Cytoprotective and Antioxidant Properties

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Bioorganic Chemistry 80 (2018) 43–56

Contents lists available at ScienceDirect

Bioorganic Chemistry
journal homepage: www.elsevier.com/locate/bioorg

Cytoprotective and antioxidant properties of organic selenides for the T


myelin-forming cells, oligodendrocytes

Saad Shaabana,b,c,f, , Dominique Vervandier-Fasseurb, Pierre Andreolettia, Amira Zarrouka,d,
Philippe Richardb, Amr Negme, Georg Manolikakesf,g, Claus Jacobh,
⁎⁎
Mustapha Cherkaoui-Malkia,
a
Univ. Bourgogne-Franche Comté, Laboratoire BioPeroxIL (Biochimie du Peroxysome, Inflammation et Métabolisme Lipidique) EA 7270, 21000 Dijon, France
b
Institut de Chimie Moléculaire de l'Université de Bourgogne, UMR6302, CNRS, Université Bourgogne Franche-Comté, F-21000 Dijon, France
c
Organic Chemistry Division, Department of Chemistry, Faculty of Science, Mansoura University, El-Gomhorya Street, 35516 Mansoura, Egypt
d
Faculté de Médecine, Laboratoire de Nutrition-Aliments Fonctionnels et Santé Vasculaire (LR12ES05), Monastir & Faculté de Médecine, Université de Monastir, Sousse,
Tunisia
e
Biochemistry Division, Department of Chemistry, Faculty of Science, Mansoura University, El-Gomhorya Street, 35516 Mansoura, Egypt
f
Institute of Organic Chemistry and Chemical Biology, Goethe-University Frankfurt, Max-von-Laue-Str. 7, 60438, Frankfurt/Main, Germany
g
Department of Chemistry, TU Kaiserslautern, Erwin-Schrödinger-Str. Geb. 54, D-67663 Kaiserslautern, Germany
h
Division of Bioorganic Chemistry, School of Pharmacy, Saarland University, Campus B2 1, D-66123 Saarbuecken, Germany

A R T I C LE I N FO A B S T R A C T

Keywords: Here a new series of twenty-one organoselenides, of potential protective activity, were synthesized and tested for
Organic selenides their intrinsic cytotoxicity, anti-apoptotic and antioxidant capacities in oligodendrocytes. Most of the organo-
Apoptosis selenides were able to decrease the ROS levels, revealing antioxidant properties. Compounds 5b and 7b showed
Neurodegeneration a high glutathione peroxidase (GPx)-like activities, which were 1.5 folds more active than ebselen. Remarkably,
Oligodendrocytes
compound 5a diminished the formation of the oligodendrocytes SubG1 peak in a concentration-dependent
Multicomponent reactions
Cytoprotective
manner, indicating its anti-apoptotic properties. Furthermore, based on the SwissADME web interface, we
performed an in-silico structure-activity relationship to explore the drug-likeness of these organoselenides,
predicting the pharmacokinetic parameters for compounds of interest that could cross the blood-brain barrier.
Collectively, we present new organoselenide compounds with cytoprotective and antioxidant properties that can
be considered as promising drug candidates for myelin diseases.

1. Introduction Oligodendrocytes support the long-term neurons integrity by myeli-


nating their axons [7]. One of the hallmarks of oligodendrocyte dys-
In the brain, the myelin sheath plays a crucial role for neuron signal function and apoptosis is their high susceptibility to oxidative stress
transmission and their survival [1]. Such a sheath is built by a multi- (OS), which results from the excess production of reactive oxygen
layered spiral extension of oligodendrocyte cell membranes to wrap the species (ROS; e.g., hydrogen peroxide, superoxide, and hydroxyl radi-
neuron’s axons [2]. Chronic erosion of this myelin sheath leads to de- cals), by cellular aerobic metabolism, particularly from mitochondrial
myelination, and disrupting the neuron integrity and the normal axonal respiration [8]. Oxidative damage, caused in a variety of neurodegen-
function [3]. Demyelination is largely associated with numerous neu- erative diseases, is mediated by excessive production of ROS, which can
rodegenerative diseases (e.g., Parkinson’ and Alzheimer’s diseases) as be linked to cell lysis or oxidative burst associated to the immune re-
well as multiple sclerosis and peroxisomal leukodystrophies rare dis- sponse [9]. This overproduction occurs when the generation of reactive
eases [4,5]. Thus for the myelination process, oligodendrocyte integrity oxygen species (ROS) exceeds the antioxidant cellular capacity, in-
is central and their dysfunction was also detected in several psychiatric cluding enzymatic (catalase, glutathione peroxidase and superoxide
disorders, including autism, schizophrenia, and depression [6]. dismutase) and non-enzymatic defense systems (glutathione and

Abbreviations: CNS, Central nervous system; MTT, 3-(4,5- dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide; DCF, 7-ketocholesterol (7kc), dichlorofluorescein; DHE, dihy-
droethidium; GPx, glutathione peroxidase; ROS, reactive oxygen species; DPPH, 2,2-diphenyl-1-picrylhydrazyl; ABTS, 2,2′-azino-bis-3-ethylbenzthiazoline-6-sulphonic acid

Corresponding author at: Organic Chemistry Division, Department of Chemistry, Faculty of Science, Mansoura University, El-Gomhorya Street, 35516 Mansoura, Egypt.
⁎⁎
Corresponding author at: Univ. Bourgogne-Franche Comté, Laboratoire BioPeroxIL EA7270, Faculté des Sciences Gabriel, 6 Bd Gabriel, 21000 Dijon, France.
E-mail addresses: dr_saad_chem@mans.edu.eg (S. Shaaban), malki@u-bourgogne.fr (M. Cherkaoui-Malki).

https://doi.org/10.1016/j.bioorg.2018.05.019
Received 31 March 2018; Received in revised form 16 May 2018; Accepted 20 May 2018
Available online 21 May 2018
0045-2068/ © 2018 Elsevier Inc. All rights reserved.
S. Shaaban et al. Bioorganic Chemistry 80 (2018) 43–56

