in ethyl alcohol solution. The white crystalline having melting temperature of 192 °C is obtained. Eluent – ethanol:hexane (5:2). 1H NMR (300 MHz, DMSO-d6) 0.96 (s, 3H, CH3), 4.59–7.25 (m, 6H, Ar), 4.81 (H, OH), 3.45 (1H, CH), 9.84 (s, 1H, NH), 3.40, 1.48 (2H, CH2). 13C NMR (75 MHz, DMSO-d6) 14.2, 28.6, 58.9, 61.7, 71.2, 71.6, 104.2, 127.1, 127.9, 128.6, 129.9, 138, 160.3, 167.2, 178.

Biochemical studies

For determination of inhibition effects of tetrahydropyrimidine-5-carboxylates (13) on CA I and II isoforms, both isoenzymes were purified from fresh human erythrocyte using affinity chromatography technique as the first experimental workCitation69,Citation70. To this end, Sepharose-4B-L-Tyrosine-sulfanilamide affinity chromatography was used for purification of both isoenzymesCitation71 via a single step described previouslyCitation72–74.

CA activity determination was realised spectrophotometric method of Verpoorte et al.Citation75 as described previouslyCitation76. In this method, changes in absorbance were recorded during 3 min at 25 °C. P-nitrophenylacetate (NPA) was used as substrate and converted by both isoenzymes to p-nitrophenolate ion. These activity determinations are described in detail in our previous studiesCitation77.

For determination of protein quantity, Bradford method was used during the purification stepsCitation78. This spectrophotometrical protein determination was explained previouslyCitation79. The bovine serum albumin was the standard for this determination which was done at 595 nmCitation80.

After the purification process of the CA isoenzymes, SDS-polyacrylamide gel electrophoresis (SDS-PAGE) has been carried out. Stacking gel containing (10 and 3%) acrylamide and (0.1%) of SDSCitation81 was used for running the process using a Minigel system (Mini-PROTEAN Tetra System). The method used for visualization of protein has been explained in detail in previous studiesCitation82. According to this method, the gel was fixed, then stained with Coomassie Brilliant Blues R-250, later on the gel was de-stained by using standard methods for detecting protein bands that belong to purified CA isoenzymesCitation83.

The inhibitory effects of tetrahydropyrimidine-5-carboxylates (13) on AChE/BChE activities were measured according to spectrophotometric method of Ellman et al.Citation84. Acetylthiocholine iodide (AChI) or butyrylthiocholine iodide (BChI) was used as substrates for the reaction. 5,5′-Dithio-bis(2-nitro-benzoic)acid (DTNB, D8130-1G, Sigma-Aldrich, Steinheim, Germany) was used for the measurement of the AChE/BChE activities. Briefly, 100 mL of Tris/HCl buffer (1 M, pH 8.0), 10 mL of sample solution dissolved in deionized water at different concentrations and 50 mL AChE/BChE solution were mixed and incubated for 10 min at 25 °C. Then 50 mL of DTNB (0.5 mM) was added. The reaction was then initiated by the addition of 50 mL of AChI/BChI. The hydrolysis of these substrates was monitored spectrophotometrically by formation of the yellow 5-thio-2-nitrobenzoate anion as the result of the reaction of DTNB with thiocholine, released by enzymatic hydrolysis of AChI/BChI, with an absorption maximum at a wavelength of 412 nmCitation85,Citation86.

The effect of novel tetrahydropyrimidine-5-carboxylates (13) on both CA isoenzymes was examined using the hydratase activity and recorded in triplicate analysis at the each used concentrationCitation87. For this purpose, different concentrations of tetrahydropyrimidine-5-carboxylates (13) were determined in preliminary assays. CA isoenzyme activities were measured in the presence of different quantities of them. The control sample activity in the absence of a tetrahydropyrimidine-5-carboxylates (13) was taken as 100%. For each tetrahydropyrimidine-5-carboxylates (13), an activity (%) – [Tetrahydropyrimidine-5-carboxylates] was drawn using conventional polynominal regression software. The half maximal inhibitory concentration (IC50) of all tetrahydropyrimidine-5-carboxylates (13) was calculated from graphsCitation88. IC50 values are measures of the effectiveness of novel tetrahydropyrimidine-5-carboxylates (13) in inhibiting both CA isoenzymes. For determination of Ki values, three different tetrahydropyrimidine-5-carboxylate (13) concentrations were used. Ki values reflect the binding affinity of the tetrahydropyrimidine-5-carboxylates (13) to both CA isoenzymes. In this way, the IC50 value is converted to an absolute inhibition constant Ki value. In this experiment, PNA was used as substrate at five different concentrations. Finally, Lineweaver–Burk curves were drawn for each inhibitorCitation89.

Fe2+ chelating ability of tetrahydropyrimidine-5-carboxylates (13) was predicted according to Dinis et al.Citation90 with slight modificationCitation91,Citation92. Fe2+-binding capacity of tetrahydropyrimidine-5-carboxylates (13) was spectrophotometrically recorded at 522 nmCitation93. In brief, to a mixture of FeCl2 (0.1 mL, 0.6 mM), tetrahydropyrimidine-5-carboxylates (13) were added at three different concentrations (10–30 μg/mL) in methanol (0.4 mL). The reactions were started by pipyrdyl solution addition (0.1 mL, 5 mM). After that, the solution was mixed and incubated at room temperature for 10 min. Finally, absorbance value of the mixture was measured spectrophotometrically at 522 nmCitation94.

Results and discussion

Compounds characterization

The synthesis of the new compounds is shown in Scheme 1. The reaction of substituted benzaldehyde with methylene active compounds such as 2-(methacryloyloxy)ethyl acetoacetate and urea in the presence of TFAA led to the desired cyclic urea. At the next stage, we have provided the transformation of obtained compounds. So, by the reaction epichlorohydrin had synthesised allyl 1-(3-choloro-2-hydroxypropyl)-4-metil-6-phenyl-2-thioxo-1,2,3,6-tetrahydropyrimidine-5-carboxylate. At the same time by the reaction 1,2-epoxobutane synthesised ethyl 1-(2-hydroxybutyl)-4-methyl-6-phenyl-2-thioxo-1,2,3,6-tetrahydropyrimidine-5-carboxylate.

In the compound, the C8, C9, C11 and C12 atoms of the phenyl ring are disordered over two sets of sites in a 0.60 (3):0.40 (3) ratio. The heterocyclic ring is essentially planar and forms a dihedral angle with the phenyl ring. The crystal packing is stabilized by intermolecular N3—H3N···O1 hydrogen bonds, which link the molecules into chains running parallel to the b axis (), with graph-set notation C(6)Citation95. Data collection: APEX2Citation96; cell refinement: SAINT-PlusCitation94; data reduction: SAINT-Plus; program(s) used to solve structure: SHELXTLCitation97; program(s) used to refine structure: SHELXTL; molecular graphics: SHELXTL; software used to prepare material for publication: SHELXTL.

Figure 1. (A) The molecular structure of the new compound. (B) Packing diagram of the title compound. N–H–O hydrogen bonds are shown as dashed lines. For clarity only one of the disordered components of the phenyl ring is shown.

Figure 1. (A) The molecular structure of the new compound. (B) Packing diagram of the title compound. N–H–O hydrogen bonds are shown as dashed lines. For clarity only one of the disordered components of the phenyl ring is shown.

Special details

Geometry

All e.s.d.s (except the e.s.d. in the dihedral angle between two l.s. planes) are estimated using the full covariance matrix. The cell e.s.d.s are taken into account individually in the estimation of e.s.d.s in distances, angles and torsion angles; correlations between e.s.d.s in cell parameters are only used when they are defined by crystal symmetry. An approximate (isotropic) treatment of cell e.s.d.s is used for estimating e.s.d.s involving l.s. planes.

Refinement

Refinement of F2 against ALL reflections. The weighted R-factor wR and goodness of fit S are based on F2, conventional R-factors R are based on F, with F set to zero for negative F2. The threshold expression of F2 > σ(F2) is used only for calculating R-factors (gt) etc. and is not relevant to the choice of reflections for refinement. R-factors based on F2 are statistically about twice as large as those based on F, and R-factors based on ALL data will be even larger.

Biochemical results

Classical CAIs usually act by complexing the zinc ion (Zn2+) in the active site of CA such as primary sulfonamides and their bioisosters, thiophenols, inorganic anions, hydroxamates and carboxylic acidsCitation98. It was reported that some derivatives characterized by different substituents including CH3, CF3, CN, F, Cl, Br, NO2, OCH3 and Ph at the ortho-, meta- and para-positions of the benzylic or benzoylic portion in their skeleton have inhibition profiles against CA isoenzymesCitation99. All the synthesized compounds were tested to evaluate their inhibitory activity towards the slower cytosolic isoform (hCA I), the more rapid cytosolic isoenzyme (hCA II) and AChE/BChE enzymes. The chemical formula of novel tetrahydropyrimidine-5-carboxylates (13) is given in Scheme 1 and their CA I and II isoforms inhibition data are summarized in . Novel tetrahydropyrimidine-5-carboxylates (13) showed effective Fe2+ chelating effects and inhibition profiles against CA isoforms, AChE and BChE enzymes. When examining the results, the following structure–activity relationship could be easily observed:

Scheme 1. The three-component condensation reactions come to an end within 2.5–3 h at 60–75 °C. The synthesized compounds were crystalline and their structure was confirmed by spectral and physico-chemical methods, among which IQ, 1H, 13C NMR and X-ray spectroscopy: The crystal structure of synthesized 2-(methacryloyloxy)ethyl 6-methyl-2-oxo-4-phenyl-1,2,3,4-tetrahydropyrimidine-5-carboxylate has been determined with X-ray structure analysis method. The structure and crystalline form of new cyclical compound has been shown in . Crystalline compound holds two crystallographical independent molecules.

Scheme 1. The three-component condensation reactions come to an end within 2.5–3 h at 60–75 °C. The synthesized compounds were crystalline and their structure was confirmed by spectral and physico-chemical methods, among which IQ, 1H, 13C NMR and X-ray spectroscopy: The crystal structure of synthesized 2-(methacryloyloxy)ethyl 6-methyl-2-oxo-4-phenyl-1,2,3,4-tetrahydropyrimidine-5-carboxylate has been determined with X-ray structure analysis method. The structure and crystalline form of new cyclical compound has been shown in Figure 1. Crystalline compound holds two crystallographical independent molecules.