ascorbic acid) [10]. The selenoprotein family, including glutathione 2. Results and discussions
peroxidases, represents one of the first-line defenses against ROS. Se-
lenoproteins play also a crucial role for neuronal function, as demon- 2.1. Design and syntheses
strated by deactivation of the tRNA [Ser] (Sec) gene in mice, resulting
in extensive neurodegeneration [11]. The second line of defense is re- The last decade has witnessed significant advance in the synthesis of
presented by cellular antioxidants, such as glutathione (GSH) and se- diverse classes of organic selenides e.g., diselenides, selenocyanates,
lenium (Se), the latter for its implication in the active center of several and seleno-heterocyclic compounds. Although, these compounds have a
glutathione peroxidases and in the catalytic activity of thioredoxin re- pivotal role in human health and disease prevention; they have limited
ductase [12]. Depleted states of the reduced form of GSH and selenium application in biological systems due to their often toxicity especially if
were associated with the progression of neurodegenerative diseases higher concentrations were administrated. Combining selenium with
[13–15]. This underlines the overarching importance of an essential better physico-chemical properties bioactive pharmacophores will in-
dietary supply of Se for mammals. Interestingly, there seems to be a teractively not only stimulate their corresponding cytotoxicity but will
connection between Se and other antioxidants on the one side, and the also foster their bioavailability. Within this context, our design strategy
role of ROS in the pathogenesis of neurodegenerative diseases on the relies on the synthesis of agents in which selenium is combined with
other [16–18]. Yeo et al., reported that Se antioxidant activity pre- different functionalities such as quinone, cyclic imides, N-substituted
vented cell apoptosis in an experimental rat spinal cord injury [19]. maleanilic and succinanilic acids, formamide, acetanilide, acetaldehyde
Numerous data revealed that Se levels decline in humans with age and diethyl acetal, and/or benzyl. Most of these functionalities are com-
that Se might play different roles in the evolution of Alzheimer’s disease monly found in bioactive compounds, thus enabling the chemical di-
[20,21]. Se deficiency leads to inhibition of the normal upregulation of versity and increasing polarity.
myelin genes in differentiating oligodendrocytes and the progression Based on our recent reports on organoselenium agents [28–35], we
from immature to mature oligodendrocytes as well [22]. Curiously, envisioned the synthesis of novel selenium-based primary aromatic
experiments using animal models reveal that organoselenium has a amines 2a-d. The amine functionality in turn can give an access to a
better bioavailability and biological activity than inorganic Se [23]. large group of agents either via isocyanide-based multicomponent re-
Therefore, during the last decade or two, the biological chemistry of actions (IMCR) (e.g., Passerini, azido-Ugi Passerini, Ugi, and Groebke-
selenium derivatives has often focused on potential antioxidant and Blackburn-Bienaymé reactions) or via the classical amines reactions
neuroprotective activities [24], and here primarily on organic rather (e.g., formylation, acetylation, etc.).
than inorganic selenium agents [25,26]. During our research on sele- The key synthons selenoamines 2a-d were synthesized in good
nium derivatives with possible antioxidant and neuroprotective activ- yields (up to 91%) via reduction of the diselenide (1) with sodium
ities, we recently developed organoselenium compounds with enhanced tetrahydridoborate (NaBH4) followed by subsequent nucleophilic sub-
antioxidant and GPx-like activities [27–32]. The mechanism by which stitution (SN) reaction with appropriate organic halides (Scheme 1).
these compounds exert their antioxidant effect is related to the mod- Chalcogen-bearing isonitriles are not commercially available and
ulation of the ROS and GSH levels in cancer cells. Interestingly, some of usually challenging to synthesize. In 2009, we reported the synthesis of
these compounds showed better antioxidant and GPx-like activities three chalcogen-based isonitriles, including the tellurium isonitrile for
than classical antioxidants (e.g., vitamin C, ebselen) [27,30,32–36]. the first time [32,34]. Therefore, our preliminary target was to convert
Based on these results, we decided to further direct our research to the amines 2a, 2b, 2c, and 2d into the corresponding isonitriles, sui-
obtain evidences regarding the structural requirements underlying the table for further applications in IMCR [29,40]. The synthesis of iso-
potential cytoprotective/neuroprotective activities of organic selenides cyanides is usually performed in two steps i.e. amine formylation em-
taking the myelin-forming cells, oligodendrocytes as a model of study. ploying acetic formic anhydride followed by subsequent dehydration
Different reports on the synthesis of organoselenium compounds using phosphoryl chloride (POCl3). In accordance with our aim, we
have already been published; however, they generally require the decided to apply the same methodology for the transformation of or-
handling of hazard reagents (e.g., KSeCN), harsh reaction conditions ganoselenium-based amines 2a-d into the corresponding isocyanides.
(e.g., strongly acidic or basic), and mostly multistep procedures Unexpectedly, acetanilides 4a-c were invariably obtained instead of the
[37,38]. Therefore, we decided to avoid rather sophisticated synthetic corresponding formamides. This promoted us to use only formic acid in
procedures and employ simple, straight forward and facile routes to- place of the acetic formic anhydride. Under neat conditions, the desired
wards the synthesis of our preliminary organoselenium-based library. corresponding formamides were obtained in good yields (up to 88%).
Here, we present the results obtained during extensive in vitro The dehydration of formamides was thereafter investigated using
evaluations of the selenium derivatives in oligodendrocytes. Besides a POCl3. A non-polar product (eluted with petroleum ether) with the
rational design of synthetic organoselenium compounds, we have characteristic isonitrile odor was detected by TLC; however, the triplet
13
aimed at gaining a deeper insight into the potential use of these com- CNMR peak (≈156 ppm) corresponding to the isocyanide carbon did
pounds as neuroprotectors in neurodegenerative diseases through the not appear, suggesting that the isocyanide may be unstable. Therefore,
protective effect on oligodendrocytes, the myelinating cells. For the we designed a one-pot synthetic approach based on the use of the in situ
chemical synthesis, we adopted both multistep and one-pot synthesis as formed isocyanide in subsequent isocyanide-based MCRs (IMCRs) such
part of a diversity-oriented approach to achieve the synthesis of the as Groebke-Blackburn-Bienaymé and Ugi reactions [40]. Unfortunately,
twenty-one organoselenides. The in vitro evaluations were conducted very poor conversion (< 5%) was constantly observed with subsequent
using immortalized murine oligodendrocyte cell lines [39]. Flow cyto- disappearance of the isocyanide from the reaction mixture (monitored
metry was used to estimate the ROS levels employing the H2-DCFDA by TLC). Hence, we concluded, that this result is related to the high
and the DHE assays. Furthermore, the antioxidant potential of these reactivity and low stability of the in situ formed isocyanides. Different
compounds was further investigated employing different chemical and dehydrating agents such as cyanuric chloride [41], triphenylphosphine
biochemical assays and the apoptotic/anti-apoptotic properties of the [42], 4-toluenesulfonyl chloride [43], DABCO [44] and methyl N-
compounds were detected by flow cytometry follow-up of the sub-G1 (triethylammoniumsulfonyl)carbamate [45] were investigated; how-
peak. Finally, to assess the structure-activity relationship of the syn- ever, these reagents turned out to be either low yielding or ineffective
thesized organoselenium compounds, we determined in silico their with our seleno-formamides.
physicochemical and pharmacokinetic parameters, and their drug- Further efforts were then directed to evaluate the reactivity of the
likeness as well through the free web tools in SwissADME. novel selenoamines 2a-d via their reactions with various anhydrides
aiming to synthesize selenocyclic imides which we recently proved to
possess enhanced antioxidant activities (e.g., GPx-mimicking and free

44
S. Shaaban et al. Bioorganic Chemistry 80 (2018) 43–56

Scheme 1. Synthesis of the selenoamines 2a-d, selenoformamides 3a-c and selenoacetamides 4a-c. Reagents and conditions: (i) Amine (1 mmol), NaBH4 (5 mmol);
(ii) Amine (1 mmol), 85% aq. HCOOH (1.2 eq), 2 h, 60 °C; (iii) Amine (1 mmol), HCOOH (1 eq), acetic anhydride (1.5 eq), 2 h, 55 °C.

radical-scavenging activity) [28–31]. Amines 2b-c reacted with maleic (2) single bond with a bond distance of 1.389 Å, C(17)eO(2) single
and succinic anhydrides in dry toluene at ambient temperature afforded bond with a bond distance of 1.405 Å, and C(17)eO(2) double bond
the corresponding N-substituted maleanilic 5a-b and N-substituted with a bond distance of 1.195 Å. Such efficient and clean pathway to
succinanilic 6a-b derivatives in quantitative yields (Scheme 2). In isomaleimides synthesis is highly useful and worth further investiga-
contrast, 2a failed to react under these conditions and a tar material tions using a wider arsenal of N-substituted selenomonoamidic acids.
was formed. This may be attributed to the side reaction(s) such as hy-
drolysis and sequential condensation and/or polymerization which
might take place at the acetal residue. Furthermore, the resulting N- 2.2. Cytoprotective and antioxidant profiles
substituted monoamidic acids 5a-b and 6a were subjected to acid-cat-
alyzed esterification and the corresponding methyl esters 7a-b and 8a 2.2.1. Organoselenides effect on the viability of the oligodendrocytes 158 N
were obtained in good yields (up to 87%). The esterification of 6b was and 158JP
also performed under the same conditions as of 6a. Unfortunately, the Oligodendrocytes play a crucial in the long-term neurons integrity
expected quinone-based methylester was not obtained; instead we ob- by myelinating their axons and thus securing the fast signal conduction
served a decomposition of 6b. We attributed this behavior to the for- [2,3]. Oligodendrocytes are the most vulnerable cells of the CNS to ROS
mation of the corresponding hydroquinone (under acidic conditions) and there are only very few reports on the cytoprotective/cytotoxic role
and subsequent cleavage of the amidic bond. of organoselenides on these myelinating cells. It was therefore of in-
As expected, selenocyclic imides 10a-b were obtained via ring-clo- terest to investigate the protective activity of the synthesized organo-
sure of the corresponding N-substituted succinanilic acids 6a-b with selenides on two immortalized murine oligodendrocytes: 158N and
acetic anhydride at 55 °C in the presence of sodium acetate (Scheme 2). 158JP cell lines. The jimpy (158JP) cell line is mutated for the pro-
Unexpectedly, kinetically controlled isomaleimides 9a-b were ob- teolipid protein PLP/DM20, a predominant myelin protein that may
tained in good yields (up to 77%) instead of the thermodynamically, have a role in the maintenance of myelin sheaths [39]. To determine
more stable, maleimides. This was confirmed via spectral data and X- the half maximal inhibitory concentration IC50 of the organoselenides, a
ray diffraction. A perspective view and atom numbering are given in range of concentrations was evaluated on both oligodendrocytes cell
Fig. 1 and crystal structure determination and refinement parameters lines. The low cytotoxic concentration will be further selected to eval-
are shown in the Experimental section. uate the compounds corresponding antioxidant and protective proper-
The X-ray single crystal structure of 9a shows a triclinic system with ties. Accordingly, MTT assay was used to evaluate the basal oligoden-
a space group of P-1. The phenyl ring is directly bonded to the imine drocytes cytotoxicity of the twenty-one organic selenides on 158N and
moiety (eN]). There are three different types of CeO bonds: C(14)eO 158JP oligodendrocytes using 7-ketocholesterol (7kc) as known cyto-
toxic positive control [46]. MTT is metabolized by the mitochondrial