Table 1. The enzyme inhibition values of some tetrahydropyrimidine-5-carboxylates (13) against human carbonic anhydrase isoenzymes I and II (hCA I and II), acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) enzymes.

  1. To describe inhibitory effects, researchers often use an IC50 value. However, a more suitable measure is the Ki constant. Ki values were calculated from Lineweaver–Burk graphsCitation89. In our study, both Ki and IC50 parameters of the tetrahydropyrimidine-5-carboxylates (13) were determined and are given in . For the cytosolic hCA I isoenzyme, tetrahydropyrimidine-5-carboxylates (13) had Ki values in the range of 429.24 ± 87.89–539.30 ± 106.70 nM (). The most significant inhibition effect was observed by novel tetrahydropyrimidine-5-carboxylate 2 (allyl 1-(3-choloro-2-hydroxypropyl)-4-metil-6-phenyl-2-thioxo-1,2,3,6-tetrahydropyrimidine-5-carboxylate), which possesses biological active groups of –CH2, –CH3, –C=O, –NH, –C=S, Cl and –OH with Ki value of 429.24 ± 87.89 nM. It is well known that these biologically active groups inhibit CA isoenzymes. On the other hand, acetazolamide (AZA), used as a CA inhibitor for the medical treatment of epileptic seizure, glaucoma, idiopathic intracranial hypertension, altitude sickness, cystinuria, periodic paralysis, central sleep apnea and dural ectasia, showed Ki value of 481.33 ± 55.33 nM. The hCA I is highly abundant in red blood cells and is found in many tissues but its precise physiological function is unknown. CA I is associated with cerebral and retinal edema and the inhibition of CA I may be a valuable tool for fighting these conditionsCitation27.

  2. The ubiquitous and physiologically predominant cytosolic isoform hCA II is associated with several diseases. For hCA II, novel tetrahydropyrimidine-5-carboxylates (13) had Ki values in the range of 391.86 ± 40.16–530.80 ± 103.60 nM. On the other hand, AZA used as a clinical CA inhibitor demonstrated Ki value of 202.70 ± 162.5 nM. As can be seen in CA I, similar to CA I, the most significant inhibition effect was observed by tetrahydropyrimidine-5-carboxylates 2 (allyl 1-(3-choloro-2-hydroxypropyl)-4-metil-6-phenyl-2-thioxo-1,2,3,6-tetrahydropyrimidine-5-carboxylate), with Ki values of 391.86 ± 40.16 nM.

  3. AChE is a metabolic serine hydrolase that catalyses the hydrolysis of ACh, thus regulating cholinergic neurotransmission. Therefore, in disorders such as AD, where there is diminished cholinergic activity, inhibition of AChE has been employed to treat some of the symptoms attributed to decreased ACh levelsCitation100,Citation101. Effective AChE and BChE inhibitors can be used for AD treatment. Most of the currently available drugs on the market including Tacrine, Rivastigmine, Donepezil and Galantamine intended to treat AD are AChE and BChE inhibitorsCitation100–102. However, during the progression of AD, brain AChE levels decline while BChE activity increases, suggesting that ACh hydrolysis may occur to a greater extent via BChE catalysis. In this regard, it has been reported that highly selective inhibition of BChE is important in raising ACh levels and improving cognitionCitation103. The inhibition effects of novel tetrahydropyrimidine-5-carboxylates (13) against AChE/BChE activities were measured according to spectrophotometric method described by Ellman et al.Citation84 Acetylthiocholine iodide/butyrylthiocholine iodide (AChI/BChI) was used as substrates for the reactions. This method is based on the amount of thiocholine released when the AChE/BChE hydrolyses the substrate AChI/BChI to thiocholine and acetate/butyrate. The product thiocholine reacts with Ellman’s reagent (5,5-bisdithionitrobenzoic acid-DTNB) to produce a yellow compound (5-thio-2-nitrobenzoate) anion, which can be detected in a wavelength of 412 nm. AChE and BChE were very effectively inhibited by novel tetrahydropyrimidine-5-carboxylates (13). It was found that Ki values were in the range of 68.48 ± 25.07–97.19 ± 19.62 nM for AChE and 104.70 ± 8.093–214.15 ± 28.77 nM for BChE, respectively (). On the other hand, Tacrine (TAC) was used as standard AChE and BChE inhibitor it had Ki values of 396.03 ± 30.66 and 209.21 ± 15.02 nM, respectively. In addition, donepezil hydrochloride, which is used for the treatment of mild-to-moderate AD and various other memory impairments, had been shown to lower AChE inhibition activity (IC50: 55.0 nM). The Ki values of novel tetrahydropyrimidine-5-carboxylates (13) for AChE and BChE were calculated from Lineweaver–Burk plotsCitation89.

  4. Finally, novel tetrahydropyrimidine-5-carboxylates (13) had also effective Fe2+ chelating effect. The distinction between different concentrations of novel tetrahydropyrimidine-5-carboxylates (13) (10–30 μg/mL) and the control value was fixed to be statistically important (p < 0.01). Furthermore, it is found that IC50 values for novel tetrahydropyrimidine-5-carboxylates (13) were found as 46.21, 53.30 and 45.89 μg/mL, respectively (). Whereas, IC50 values belonging to Fe2+ chelating capacity of positive controls like trolox, α-tocopherol, BHT, BHA and EDTA was found to be 19.80, 36.47, 23.89, 30.13 and 16.90 μg/mL, respectively. A lower IC50 value reflects a higher Fe2+ binding activity. These results clearly introduce that Fe2+ chelating effect of novel tetrahydropyrimidine-5-carboxylates (13) was close to trolox, α-tocopherol, BHT, but lower than that of other standards. Fe2+ ions are the most efficient pro-oxidants in pharmacology systems and foodCitation104–106. Ferrozine can create complexes with Fe2+. In the presence of Fe2+ chelating compounds, Ferrozine–Fe2+ complex formation is broken down, resulting in a decrease in the red colour of Ferrozine–Fe2+ complexCitation107–109.

Table 2. Determination of half maximal concentrations (IC50) of Fe2+ chelating of some tetrahydropyrimidine-5-carboxylates (13) and standard compounds.

Conclusion

Novel tetrahydropyrimidine-5-carboxylates (13) used in the present study demonstrated effective inhibition profiles against hCA isoforms, AChE and BChE enzymes. Additionally, these compounds demonstrated effective metal chelating properties. Novel tetrahydropyrimidine-5-carboxylates (13) identified their potential CA isoenzymes, and AChE and BChE enzyme inhibitors. In this study, nanomolar level of Ki values was observed for all novel tetrahydropyrimidine-5-carboxylates and these compounds can be selective inhibitor of both cytosolic CA isoenzymes and AChE and BChE enzymes. Also, they can be used as metal chelators in related applications.

Acknowledgements

I.G. and S.H.E. would like to extend their sincere appreciation to the Distinguished Scientist Fellowship Program at King Saud University for supporting this research.

Declaration of interest

The authors declare no conflict of interest.