45
S. Shaaban et al. Bioorganic Chemistry 80 (2018) 43–56

Scheme 2. Reactions of selenoamines 2a-d with anhydrides and subsequent dehydration/esterification. Reagents and conditions: (i) amine (1 mmol), toluene (5 ml),
anhydride (1 mmol), r.t, 3 h; (ii) amide-acid (1 mmol), MeOH (10 ml), conc.H2SO4 (200 µl), 8 h, r.t; (iii) amide-acid (1 eq), 100 mg NaOAc, Ac2O (3 ml), 2 h, 50–60 °C.

enzyme succinate dehydrogenase giving the blue formazan product. down to 9 and 3 µM in 158N cells and 158JP, respectively. Surprisingly,
Hence, this assay provides information on viability (and/or cell pro- the cytotoxicity of acetanilides 4a and 4c was more pronounced in the
liferation) and mitochondrial metabolism. The concentration producing case of 158JP cells compared to 158N cells (IC50 = 0.02 and 14 µM,
50% growth inhibition (IC50, Table 1) and the maximum concentration respectively). These compounds may act as pro-oxidants and their
without toxicity were estimated for each of the compounds from the toxicity was significant in 158JP owing to the fact that these cells have
respective dose-response curves. higher ROS levels (10-fold) compared to 158N, due to the known higher
As shown in Table 1, most of the compounds did not exhibit any mitochondrial activity and redox status in 158JP [47]. A possible ex-
effect on the viability (IC50 ≥ 100 µM) of 158N and 158JP at the con- planation of acetanilides derivatives cytotoxicity, is may be related to
centration range used, except for the quinoid-based compounds 6b and the accumulation of paracetamol, a well-known cytotoxin, produced
10b and N-substituted maleanilic ester 7a, they showed IC50 values during the intracellular xenobiotic metabolism of acetanilides [48].

Fig. 1. Ortep view of of (Z)-5-((4-((4-bromophenyl)


selanyl)phenyl)imino)furan-2(5H)-one (9a). Selected
bond lengths (Å): C11-N1 1.414, C14-N1 1.266, C14-
O2 1.389, C17-O1 1.195, C17-O2 1.405. Selected
bond angles (o): C14-N1-C11 126.4, N1-C14-O2
126.1, C14-O2-C17 107.77, O1-C17-O2 120.0.

46
S. Shaaban et al. Bioorganic Chemistry 80 (2018) 43–56

Table 1
Newly synthesized organoselenides, yields and their effect on the viability of 158N and 158JP cells.a
R1SePhR2 R1 R2 Yields % IC50 (µM)

158N 158JP

2a eCH2CH(OC2H5)2 eNH2 89 54 -b
2b eCH2C6H5 eNH2 85 -b -b
2c eCH2-4-Br-C6H4 eNH2 91 -b -b
2d -3-Me-1,4-naphthoquinone eNH2 89 27 -b
3a eCH2CH(OC2H5)2 eNHCHO 82 25 -b
3b eCH2-4-Br-C6H4 eNHCHO 88 43 91
3c -3-Me-1,4-naphthoquinone eNHCHO 81 39 -b
4a eCH2CH(OC2H5)2 eNHCOCH3 89 14 0.02
4b -CH2C6H5 eNHCOCH3 92 -b -b
4c 3-Me-1,4-naphthoquinone eNHCOCH3 75 11 0.02
5a eCH2-4-Br-C6H4 eNHCOCH]CHCOOH 94 -b -b
5b 3-Me-1,4-naphthoquinone eNHCOCH]CHCOOH 91 -b 35
6a eCH2-4-Br-C6H4 eNHCOCH2CH2COOH 90 54 -b
6b 3-Me-1,4-naphthoquinone eNHCOCH2CH2COOH 93 26 3
7a eCH2-4-Br-C6H4 eNHCOCH]CHCOOMe 86 9 20
7b 3-Me-1,4-naphthoquinone eNHCOCH]CHCOOMe 84 -b -b
8a eCH2-4-Br-C6H4 -NHCOCH2CH2COOMe 87 -b 83
9a eCH2-4-Br-C6H4 77 29 50

9b 3-Me-1,4-naphthoquinone 75 -b -b

10a eCH2-4-Br-C6H4 76 -b -b

10b 3-Me-1,4-naphthoquinone 76 16 5

7kc – – – 37 74

a
The cytotoxicity was measured after one day of treatment with different concentrations (0, 1, 10, 20, 50 and 100 µM) of the organic selenides using the MTT
assay. The IC50 was estimated as the mean of two parallel experiments; 7kc was used as a positive control.
b
Means no growth inhibition was observed at the tested concentration range.

2.2.2. Assessment of antioxidant profiles of the selenide compounds intracellular ROS levels at all the tested concentrations proposing a pro-
Organoselenium compounds are generally good nucleophiles and oxidant activity of these compounds (Table 2).
this fact led to the rational design of several synthetic organoselenides Interestingly, the pro-oxidant activity was more pronounced in the
as antioxidants, chemopreventive, and neuroprotective agents case of the quinoid compounds 2d which showed the highest H2-DCF
[36–38,49,50]. To the best of our knowledge, the underlying me- fluorescence intensity. This is hardly surprising, as quinones in general
chanism(s) of action is (are) not fully understood. Organoselenium and selenium-containing naphthoquinones, in particular, are well
compounds can either act as pro-oxidants or antioxidants depending on known to generate ROS in a catalytic fashion - and via the reduction of
the redox status of their environment. In normal cells, organic selenides dioxygen in the presence of a suitable reducing agent.
behave as antioxidants and protect cells from ROS. In contrast, these
compounds switch into pro-oxidants in cells with elevated ROS levels 2.2.2.2. Effect of organoselenium compounds on superoxide radical anion
[51–54]. (O2.−) production by oligodendrocytes. In living cells and under aerobic
Taking into account that the CNS is prone to oxidative damage and conditions, quinones undergo redox cycling with triplet oxygen to
in the light of our search for new neuroprotective agents, our aim is to generate superoxide radical anion (O2%−). The later causes oxidative
further investigate the antioxidant potential of the novel organic sele- DNA damage, apoptosis and subsequent cell death [55]. This
nides employing different biochemical and chemical assays. biochemical feature is responsible for most of the quinone-based
drug’s activity. From this perspective, all the selenoquinones were
2.2.2.1. Effect of organoselenides on oligodendrocytes ROS selected for further estimation of their respective ability to produce
production. ROS levels were estimated by flow cytometry in 158N O2%− by the DHE assay. The latter is believed to be a selective “gold
cells treated with different concentrations (0, 10, 20 and 50 µM) of the standard” O2%−detector. DHE is a lipophilic cell-permeable probe,
organoselenium compounds employing the commonly used H2-DCFDA which is rapidly oxidized by O2%−, to give the fluorescent compound 2-
and DHE assays for the detection of ROS and of superoxide anions hydroxyethidium [55]. As shown in Table 3, and in agreement with the
(O2%−), respectively [55,56]. The commercial α-Tocopherol (vitamin literature and our hypothesis of quinones as ROS-generating
E) was used as a standard antioxidant (positive control). substances, all quinoid-based compounds were able to increase the
In the H2-DCFDA assay, most of the compounds were able to de- O2%− levels, compared to vitamin E. Interestingly, compound 2d was
crease the ROS levels in 158N cells. Interestingly, compounds 2b, 2c, among the most active compounds and was able to induce a dose-
3b, 4a, 4b, 4c, 5a, 5b, 6b, and 9a showed a significant decrease in the dependent increase of O2%− in 158N cells.
H2-DCF fluorescence intensity, thus lower ROS production at 10 and
20 µM compared to vitamin E (Table 2). On the other hand, compounds 2.2.3. Anti-apoptotic effect of selenide compounds on the oligodendrocytes
7a, 8a and 10a and most of the quinone-based organoselenium 7b, 9b, Apoptosis was estimated by staining fixed 158N cells with PI fol-
and 10b were able to mediate significant overproduction of the lowed employing flow cytometry quantification of the sub-G1 peak in

47
S. Shaaban et al. Bioorganic Chemistry 80 (2018) 43–56

Table 2 any effect on the development of the SubG1 peak (data not shown).
Evaluation of the effects of compounds on ROS production by H2-DCFDA as- Compound 5a is reducing ROS levels, which in essence is an anti-
say.a oxidative and hence often also an anti-apoptotic event. Admittedly,
10 µM Foldb 20 µM Foldb 50 µM Foldb some of the other compounds (e.g., 5b, 6b and 9a) are a lot better in
removing ROS, yet, they may not be as good in crossing membranes.
2a 71 ± 5* 0.75 81 ± 10* 0.86 82 ± 10* 0.87 This may be due to that 5a is probably amphiphilic and hence a good
2b 38 ± 4* 0.40 38 ± 5* 0.40 33 ± 2* 0.35
molecule to enter cells, whilst others are not. Compound 5a is also an
2c 36 ± 8* 0.38 41 ± 2* 0.43 48 ± 2* 0.51
2d 700 ± 143* 7.44 854 ± 135 9.08 935 ± 307* 9.94 antioxidant (albeit a weak one) and may act via its unsaturated bond.
3a 43 ± 2* 0.45 46 ± 1* 0.49 71 ± 209 0.76 There is another possible mode of action: is an attack of 5a on cysteine
3b 34 ± 6* 0.36 33 ± 5* 0.35 38 ± 8* 0.40 proteins via its unsaturated double bond, and this may result in an
3c 124 ± 7 1.32 115 ± 9 1.22 95 ± 14 1.01
upregulation of antioxidant pathways and hence anti-apoptotic action
4a 45 ± 2* 0.48 49 ± 11* 0.52 54 ± 20* 0.57
4b 20 ± 14* 0.21 15 ± 6* 0.16 13 ± 3* 0.14
as well (for instance via Nrf-2). All this is, of course, speculation, and
4c 7 ± 2* 0.07 5 ± 1* 0.05 6 ± 1* 0.06 these results worth further in-depth studies to substantiate the anti-
5a 42 ± 10* 0.45 36 ± 20* 0.38 31 ± 20* 0.32 apoptotic effect of 5a. Induction of anti-apoptotic effect in oligoden-
5b 11 ± 2* 0.12 8 ± 3* 0.09 10 ± 4* 0.11 drocytes by 5a is a key target for chemotherapeutic and chemopre-
6a 72 ± 32 0.77 64 ± 14* 0.68 51 ± 19* 0.54
ventive applications of such a compound, opening the way for further
6b 15 ± 0* 0.16 24 ± 0* 0.26 26 ± 1* 0.28
7a 127 ± 52 1.35 133 ± 50 1.42 103 ± 50 1.10 pharmacological studies.
7b 241 ± 74* 2.56 218 ± 48* 2.32 250 ± 71* 2.66
8a 127 ± 40 1.35 135 ± 47 1.44 140* ± 40 1.49 2.2.4. Glutathione peroxidase-like activity assay
9a 7 ± 4* 0.07 8 ± 4* 0.09 11 ± 4* 0.12
Many organoselenium compounds have long been known to mimic
9b 226 ± 79* 2.40 216 ± 51* 2.30 178 ± 23* 1.90
10a 179 ± 150 1.90 164 ± 100 1.74 147 ± 75* 1.60
the antioxidant activity of the GPx selenoenzyme. Among these com-
10b 217 ± 11* 2.30 192 ± 82 2.04 197 ± 54* 2.10 pounds, the non-toxic seleno-organic drug, ebselen has been extensively
Vitamine E 94 ± 5 studied because of its superior GPx mimetic activity. Recently, we have
a
reported different selenium-based organic compounds that possess
158N Cells were cultured in the presence of different compounds (C1 = 10; higher catalytic GPx-like activity than ebselen. In view of the former,
C2 = 20; C3 = 50 µM); Flow cytometry technique was used to evaluate ROS
the novel organic selenides were evaluated for their GPx mimicking
levels via staining cells with H2-DCF; data are shown as mean ± SD and re-
activity [30,60].
present the absolute values after corrections (subtraction of the untreated cells
The GPx catalyzes the reduction of ROOR, in presence of glu-
fluorescence).
b tathione, thus protecting cells from oxidative damage by H2O2 [61–63].
Fold of the compounds was calculated as follow: result assay at the given
The reaction takes place at the enzyme catalytic site, which contains
concentration/result of Vitamin E. significance of the difference between the
selenium redox center as a selenocysteine amino acid. It is worthwhile
DMSO and compounds. Treated cells are indicated by (Mann–Whitney test; *:
to mention that selenium is an essential cofactor in all the selenocys-
P < 0.05); vitamin E (50 µM) was used as a positive control.
teine-based GPx family members for the manifestation of their corre-
sponding antioxidant activities [64].
Table 3
Evaluation of the effects of compounds on superoxide anions production by
The catalytic GPx mimic activities of the newly synthesized organic-
DHE assay. selenides were evaluated employing the NADPH-reductase coupled
assays [56–68]. The latter is repeatedly used for the evaluation of the
DHE assay (% of positive cells)
GPx-like activity of organochalcogens. The assay principle depends on
Vitamine E 74 ± 1* the reduction of H2O2 by GPx followed by oxidation of glutathione
10 μM 20 μM 50 μM disulfide (GSSG) and NADPH to glutathione (GSH, reduced form) and
2d 264 ± 25* 406 ± 29* 417 ± 31* NADP+, respectively. The latter can be spectrophotometrically detected
3c 115 ± 8 109 ± 1 117 ± 6
by a decrease in the absorbance at 340 nm. The known, standard GPx
4c 109 ± 12 121 ± 1 120 ± 4
5b 102 ± 4 105 ± 4 112 ± 9 mimic compound, ebselen is used as the positive control. The back-
6b 211 ± 38* 220 ± 41* 252 ± 41* ground reaction of GSH with H2O2 was measured and the activity of the
9b 166 ± 32* 172 ± 33* 179 ± 31* GPx was calculated after correction. The reaction was followed till the
10b 133 ± 37 151 ± 28* 218 ± 40* end and the rate of the reaction was constantly linear (Fig. 3).
7b 86 ± 15 87 ± 15 91 ± 30
Interestingly, as shown in Fig. 3, the quinoid-based N-substituted
158N Cells were grown in the presence of different compounds (C1 = 10; maleanilic 5b acid and its corresponding methyl esters 7b were more
C2 = 20; C3 = 50 µM); Flow cytometry techniques was used to estimate the active (≈1.5-fold) than ebselen. The rest of the compounds showed
O2%− production via staining with DHE; Data are shown as mean ± SD and moderate GPx activity, particularly: 2d, 3b, 8a and 9b compounds.
expressed as % control; Significance of the difference between the DMSO and
compounds. Treated cells are indicated by (Mann–Whitney test; *: P < 0.05); 2.2.5. Assessment of antioxidant/pro-oxidant activity: DPPH, ABTS radical
Vitamin E (50 µM) was used as a positive control. scavenging, and bleomycin DNA damage assays
Among different assays used for the accounting of the antioxidant
the cell cycle. The sub-G1 is indicator of apoptotic cells and can be activities of organic compounds, the 2,2-diphenyl-2-picrylhydrazyl
identified by a nuclear fragmentation and/or condensation associated (DPPH) and 2,2′-azinobis-3-ethylbenzothioazoline-6-sulphonate (ABTS)
with an internucleosomal fragmentation of the DNA [57,58]. 7Kc is chemical assays are usually used to evaluate the radical scavenging
known to induce oxidative stress and trigger apoptosis in 158N murine activities of natural products, foods (e.g., vegetables, fruits and bev-
oligodendrocytes and therefore was used as apoptosis promoter and the erages) and several organoselenium compounds [27,30,69,70]. Fur-
positive control [59]. thermore, the bleomycin-induced DNA damage assay isconsidered as an
To detect apoptosis, cell cycle analysis was performed after treat- accurate method to evaluate the pro-oxidant activity of many food
ment of 158N cells with different concentrations (50, 10, 5 and 1 μM) of antioxidants and antineoplastic agents [69,70].
organoselenium compounds treated with or without 7kc. Interestingly, As presented in Fig. S1 and Table S1 (Supporting Information), most
compound 5a was able to diminish the formation of the SubG1 peak in of the compounds showed good-moderate scavenging activity com-
a concentration-dependent manner, which is an indicator of its anti- pared to vitamine C. On the other hand, selenoquinones 2d, 3c, 4c, 5b,
apoptotic properties (Fig. 2). The remaining compounds did not show 6b, 7b, 9b and 10b showed between 40 and 60% pro-oxidant activities,