References

  • Dalinger D, Stadler A, Kappe CO. Solid- and solution-phase synthesis of bioactive dihydropyrimidines. Pure Appl Chem 2004;76:1017–24
  • Kotharkar SA, Nagawade RR, Shinde DB. Chlorosulfonic acid catalyzed highly efficient solvent-free synthesis of 3,4-dihydropyr-imidin-2(1IBHD)-ones and thiones. Ukrain Bioorg Acta 2006;4:17–21
  • Lu J, Bai Y, Wang Z, et al. One-pot synthesis of 3,4-dihydropyr-imidin-2(1H)-ones using lanthanum chloride as a catalyst. Tetrahedron Lett 2000;41:9075–8
  • Salehi P, Davizi M, Zolfigol AM, Fard MA. Silica sulfuric acid: an efficient and reusable catalyst for the one-pot synthesis of 3,4-dihydropyrimidin-2(1H)-ones. Tetrahedron Lett 2003;44:2889–91
  • Öztaşkın N, Çetinkaya Y, Taslimi P, et al. Antioxidant and acetylcholinesterase inhibition properties of novel bromophenol derivatives. Bioorg Chem 2015;60:49–57
  • Kalın P, Gülçin İ, Gören AC. Antioxidant activity and polyphenol content of Vaccinium macrocarpon. Rec Nat Prod 2015;9:496–502
  • Sehitoglu MH, Han H, Kalin P, et al. Pistachio (Pistacia vera L.) Gum: a potent inhibitor of reactive oxygen species. J Enzyme Inhib Med Chem 2015;30:264–9
  • Çakmakçı S, Topdaş EF, Kalın P, et al. Antioxidant capacity and functionality of oleaster (Elaeagnus angustifolia L.) flour and crust in a new kind of fruity ice cream. Int J Food Sci Technol 2015;50:472–81
  • Göçer H, Akıncıoğlu A, Öztaşkın N, et al. Synthesis, antioxidant and antiacetylcholinesterase activities of sulfonamide derivatives of dopamine related compounds. Arch Pharm 2013;346:783–92
  • Bursal E, Köksal E, Gülçin İ, et al. Antioxidant activity and polyphenol content of cherry stem (Cerasus avium L.) determined by LC–MS/MS. Food Res Int 2013;51:66–74
  • Gülçin İ, Elmastaş M, Aboul-Enein HY. Antioxidant activity of clove oil – a powerful antioxidant source. Arab J Chem 2012;5:489–99
  • Gülçin İ. Antioxidant activity of food constituents – an overview. Arch Toxicol 2012;86:345–91
  • Çetinkaya Y, Göçer H, Menzek A, Gülçin İ. Synthesis and antioxidant properties of (3,4-dihydroxyphenyl)(2,3,4-trihydroxyphenyl)methanone and its derivatives. Arch Pharm 2012;345:323–34
  • Gülçin İ, Beydemir S, Topal F, et al. Apoptotic, antioxidant and antiradical effects of majdine and isomajdine from Vinca herbacea Waldst. and kit. J Enzyme Inhib Med Chem 2012;27:587–94
  • Bursal E, Gülçin İ. Polyphenol contents and in vitro antioxidant activities of lyophilized aqueous extract of kiwifruit (Actinidia deliciosa). Food Res Int 2011;44:1482–9
  • Gülçin İ, Topal F, Çakmakçı R, et al. Pomological features, nutritional quality, polyphenol content analysis and antioxidant properties of domesticated and three wild ecotype forms of raspberries (Rubus idaeus L.). J Food Sci 2011;76:C585–93
  • Gülçin İ, Topal F, Oztürk Sarikaya SB, et al. Polyphenol contents and antioxidant properties of medlar (Mespilus germanica L.). Rec Nat Prod 2011;5:158–75
  • Köksal E, Bursal E, Dikici E, et al. Antioxidant activity of Melissa officinalis leaves. J Med Plants Res 2011;5:217–22
  • Göçer H, Gülçin İ. Caffeic acid phenethyl ester (CAPE): correlation of structure and antioxidant properties. Int J Food Sci Nut 2011;62:821–5
  • Gülçin İ. Antioxidant activity of eugenol – a structure and activity relationship study. J Med Food 2011;14:975–85
  • Gülçin İ, Huyut Z, Elmastaş M, Aboul-Enein HY. Radical scavenging and antioxidant activity of tannic acid. Arab J Chem 2010;3:43–53
  • Gülçin İ, Bursal E, Şehitoğlu HM, et al. Polyphenol contents and antioxidant activity of lyophilized aqueous extract of propolis from Erzurum, Turkey. Food Chem Toxicol 2010;48:2227–38
  • Gülçin İ, Kirecci E, Akkemik E, et al. Antioxidant and antimicrobial activities of an aquatic plant: Duckweed (Lemna minor L.). Turk J Biol 2010;34:175–88
  • Valko M, Jomova K, Rhodes CJ, et al. Redox‐ and non‐redox‐metal‐induced formation of free radicals and their role in human disease. Arch Toxicol 2016;90:1–37
  • Şerbetçi Tohma H, Gülçin İ. Antioxidant and radical scavenging activity of aerial parts and roots of Turkish liquorice (Glycyrrhiza glabra L.). Int J Food Propert 2010;13:657–71
  • Kehrer JP. The Haber–Weiss reaction and mechanisms of toxicity. Toxicology 2000;149:43–50
  • Taslimi P, Gulcin İ, Ozgeris B, et al. The human carbonic anhydrase isoenzymes I and II (hCA I and II) inhibition effects of trimethoxyindane derivatives. J Enzyme Inhib Med Chem 2016;31:152–7
  • Scozzafava A, Kalın P, Supuran CT, et al. The impact of hydroquinone on acetylcholine esterase and certain human carbonic anhydrase isoenzymes (hCA I, II, IX, and XII). J Enzyme Inhib Med Chem 2015;30:941–6
  • Akıncıoğlu A, Akıncıoğlu H, Gülçin I, et al. Discovery of potent carbonic anhydrase and acetylcholine esterase inhibitors: novel sulfamoylcarbamates and sulfamides derived from acetophenones. Bioorg Med Chem 2015;23:3592–602