48
S. Shaaban et al. Bioorganic Chemistry 80 (2018) 43–56

Fig. 2. Anti-apoptotic effect of the organoselenium


derivative 5a on oligodendrocytes. The anti-apop-
totic effect was measured by cytometry using pro-
pidium iodide. The organoselenium compound 5a
was used at different concentrations (50, 10, 5, and
1 μM) to evaluate the percentage of cells in Sub-G1
phase during the cell cycle of 158N cells treated
with/without 7kc (50 μM) for 24 h. Data shown are
mean ± SD.

when compared to vitamin C (bleomycin-reducing activity considered which ranges from 1 (very easy synthesis) to 10 (very difficult synth-
as 100%). esis). This computation of SA score was implemented by Daina et al.
These results were in agreement with that obtained from DCF and [72] in SwissADME interface to assist the medicinal chemist for the
DHE assays that selenoquinones have more pro-oxidant properties identification of potentially problematic fragments and eventual lead
compared to the rest of organoselenium compounds. Although the optimization.
DPPH and the ABTS assays are widely used, they are only in vitro In parallel, to compare the structure-activity relationship of the
models and do not necessarily translated into in vivo action, in parti- synthesized organoselenide derivatives, we summarized the obtained
cular, the ADME factors are not covered here either. experimental results as a heatmap of these compounds taking into ac-
count their chemical phylogeny (Fig. 4): where the scalable red color
2.3. In silico structure-activity relationship indicates the negative effect, while the scalable green color reveals the
positive effect of the tested compounds. The heatmap of our experi-
Based on the molecular structure, we explored in silico the synthe- mental results (Fig. 4) shows that selenoamines 2a (diethoxyethyl se-
sized organoselenides drugability by performing a computer-aided lanyl aniline) and 2d (selanyl dihydronaphthalene, dione), derived
structure-activity prediction through the free web tools in SwissADME, from diselinide as a precursor, exhibited a low or a mild cytotoxicity
which is a user-friendly interface website (http://www.swissadme.ch) respectively, while selenylaniline derivatives 2b and 2c revealed a
to evaluate several physicochemical and pharmacokinetic parameters protective effect on oligodendrocytes. It’s noteworthy, that selenoa-
[71]. Among these parameters showed in Fig. 4: the topological polar mines, as selenocystine, induce ROS production and exert their pro-
surface area (TPSA) based on the summation of tabulated surface oxidant action through depletion of the cellular antioxidant glutathione
contributions of polar fragments in the compound; the lipophilicity [73–75]. Interestingly, compound 2d, which show a higher cytotoxicity
represented by the n-octanol/water partition coefficient (log Po/w), of the 2-serie compounds, presents also strong pro-oxidant and DNA
which is a key physicochemical parameter in drug discovery; the damage activities and a lower antioxidant property and all 2d deriva-
pharmacokinetic (i.e. gastrointestinal (GI) absorption, blood-brain tives, containing the 2-methyl-3-selanylnaphthalene-1,4-dione moiety,
barrier (BBB) permeability), and the synthetic accessibility (SA) score, conserved a significant pro-oxidant and DNA damage activities.

Fig. 3. GPx-like activity assay (µM min−1). The reduction of H2O2 by GPx-like activity was tracked by oxidation of glutathione disulfide (GSSG) and NADPH to
glutathione (GSH, reduced form) and NADP+, respectively. The latter was detected by a decrease in the absorbance at 340 nm.

49
S. Shaaban et al. Bioorganic Chemistry 80 (2018) 43–56

Fig. 4. Chemical phylogeny heatmap and drug-likeness descriptors of the synthesized organoselinides. Chemical phylogeny was obtained taking into account the
synthesis design described for each compound in the result section and the experimental results were reported as a heatmap, in which the scalable red color indicates
the negative effect, while the scalable green color reveals the positive effect of the tested compounds respectively. The color scale was determined from the
experimental values presented in figures and tables. TPSA: topological polar surface area; log Po/w: n-octanol/water partition coefficient; GI: gastrointestinal; BBB:
blood-brain barrier.

However, among 2d derivatives, three compounds (5b, 7b and 9b) respectively). Within this context, the 7a esterified derivative with a
exhibited a protective effect on oligodendrocytes and showed a GPx- lower TPSA (55.40 Å2) and a poor solubility, showed a strong cyto-
like activity, but only compound 5b revealed antioxidant and anti- toxicity and a significant GPx activity (Fig. 4). While compound 5a
apoptotic properties (Fig. 4). Several studies reviewed by Bhabak and which derived from 2c, with a TPSA of 66.40 Å2, showed protective and
Mugesh [76] described the mechanism of GPx activity of Ebselen and highest anti-apoptotic effects on oligodendrocytes as well as a good
related organoselenium compounds, for which the strength of Se⋯O antioxidant property, but has a low GPx activity. Thus, compound 5a
non-bonded interactions in the selenenyl sulfides fine-tuned the balance presents interesting protective properties for oligodendrocytes and
between the antioxidant scavenging and the GPx activity. Additionally, could become a lead compound in the development of new drug. The
7b showed a negative predicted pharmacokinetic to cross the blood- pioneer work on drug-likeness was setup by Lipinski et al. [80] as a
brain barrier. Structurally, compound 7b is an esterified derivative of rule-of-five to predict poor bioavailability for a drug candidate when it
the N-substituted monoamidic acid 5b. Nonetheless, a strong correla- contains more than 5H-bond donors, 10H-bond acceptors, a molecular
tion exists between the absorbed fraction and the hydrogen bond de- weight superior to 500 and a calculated log Po/w higher than 5. Re-
scriptors [77]. Here the physicochemical analysis in SwissADME re- markably, all the substituted organoselenides that we synthesized show
vealed that compound 7b has additional 4 rotatable bonds and one H- favorable Lipinski rules (with zero violation) indicative of their like-
bond donor, which may account for its unfavorable molecular dynamic. lihood drug-likeness to become an oral drug with respect to bioavail-
Furthermore, TPSA was defined by Ertl et al. [78] as the sum of the ability. Pharmacokinetically, except compounds 6b and 7b, all the
surface areas of polar fragments in a molecule. As documented by other synthesized organoselenides would potentially cross the blood-
Hitchcook et al. [77], compounds with PSA < 90 Å2 are considered brain barrier. The predicted synthetic accessibility scores range from
easily absorbable with higher capacity in penetrating cellular mem- 2.6 to 3.8, revealing that these compounds are easily synthesizable.
branes, while the preferred range is compounds with PSA < 70 Å2. Such prediction is in agreement with the obtained good experimental
Moreover, substances with high TPSA are more prone to better interact synthesis yields ranging from 77% to 91%. Collectively, the in silico and
with hydrophobic environment of their transporter proteins, as shown in cellulo collected data are in favor of additional investigations as the
for the Multidrug resistance-associated protein 1 [79]. organotypic cerebellar slice model to study the effects of lead com-
In line to this, compounds 5b, 6b, and 7b are either equally or pound on axon demyelination.
above the PSA limit (72.80 Å2, 100.54 Å2, and 89.54 Å2, respectively).
Interestingly, compounds 2c, 5a and 7a showed lower polar surface
than the recommended PSA limit (26.02 Å2, 66.40 Å2, and 55.40 Å2

50
S. Shaaban et al. Bioorganic Chemistry 80 (2018) 43–56

3. Conclusion obtained on 7kc treated 158N cells [85]. Organoselenium compounds


were prepared in DMSO and then diluted with DMEM up to 1 mM.
In the present work, we synthesized, in good yields, twenty-one
novel organoselenium compounds and evaluated their potential anti- 4.2.2. MTT assay
oxidant and protective activities in silico and in cellulo on oligoden- The MTT assay was carried out on 158N and 158JP cells after one
drocytes. It’s noteworthy that most of the organic selenides demon- day of incubation with 7kc (50 µM) different concentrations (0, 1, 10,
strated a cytoprotective effect in two different oligodendrocyte cell 20, 50 and 100 µM) of the organic selenides. In live cells, the MTT is
lines, indicating that these compounds may represent chemoprotective reduced to formazan by the succinate dehydrogenase mitochondrial
activity. Therefore, the potential antioxidant properties of the com- enzyme. The microplate reader Tecan Sunrise (Tecan, Lyon, France)
pounds were evaluated employing different well-established biological was used for the reading of the plates at 570 nm. The IC50 values and
and chemical assays, including H2-DCFDA, DHE, DPPH, ABTS, bleo- the maximum concentrations without toxicity were estimated for each
mycin DNA damage and GPx-like assays. Our data demonstrated that of the compounds from the respective dose-response curves.
most of the selenium-based quinones manifested a pro-oxidant activity,
whereas, the rest of the compounds demonstrated antioxidant activity. 4.2.3. H2-DCFDA and DHE assays
Within this context, organoselenium compounds 2c, 2b, 3b, 4a, 4b, 4c, ROS overproduction and O2%− and were detected with DCF and
5a, 5b, 6b, and 9a were able to decrease the intracellular ROS levels, as DHE, respectively. Cultured cells were treated with trypsin re-sus-
measured by H2-DCFDA assay, in oligodendrocytes compared to vi- pended in PBS (106 cells/mL) and kept in the dark for half hour at 37 °C
tamin E. On the other hand, selenoquinones 2d, 3c, 7b, 9b and 10b with H2-DCFDA or DHE at 10 μM and 2 μM and cells were analyzed by
increased the ROS levels in oligodendrocytes. Furthermore, the apop- flow cytometry [55,56]. The green fluorescence of 2′,7′- dichloro-
totic/anti-apoptotic properties of the compounds were detected by flow fluorescein resulting from the oxidation of H2-DCFDA was analyzed by
cytometric estimation of the sub-G1 peak in oligodendrocytes treated flow cytometry and collected through a 520/10-nm band pass filter.
with the 7kc apoptosis inducer. Interestingly, a significant anti-apop- DHE, a non-fluorescent compound, rapidly oxidized in ethidium under
totic effect was observed in the case of compound 5a suggesting the the action of O2%− to give 2-hydroxyethidium that intercalates with
potential protective activity of this compound. Moreover, compounds DNA giving red fluorescence of ethidium collected through a 590/10-
5b and 7b displayed good GPx-like (≈1.5-fold more active than eb- nm band pass filter using an excitation of 488 nm and an emission
selen) which in turn may also account for their protective actions. These wavelength of 575 nm [55,56]. Flomax (Partec) or FlowJo (Tree Star
results were further confirmed by the ABTS and DPPH scavenging ac- Inc.) software were used for the data analysis.
tivity and the bleomycin dependent DNA damage assay, where most of
the compounds showed good scavenging activity with respect to as- 4.2.4. Apoptosis status and detection of Sub-G1 cells
corbic acid and protected DNA from damage; whereas, selenium-based The analysis of the cell cycle can reveal a Sub-G1 peak, which is
quinones exhibited low scavenging activity and caused DNA damage. representative of apoptotic cells. Cell cycle analysis was detected on
Together, the present results point to antioxidant properties of 158N cells stained with Propidium Iodide (PI).
some, non-cytotoxic, organic selenides resulting from their capacity to Flow cytometric analyses were measured on a Galaxy flow cyt-
reduce ROS generation. These compounds are considered promising ometer. Cells were prepared by re-suspension in cold ethanol (80%,
candidates and might provide a new avenue for innovative antioxidant −20 °C) for 2 h. Cells were then washed with phosphate buffer, and re-
research into prototype drugs for neurodegenerative disorders. suspended in phosphate buffer (300 μl) containing PI (80 μg/mL) and
RNase (200 μg/mL) and kept for 1 h at 37 °C.PBS 2 ml was then added,
4. Experimental section and Fluorescence of propidium iodide was recorded using a
590 ± 10 nm band filter. The percentage of cells in different cell cycle
4.1. Material and methods phase was manually estimated.

All chemicals were obtained from Sigma. Solvents were dried prior 4.2.5. Gpx-like activity
to use. The spectroscopic studies were measured at the “Pôle Chimie The GPx-kit was purchased from Biodiagnostic (Egypt) and used
Moléculaire” de l'Université de Bourgogne (PACSMUB)”. The mass of according to the reported method [86]. The sample was prepared by
the organic selenides was obtained on an Electro-Spray Ionization Mass adding assay buffer and NADPH reagent (0.1 ml; 24 µmol Glutathione,
Spectrometry (Thermo LTQ Orbitrap XL ESI–MS) and high-resolution 4.8 µmol NADPH and 12 units Glutathione reductase), and organic se-
mass spectra. 1H (300.13 MHz) and 13C (75.5 MHz) NMR spectra were lenides (0.01 ml, 41 µM). H2O2 (0.8 mM) was then added and the ab-
analyzed on Bruker 300 Avance III, spectrometers. The values of the sorbance was kinetically recorded every 1 min at wavelength 340 nm
chemical shifts (δ) are presented in parts per million relative to tetra- over three successive minutes. A340nm/min was calculated. Ebselen
methylsilane, using deuterated solvent as an internal standard. All (41 µM) was used as the positive control. For colored organoselenium
chemicals were purchased from Sigma e.g., 7-ketocholesterol, and as- compounds, their individual absorbance was subtracted at the used
corbic acid and vitamin E. More details on the spectroscopic data can be wavelength.
found in the Supporting Informations. 2-methyl-3-bromo-1,4-naph-
thaoquinone was synthesized according the reported literature method 4.2.6. Synthesis and characterization
[81]. Compound 1 was synthesized according to a literature reported 4.2.6.1. Synthesis of 2-methyl-3-bromo-l,4-naphthoquinone [81,84]. 2-
method [82,83,84]. Methyl-1,4-naphthoquinone (11 mg, 63 mmol), anhydrous sodium
acetate (1.1 mg, 34 mmol) were mixed together in glacial acetic acid
4.2. Biological assays (50 ml). Bromine (2 ml) was then added and the mixture was allowed to
settle in the dark for three days. A yellow crystalline mass formed,
4.2.1. Cytotoxicity assay which was removed. The supernatant liquid was poured on to 500 ml of
Murine oligodendrocytes (158N and 158JP) were seeded at 240,000 distilled water. The formed precipitate was filtered off and
cells per 24-well plates, and were grown using the Dulbecco’s Modified recrystallized from ethanol.
Eagle Medium supplemented with penicillin antibiotics (1% v/v, 4.2.6.1.1. General procedure Ib: Reduction of 1 and subsequent
streptomycin) and fetal bovine serum (5% v/v) and kept at 37 °C in modification of selenium based-amines (2a-c). Compound 1 (1 mmol)
atmosphere containing 5% CO2. The concentration of 7kc [57–59] and and appropriate halo derivative (2.2 mmol) were dissolved in EtOH
vitamin E, and the time of treatment were chosen based on data (20 ml). Sodium tetrahydridoborate (189.15 mg, 5 mmol) was added