  • Bozdag M, Carta F, Vullo D, et al. Dithiocarbamates with potent inhibitory activity against the Saccharomyces cerevisiae b-carbonic anhydrase. J Enzyme Inhib Med Chem 2016;31:132–6
  • Yıldırım A, Atmaca U, Keskin A, et al. N-Acylsulfonamides strongly inhibit human carbonic anhydrase isoenzymes I and II. Bioorg Med Chem 2015;23:2598–605
  • Boztaş M, Çetinkaya Y, Topal M, et al. Synthesis and carbonic anhydrase isoenzymes I, II, IX, and XII inhibitory effects of dimethoxy-bromophenol derivatives incorporating cyclopropane moieties. J Med Chem 2015;58:640–50
  • Scozzafava A, Passaponti M, Supuran CT, Gülçin İ. Carbonic anhydrase inhibitors: guaiacol and catechol derivatives effectively inhibit certain human carbonic anhydrase isoenzymes (hCA I, II, IX, and XII). J Enzyme Inhib Med Chem 2015;30:586–91
  • De Luca V, Del Prete S, Carginale V, et al. A failed tentative to design a super carbonic anhydrase having the biochemical properties of the most thermostable CA (SspCA) and the fastest (SazCA) enzymes. J Enzyme Inhib Med Chem 2015;30:989–94
  • Arabaci B, Gülçin İ, Alwasel S. Capsaicin: a potent inhibitor of carbonic anhydrase isoenzymes. Molecules 2015;19:10103–14
  • Göçer H, Akıncıoğlu A, Göksu S, et al. Carbonic anhydrase and acetylcholine esterase inhibitory effects of carbamates and sulfamoylcarbamates. J Enzyme Inhib Med Chem 2015;30:316–20
  • Arslan M, Şentürk M, Fidan İ, et al. Synthesis of 3,4-dihydroxypyrrolidine-2,5-dione and 3,5-dihydroxybenzoic acid derivatives and evaluation of the carbonic anhydrase I and II inhibition. J Enzyme Inhib Med Chem 2015;30:896–900
  • Akbaba Y, Bastem E, Topal F, et al. Synthesis and carbonic anhydrase inhibitory effects of novel sulfamides derived from 1-aminoindanes and anilines. Arch Pharm 2014;347:950–7
  • Göksu S, Naderi A, Akbaba Y, et al. Carbonic anhydrase inhibitory properties of novel benzylsulfamides using molecular modeling and experimental studies. Bioorg Chem 2014;56:75–82
  • Singasane N, Kharkar PS, Ceruso M, et al. Inhibition of carbonic anhydrase isoforms I, II, IX and XII with Schiff’s bases incorporating iminoureido moieties. J Enzyme Inhib Med Chem 2015;30:901–7
  • Güney M, Coşkun A, Topal F, et al. Oxidation of cyanobenzocycloheptatrienes: synthesis, photooxygenation reaction and carbonic anhydrase isoenzymes inhibition properties of some new benzotropone derivatives. Bioorg Med Chem 2014;22:3537–43
  • Topal M, Gülçin İ. Rosmarinic acid: a potent carbonic anhydrase isoenzymes inhibitor. Turk J Chem 2014;38:894–902
  • Çetinkaya Y, Göçer H, Gülçin İ, Menzek A. Synthesis and carbonic anhydrase isoenzymes inhibitory effects of brominated diphenylmethanone and its derivatives. Arch Pharm 2014;347:354–9
  • Akıncıoğlu A, Topal M, Gülçin İ, Göksu S. Novel sulfamides and sulfonamides incorporating tetralin scaffold as carbonic anhydrase and acetylcholine esterase inhibitors. Arch Pharm 2014;347:68–76
  • Çetinkaya Y, Göçer H, Göksu S, Gülçin İ. Synthesis and carbonic anhydrase isoenzymes inhibitory effects of novel benzylamine derivatives. J Enzyme Inhib Med Chem 2014;29:168–74
  • Akbaba Y, Akıncıoğlu A, Göçer H, et al. Carbonic anhydrase inhibitory properties of novel sulfonamide derivatives of aminoindanes and aminotetralins. J Enzyme Inhib Med Chem 2014;29:35–42
  • Aksu K, Nar M, Tanç M, et al. The synthesis of sulfamide analogues of dopamine related compounds and their carbonic anhydrase inhibitory properties. Bioorg Med Chem 2013;21:2925–31
  • Akıncıoğlu A, Akbaba Y, Göçer H, et al. Novel sulfamides as potential carbonic anhydrase isoenzymes inhibitors. Bioorg Med Chem 2013;21:1379–85
  • Gülçin İ, Beydemir S. Phenolic compounds as antioxidants: carbonic anhydrase isoenzymes inhibitors. Mini Rev Med Chem 2013;13:408–30
  • Supuran CT, Scozzafava A. Carbonic anhydrases as targets for medicinal chemistry. Bioorg Med Chem 2007;15:4336–50
  • Nar M, Çetinkaya Y, Gülçin İ, Menzek A. (3,4-Dihydroxyphenyl)(2,3,4-trihydroxyphenyl)methanone and its derivatives as carbonic anhydrase isoenzymes inhibitors. J Enzyme Inhib Med Chem 2013;28:402–6
  • Supuran CT. Carbonic anhydrases: from biomedical applications of the inhibitors and activators to biotechnological use for CO2 capture. J Enzyme Inhib Med Chem 2013;28:229–30
  • Öztürk Sarıkaya SB, Topal F, Şentürk M, et al. In vitro inhibition of α-carbonic anhydrase isozymes by some phenolic compounds. Bioorg Med Chem Lett 2011;21:4259–62
  • Şentürk M, Gülçin İ, Beydemir Ş, et al. In vitro inhibition of human carbonic anhydrase I and II isozymes with natural phenolic compounds. Chem Biol Drug Des 2011;77:494–9
  • Innocenti A, Gülçin İ, Scozzafava A, Supuran CT. Carbonic anhydrase inhibitors. Antioxidant polyphenol natural products effectively inhibit mammalian isoforms I–XV. Bioorg Med Chem Lett 2010;20:5050–53
  • Innocenti A, Öztürk Sarıkaya SB, Gülçin İ, Supuran CT. Carbonic anhydrase inhibitors. Inhibition of mammalian isoforms I–XIV with a series of natural product polyphenols and phenolic acids. Bioorg Med Chem 2010;18:2159–64
  • Şentürk M, Gülçin İ, Daştan A, et al. Carbonic anhydrase inhibitors. Inhibition of human erythrocyte isozymes I and II with a series of antioxidant phenols. Bioorg Med Chem 2009;17:3207–11
  • Öztürk Sarıkaya SB, Gülçin İ, Supuran CT. Carbonic anhydrase inhibitors. Inhibition of human erythrocyte isozymes I and II with a series of phenolic acids. Chem Biol Drug Des 2010;75:515–20
  • Çoban TA, Beydemir Ş, Gülçin İ, Ekinci D. The inhibitory effect of ethanol on carbonic anhydrase isoenzymes: in vivo and in vitro studies. J Enzyme Inhib Med Chem 2008;23:266–70
  • Akinyemi AJ, Thome GR, Morsch VM, et al. Effect of dietary supplementation of ginger and turmeric rhizomes on ectonucleotidases, adenosine deaminase and acetylcholinesterase activities in synaptosomes from the cerebral cortex of hypertensive rats. J Appl Biomed 2016;14:59–70
  • Aksu K, Topal F, Gülçin I, et al. Acetylcholinesterase inhibitory and antioxidant activities of novel symmetric sulfamides derived from phenethylamines. Arch Pharm 2015;348:446–55
  • Polat Köse L, Gülçin İ, Gören AC, et al. LC–MS/MS analysis, antioxidant and anticholinergic properties of galanga (Alpinia officinarum Hance) rhizomes. Ind Crops Prod 2015;74:712–21
  • Cardoso AM, Bagatini MD, Martins CC, et al. Exercise training prevents ecto-nucleotidases alterations in platelets of hypertensive rats. Mol Cell Biochem 2012;371:147–56
  • Topal M, Gocer H, Topal F, et al. Antioxidant, antiradical and anticholinergic properties of cynarin purified from the illyrian thistle (Onopordum illyricum L.). J Enzyme Inhib Med Chem 2016;31:266–75
  • Ye C, Wang MQ, Zhong X, et al. Highly sensitive electrochemiluminescence assay of acetylcholinesterase activity based on dual biomarkers using Pd–Au nanowires as immobilization platform. Biosen Bioelectron 2016;79:34–40
  • Koukoulitsa C, Villalonga-Barber C, Csonka R, et al. Biological and computational evaluation of resveratrol inhibitors against Alzheimer’s disease. J Enzyme Inhib Med Chem 2016;31:67–77
  • He D, Wu H, Wei Y, et al. Effects of harmine, an acetylcholinesterase inhibitor, on spatial learning and memory of APP/PS1 transgenic mice and scopolamine-induced memory impairment mice. Eur J Pharmacol 2015;768:96–107
  • Inestrosa NC, Alvarez A, Perez CA, et al. Acetylcholinesterase accelerates assembly of amyloid-beta-peptides into Alzheimer's fibrils: possible role of the peripheral site of the enzyme. Neuron 1996;16:881–91
  • Çoban TA, Beydemir Ş, Gülçin İ, Ekinci D. Morphine inhibits erythrocyte carbonic anhydrase in vitro and in vivo. Biol Pharm Bull 2007;30:2257–61
  • Hisar O, Beydemir Ş, Gülçin İ, et al. The effect of melatonin hormone on carbonic anhydrase enzyme activity in rainbow trout (Oncorhynchus mykiss) erythrocytes in vitro and in vivo. Turk J Vet Anim Sci 2005;29:841–5
  • Atasever A, Özdemir H, Gülçin İ, Küfrevioğlu Öİ. One-step purification of lactoperoxidase from bovine milk by affinity chromatography. Food Chem 2013;136:864–70
  • ArasHisar Ş, Hisar O, Beydemir Ş, et al. Effect of vitamin E on carbonic anhydrase enzyme activity in rainbow trout (Oncorhynchus mykiss) erythrocytes in vitro and in vivo. Acta Vet Hung 2004;52:413–22
  • Beydemir Ş, Gülçin İ. Effect of melatonin on carbonic anhydrase from human erythrocyte in vitro and from rat erythrocyte in vivo. J Enzyme Inhib Med Chem 2004;19:193–7
  • Gülçin İ, Beydemir Ş, Büyükokuroğlu ME. In vitro and in vivo effects of dantrolene on carbonic anhydrase enzyme activities. Biol Pharm Bull 2004;27:613–6
  • Verpoorte JA, Mehta S, Edsall JT. Esterase activities of human carbonic anhydrase. J Biol Chem 1967;242:4221–9
  • Hisar O, Beydemir Ş, Gülçin İ, et al. Effect of low molecular weight plasma inhibitors of rainbow trout (Oncorhyncytes mykiss) on human erythrocytes carbonic anhydrase-II isozyme activity in vitro and rat erythrocytes in vivo. J Enzyme Inhib Med Chem 2005;20:35–9
  • Taslimi P, Gülçin İ, Öztaşkın N, et al. The effects of some bromophenol derivatives on human carbonic anhydrase isoenzymes. J Enzyme Inhib Med Chem. 2016. [Epub ahead of print].http://dx.doi.org/10.3109/14756366.2015.1054820
  • Bradford MM. A rapid and sensitive method for the quantitation of protein utilizing the principle of protein dye binding. Anal Biochem 1976;72:248–54
  • Gülçin İ, Küfrevioğlu Öİ, Oktay M. Purification and characterization of polyphenol oxidase from nettle (Urtica dioica L.) and inhibition effects of some chemicals on the enzyme activity. J Enzyme Inhib Med Chem 2005;20:297–302
  • Beydemir Ş, Gülçin İ, Hisar O, et al. Effect of melatonin on glucose-6-phospate dehydrogenase from rainbow trout (Oncorhynchus mykiss) erythrocytes in vitro and in vivo. J Appl Anim Res 2005;28:65–8