51
S. Shaaban et al. Bioorganic Chemistry 80 (2018) 43–56

portion wise over 0.5 h. Then the reaction was reflux for additional 3 h. 4.2.6.2. 4-((2,2-Diethoxyethyl)selanyl)aniline (2a). Compound 2a was
The organic layer was dried and evaporated under vacuum. The residue prepared following procedure Ib from 1 (344 mg, 1 mmol),
was purified by silica gel chromatography. bromoacetaldehyde diethyl acetal (331 μl, 2.2 mmol), Aliquat 336
4.2.6.1.2. General procedure Ia: Reduction of 1 and subsequent (45 mg, 5% mol) and sodium tetrahydridoborate (189.15 mg,
modification of selenium based-amines (2d). Compound 1 (1 mmol) and 5 mmol). The progress of the product formation was followed by TLC
appropriate halo derivative (2.2 mmol) were dissolved in EtOAc petroleum ether: EtOAc = 6:1, Rf = 0.39, purified by column silica gel
(20 ml). Water (20 ml) and N-Methyl-N,N,N-trioctylammonium chromatography with petroleum ether: EtOAc = 6:1.5. Colorless oil;
chloride (Aliquat 336) (45 mg, 5% mol) were then added. Sodium Yield: 258.1 mg (89%). 1H NMR (300 MHz, CDCl3) δ 7.42–7.34 (d,
tetrahydridoborate (189.15 mg, 5 mmol) was added portion wise over J = 7.37 HZ, 2H, Ar-H), 6.62–6.55 (d, J = 6.58, 2H, Ar-H), 4.66 (t,
1 h. Then the reaction was stirred for additional 3 h. The organic layer J = 5.7 Hz, 1H, CH), 3.67–3.46 (m, 4H, 2CH2), 3.03 (d, J = 5.7 Hz, 2H,
was dried and evaporated under vacuum. The residue was purified by SeCH2), 1.19 (t, J = 7.1 Hz, 6H, 2CH3); 13C NMR (75 MHz, CDCl3) δ
silica gel chromatography. 146.17, 135.90, 117.11, 115.70, 102.30, 61.70, 32.23, 15.23; MS (ESI):
4.2.6.1.3. General procedure IIa: Preparation of N-acetyl derivatives m/z = found 312.04 [M++Na]; calcd. 289.06 [M+]; HRMS calcd. for
(4a-c). A mixture of formic acid (1.0–1.2 equivalents) and acetic C12H19NO2Se [M++Na]: 312.0498, found 312.04597 [M++Na].
anhydride (1.5–2.0 equivalents) was heated at 55 °C for 2 h and then
one equivalent of the appropriate amine (2a-d) was added. Heating was 4.2.6.3. 4-(Benzylselanyl)aniline (2b). Compound 2b was prepared
continued for another 2 h. After complete consumption of the starting following procedure Ib from 1 (344 mg, 1 mmol), benzyl chloride
material (as judged by TLC analysis), the reaction was cooled and (253 μl, 2.2 mmol), Aliquat 336 (45 mg, 5% mol) and sodium
poured onto water. The formed precipitated was collected and purified tetrahydridoborate (189.15 mg, 5 mmol). The progress of the product
by silica gel chromatography. formation was followed by TLC petroleum ether: EtOAc = 6:1,
4.2.6.1.4. General procedure IIb: Preparation of N-acetyl derivatives Rf = 0.36, purified by column silica gel chromatography with
(4a-c). Acetic anhydride (2.0 equivalents) was added to one equivalent petroleum ether: EtOAc = 6:1.5. Colorless oil; Yield: 223.55 mg
of the appropriate amine (2a-d) and the reaction was heated for two (85%). 1H NMR (300 MHz, CDCl3) δ 7.28–7.18 (m, 2H, Ar-H),
hours at 60 °C. The reaction was cooled and then poured onto ice. The 7.14–7.08 (m, 2H, Ar-H), 6.94–6.81 (m, 2H, Ar-H), 6.50–6.39 (m, 2H,
formed precipitated was purified by silica gel chromatography. Ar-H), 3.85 (s, 2H, SeCH2), 3.62 (s, 2H, NH2); 13C NMR (126 MHz,
4.2.6.1.5. General procedure III: Preparation of N-formyl derivatives CDCl3) δ 146.68, 138.65, 137.03, 131.32, 130.48, 120.33, 116.48,
(3a-c). A solution of the amine (1.0 mmol) and 85% aqueous formic 115.63, 32.51; MS (ESI): m/z = found 263.85 [M++1]; calcd. 263.02
acid (1.2 mmol) was heated at 60 °C for 2 h. After the disappearance of [M+]; HRMS calcd. for C13H13NSe [M++1]: 264.02860, found
the amine, the reaction was poured on to water and the crude N-formyl 264.02748 [M++1].
compound formed essentially as a solid product was purified by silica
gel chromatography. 4.2.6.4. 4-((4-Bromobenzyl)selanyl)aniline (2c). Compound 2c was
4.2.6.1.6. General procedure IV: Preparation of amide-acids (5a-b prepared following procedure Ib from 1 (344 mg, 1 mmol), 4-
and 6a-b). To a solution of amine (1 mmol) in dry toluene (5 ml), the bromobenzyl bromide (550 mg, 2.2 mmol), Aliquat 336 (45 mg,
appropriate anhydride (1 mmol) was added and the mixture was stirred 5% mol) and sodium tetrahydridoborate (189.15 mg, 5 mmol). The
at for 3 h. The amide-acid precipitate was collected and further washed progress of the product formation was followed by TLC petroleum
with hot-toluene and dried under vacuum. The purity of the amide- ether: EtOAc = 6:1, Rf = 0.36, purified by column silica gel
acids was satisfactory for further use. If necessary, further purification chromatography with petroleum ether: EtOAc = 6:1.5. White solid;
using silica gel chromatography was performed. Yield: 310.31 mg (91%); mp 156–158 °C. 1H NMR (300 MHz, DMSO) δ
4.2.6.1.7. General procedure V: preparation of cyclic imides (10a-b) 7.37–7.33 (m, 2H, Ar-H), 7.24–7.20 (m, 2H, Ar-H), 7.01–6.94 (m, 2H,
and/or isomaleimides (9a-b). To an appropriate amide-acid (one Ar-H), 6.59–6.55 (m, 2H, Ar-H), 3.90 (s, 2H, SeCH2); 13C NMR
equivalent) and 100 mg of sodium acetate, acetic anhydride (3 ml) (75 MHz, DMSO) δ 146.65, 138.63, 137.04, 131.31, 130.45, 120.32,
was added and the mixture was gently heated for 2 h at 50–60 °C. Ice 116.49, 115.61, 32.50; MS (ESI): m/z = found 364.84 [M++Na];
was added and the resulting mixture was extracted with CH2Cl2 calcd. 340.93 [M+]; HRMS calcd. for C13H12BrNSe [M++1]:
(200 ml), dried with Mg2SO4. CH2Cl2 was evaporated and the residue 341.93911, found 341.93760 [M++1].
was purified by silica gel chromatography.
4.2.6.1.8. General procedure VI: preparation of methyl ester (7a-b 4.2.6.5. 2-((4-Aminophenyl)selanyl)-3-methylnaphthalene-1,4-dione
and 8a). An appropriate amide-acid (1 mmol) was suspended in (2d). Compound 2d was prepared following procedure Ia from 1
methanol (10 ml) and then conc.H2SO4 (200 µl) was added and the (344 mg, 1 mmol), 2-methyl-3-bromo-l,4-naphthoquinone (550 mg,
reaction was stirred at ambient temperature for 8 h. The mixture was 2.2 mmol), Aliquat 336 (45 mg, 5% mol) and sodium
poured onto ice and the formed precipitate was purified by silica gel tetrahydridoborate (189.15 mg, 5 mmol). The progress of the product
chromatography. formation was followed by TLC petroleum ether: EtOAc = 4:1.5,
4.2.6.1.9. Crystallization and X-ray data collection of 9a. Colorless Rf = 0.32, purified by column silica gel chromatography with
single crystals of C17H12NO2SeBr were obtained by the slow petroleum ether: EtOAc = 3:1. Brown solid; Yield: 305.27 mg (89%);
evaporation method. A suitable crystal with dimensions of mp 162–164 °C. 1H NMR (300 MHz, DMSO) δ 7.97 (ddt, J = 11.8, 6.9,
0.37 × 0.37 × 0.12 mm3 was selected, mounted on glass fibers 3.4 Hz, 2H, Ar-H), 7.87–7.77 (m, 2H, Ar-H), 7.28–7.19 (m, 2H, Ar-H),
and diffraction data were collected on a Bruker D8 diffractometer 6.55–6.44 (m, 2H, Ar-H), 5.39 (s, 2H, NH2), 1.98 (s, 3H, CH3); 13C NMR
using graphite monochromated MoK/a radiation (λ = 0.71073 Å). (75 MHz, CDCl3) δ 181.84, 181.73, 149.10, 147.31, 147.24, 135.69,
The crystal was kept at 100 K during data collection. APEX2 [87] was 134.08, 133.77, 131.69, 131.65, 126.34, 126.27, 114.65, 111.92,
used for the unit cell determination and intensity data collection. Using 16.50; MS (ESI): m/z = found 344.95 [M++1]; calcd. 343.01 [M+];
Olex2 [88], the structure was solved with the ShelXT [89] and structure HRMS calcd. for C17H13NO2Se [M++Na]: 366.00037, found
solution program using Direct Methods and refined with the XL [89] 365.99930 [M++Na].
refinement package using Least Squares minimisation. For more details
regarding the crystal parameters and data collection statistics for 9a, 4.2.6.6. N-(4-((2,2-Diethoxyethyl)selanyl)phenyl)formamide
please refer to the supporting information. (3a). Compound 3a was prepared following procedure III from 2a