  • Köksal E, Gülçin İ. Purification and characterization of peroxidase from cauliflower (Brassica oleracea L.) buds. Protein Peptide Lett 2008;15:320–6
  • Şentürk M, Gülçin İ, Çiftci M, Küfrevioğlu Öİ. Dantrolene inhibits human erythrocyte glutathione reductase. Biol Pharmacol Bull 2008;31:2036–9
  • Gülçin İ, Beydemir Ş, Çoban TA, Ekinci D. The inhibitory effect of dantrolene sodium and propofol on 6-phosphogluconate dehydrogenase from rat erythrocyte. Fresen Environ Bull 2008;17:1283–7
  • Ellman GL, Courtney KD, Andres V, Featherston RM. A new and rapid colorimetric determination of acetylcholinesterase activity. Biochem Pharmacol 1961;7:88–95
  • Gülçin İ, Scozzafava A, Supuran CT, et al. Rosmarinic acid inhibits some metabolic enzymes including glutathione S-transferase, lactoperoxidase, acetylcholinesterase, butyrylcholinesterase, and carbonic anhydrase isoenzymes. J Enzyme Inhib Med Chem. 2016. [Epub ahead of print].http://dx.doi.org/10.3109/14756366.2015.1135914
  • Gülçin İ, Scozzafava A, Supuran CT, et al. The effect of caffeic acid phenethyl ester (CAPE) metabolic enzymes including acetylcholinesterase, butyrylcholinesterase, glutathione S-transferase, lactoperoxidase and carbonic anhydrase ısoenzymes I, II, IX and XII. J Enzyme Inhib Med Chem 2016. [Epub ahead of print]. http://dx.doi.org/10.3109/14756366.2015.1094470
  • Gülçin İ, Yıldırım A. Purification and characterization of peroxidase from Brassica oleracea var. Acephala. Asian J Chem 2005;17:2175–83
  • Şişecioğlu M, Çankaya M, Gülçin İ, Özdemir M. The inhibitory effect of propofol on lactoperoxidase. Protein Peptide Lett 2009;16:46–49
  • Lineweaver H, Burk D. The determination of enzyme dissociation constants. J Am Chem Soc 1934;56:658–66
  • Dinis TCP, Madeira VMC, Almeida LM. Action of phenolic derivatives (acetoaminophen, salycilate, and 5-aminosalycilate) as inhibitors of membrane lipid peroxidation and as peroxyl radical scavengers. Arch Biochem Biophys 1994;315:161–9
  • Ak T, Gülçin İ. Antioxidant and radical scavenging properties of curcumin. Chem Biol Interact 2008;174:27–37
  • Gülçin İ, Elias R, Gepdiremen A, et al. Antioxidant secoiridoids from fringe tree (Chionanthus virginicus L.). Wood Sci Technol 2009;43:195–212
  • Talaz O, Gülçin İ, Göksu S, Saracoglu N. Antioxidant activity of 5,10-dihydroindeno[1,2-b]indoles containing substituents on dihydroindeno part. Bioorg Med Chem 2009;17:6583–9
  • Bruker. APEX2. Madison, Wisconsin, USA: Bruker AXS; 2005
  • Bernstein J, Davis RE, Shimoni L, Chang NL. Hydrogen-bond pattern functionality and graph sets. Angew Chem Int Ed Engl 1995;34:1555–73
  • Bruker. SAINT-Plus. Madison, Wisconsin, USA: Bruker AXS; 2001
  • Srinivas KV, Das B. Iodine catalyzed one-pot synthesis of 3,4-dihydropyrimidin-2(1h)-ones and thiones: a simple and efficient procedure for the Biginelli reaction. Synthesis 2004;13:2091–3
  • Supuran CT. Carbonic anhydrases: novel therapeutic applications for inhibitors and activators. Nat Rev Drug Discov 2008;7:168–81
  • De Simone G, Pizika G, Monti SM, et al. Hydrophobic substituents of the phenylmethylsulfamide moiety can be used for the development of new selective carbonic anhydrase inhibitors. BioMed Res Int 2014;523210
  • Grutzendler J, Morris JC. Cholinesterase inhibitors for Alzheimer's disease. Drugs 2001;61:41–52
  • Bachurin SO. Medicinal chemistry approaches for the treatment and prevention of Alzheimer's disease. Med Res Rev 2003;23:48–88
  • Stepankova S, Komers K. Cholinesterases and cholinesterase inhibitors. Curr Enzyme Inhib 2008;4:160–71
  • Makhaeva GF, Boltneva NP, Lushchekina SV, et al. Synthesis, molecular docking and biological evaluation of N, N-disubstituted 2-aminothiazolines as a new class of butyrylcholinesterase and carboxylesterase inhibitors. Bioorg Med Chem 2016;24:1050–62
  • Gülçin İ, Elias R, Gepdiremen A, et al. Antioxidant activity of bisbenzylisoquinoline alkaloids from Stephania rotunda: cepharanthine and fangchinoline. J Enzyme Inhib Med Chem 2010;25:44–53
  • Gülçin İ. Antioxidant properties of resveratrol: a structure–activity insight. Innov Food Sci Emerg 2010;11:210–18
  • Balaydın HT, Gülçin İ, Menzek A, et al. Synthesis and antioxidant properties of diphenylmethane derivative bromophenols including a natural product. J Enzyme Inhib Med Chem 2010;25:685–95
  • Gülçin İ. Antioxidant activity of L-adrenaline: an activity-structure insight. Chem Biol Interact 2009;179:71–80
  • Köksal E, Gülçin İ, Öztürk Sarıkaya SB, Bursal E. On the in vitro antioxidant activity of silymarin. J Enzyme Inhib Med Chem 2009;24:395–405
  • Wood LG, Gibson PG, Garg ML. A review of the methodology for assessing in vivo antioxidant capacity. J Sci Food Agric 2006;86:2057–66

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.