52
S. Shaaban et al. Bioorganic Chemistry 80 (2018) 43–56

(289 mg, 1 mmol) and 85% aqueous formic acid (1.2 mmol). The 134.54, 128.50, 127.87, 126.31, 124.32, 120.06, 31.70, 22.42; MS
progress of the product formation was followed by TLC petroleum (ESI): m/z = found 327.94 [M++Na]; calcd. 305.03 [M+]; HRMS
ether: EtOAc = 8:1, Rf = 0.40, purified by column silica gel calcd. for C15H15NOSe [M++ Na]: 328.02111, found 328.01998
chromatography with petroleum ether: EtOAc = 8:1.5. Brown solid; [M++Na].
Yield: 259.94 mg (82%); mp 175–177 °C. 1H NMR (300 MHz, CDCl3) δ
8.66 (s, 1H, CHO), 8.28 (s, 1H, NH), 7.50–7.34 (m, 3H, Ar-H), 4.2.6.11. N-(4-((3-Methyl-1,4-dioxo-1,4-dihydronaphthalen-2-yl)selanyl)
6.96–6.88 (m, 1H, Ar-H), 4.62 (td, J = 5.6, 3.0 Hz, 1H, CH), phenyl)acetamide (4c). Compound 4c was prepared following
3.66–3.40 (m, 4H, 2CH2), 3.00 (d, J = 5.6 Hz, 2H, SeCH2), 1.12 (t, procedure II from 2-((4-aminophenyl)selanyl)-3-methylnaphthalene-
J = 7.1 Hz, 6H, 2CH3); 13C NMR (75 MHz, CDCl3) δ 162.52, 159.18, 1,4-dione 2d (344 mg, 1 mmol) and acetic anhydride (2.0
136.16, 135.92, 134.47, 133.84, 126.57, 125.76, 120.55, 119.28, equivalents). The progress of the product formation was followed by
102.28, 62.13, 31.64, 15.22; MS (ESI): m/z = found 339.95 TLC petroleum ether: EtOAc = 8:1, Rf = 0.43, purified by column silica
[M++Na]; calcd. 317.05 [M+]; HRMS calcd. for C13H19NO3Se [M+]: gel chromatography with petroleum ether: EtOAc = 8:3. Brown solid;
317.05123, found 199.96062 [M+-CH2CH(OC2H5)2]. Yield: 288.75 mg (75%); mp 125–127 °C. 1H NMR (300 MHz, DMSO) δ
8.04–7.92 (m, 2H, Ar-H), 7.89–7.78 (m, 2H, Ar-H), 7.49 (qd, J = 6.7,
4.2.6.7. N-(4-((4-Bromobenzyl)selanyl)phenyl)formamide 3.4 Hz, 4H, Ar-H), 2.07 (s, 3H, CH3), 2.03 (s, 3H, CH3); 13C NMR
(3b). Compound 3b was prepared following procedure III from 2c (75 MHz, DMSO) δ 182.33, 181.60, 168.89, 149.50, 146.11, 139.51,
(341 mg, 1 mmol) and 85% aqueous formic acid (1.2 mmol). The 134.57, 134.01, 132.16, 126.96, 122.88, 120.29, 24.47, 17.96; MS
progress of the product formation was followed by TLC petroleum (ESI): m/z = found 384.83 [M+]; calcd. 385.02 [M+]; HRMS calcd. for
ether: EtOAc = 8:1, Rf = 0.36, purified by column silica gel C19H15NO3Se [M++Na]: 408.01094, found 408.00933 [M++Na].
chromatography with petroleum ether: EtOAc = 6:1. White solid;
Yield: 324.72 mg (88%); mp 158–160 °C.1H NMR (300 MHz, MeOD) δ 4.2.6.12. (E)-4-((4-((4-Bromobenzyl)selanyl)phenyl)amino)-4-oxobut-2-
8.47 (s, 1H, CHO), 7.56–7.38 (m, 5H, Ar-H), 7.13–6.97 (m, 3H, Ar-H), enoic acid (5a). Compound 5a was prepared following procedure IV
4.11 (s, 2H, SeCH2); 13C NMR (75 MHz, MeOD) δ 167.77, 132.13, from 4-((4-bromobenzyl)selanyl)aniline 2c (344 mg, 1 mmol) and
131.46, 127.58, 127.54, 126.51, 116.38, 115.17, 28.02; MS (ESI): m/ maleic anhydride (98 mg, 1 mmol). Its formation was monitored by
z = found 391.86 [M++Na]; calcd. 368.93 [M+]; HRMS calcd. for TLC chloroform: methanol = 8:1, Rf = 0.36, purified by column silica
C14H12BrNOSe [M++Na]: 391.91597, found 391.91555 [M++Na]. gel chromatography with chloroform: methanol = 6:1. Yellow solid;
Yield: 412.66 mg (94%); mp 215–217 °C. 1H NMR (300 MHz, DMSO) δ
4.2.6.8. N-(4-((3-Methyl-1,4-dioxo-1,4-dihydronaphthalen-2-yl)selanyl) 10.45 (s, 1H, COOH), 7.55 (d, J = 8.6 Hz, 2H, Ar-H), 7.46–7.37 (m, 4H,
phenyl)formamide (3c). Compound 3c was prepared following Ar-H), 7.17 (d, J = 8.4 Hz, 2H, Ar-H), 6.45 (d, J = 12.1 Hz, 1H, =CH),
procedure III from 2d (343 mg, 1 mmol) and 85% aqueous formic 6.30 (d, J = 12.0 Hz, 1H, CH=), 4.04 (s, 2H, SeCH2); 13C NMR
acid (1.2 mmol). The progress of the product formation was followed by (75 MHz, DMSO) δ 167.31, 163.71, 139.14, 138.42, 134.02, 132.06,
TLC petroleum ether: EtOAc = 8:1, Rf = 0.36, purified by column silica 131.60, 131.35, 130.90, 124.14, 120.54, 120.15, 30.71; MS (ESI): m/
gel chromatography with petroleum ether: EtOAc = 8:3. Brown solid; z = found 439.97 [M++1]; calcd. 438.93 [M+]; HRMS calcd. for
Yield: 300.51 mg (81%); mp 168–170 °C. 1H NMR (300 MHz, DMSO) δ C17H14BrNO3Se [M++Na]: 461.92145, found 461.92022 [M++Na].
10.26 (s, 1H, CHO), 8.27 (d, J = 1.9 Hz, 1H, CHO), 8.04–7.97 (m, 2H,
Ar-H), 7.85–7.80 (m, 2H, Ar-H), 7.52 (t, J = 2.5 Hz, 4H, Ar-H), 2.09 (s, 4.2.6.13. (Z)-4-((4-((3-Methyl-1,4-dioxo-1,4-dihydronaphthalen-2-yl)
3H, CH3); 13C NMR (75 MHz, DMSO) δ 182.33, 181.52, 160.17, 149.86, selanyl)phenyl)amino)-4-oxobut-2-enoic acid (5b). Compound 5b was
138.32, 134.57, 134.03, 132.25, 126.98, 126.78, 123.89, 120.52, prepared following procedure IV from 2-((4-aminophenyl)selanyl)-3-
18.06; MS (ESI): m/z = found394.01 [M++Na]; calcd. 371.01 [M+]; methylnaphthalene-1,4-dione 2d (343 mg, 1 mmol) and maleic
HRMS calcd. for C18H13NO3Se [M++Na]: 393.99529, found anhydride (98 mg, 1 mmol). Its formation was monitored by TLC
393.99469 [M++Na]. chloroform: methanol = 8:1, Rf = 0.36, purified by column silica gel
chromatography with petrol chloroform: methanol = 6:1. Brown solid;
4.2.6.9. N-(4-((2,2-diethoxyethyl)selanyl)phenyl)acetamide Yield: 401.31 mg (91%); mp 207–209 °C. 1H NMR (300 MHz, DMSO) δ
(4a). Compound 4a was prepared following procedure IIa from 2a 10.43 (s, 1H, COOH), 8.07–7.93 (m, 2H, Ar-H), 7.90–7.77 (m, 2H, Ar-
(289 mg, 1 mmol) and acetic anhydride (2.0 equivalents). The progress H), 7.63–7.51 (m, 2H, Ar-H), 7.29–7.10 (m, 2H, Ar-), 6.46 (dd,
of the product formation was followed by TLC petroleum ether: J = 12.0, 2.6 Hz, 1H, =CH), 6.30 (dd, J = 12.0, 1.8 Hz, 1H, CH=),
EtOAc = 4:3, Rf = 0.30, purified by column silica gel 2.30 (s, 1H, NH), 2.09 (s, 3H, CH3); 13C NMR (75 MHz, DMSO) δ
chromatography with petroleum ether: EtOAc = 1:1. Yellow solid; 182.33, 181.52, 167.28, 149.94, 134.58, 134.35, 133.86, 132.27,
Yield: 294.84 mg (89%); mp 165–167 °C. 1H NMR (300 MHz, CDCl3) 129.36, 128.67, 127.00, 126.78, 125.78, 124.20, 120.79, 18.10; MS
δ 8.37 (s, 1H, NH), 7.40–7.32 (m, 4H, Ar-H), 4.60 (t, J = 5.6 Hz, 1H, (ESI): m/z = found 464.03 [M++Na]; calcd. 441.01 [M+]; HRMS
CH), 3.63–3.37 (m, 4H, 2CH2), 3.01–2.93 (m, 2H, CH2), 2.06 (s, 3H, calcd. for C21H15NO5Se [M++1]: 464.00077, found 463.99922
COCH3), 1.10 (t, J = 7.1 Hz, 6H, 2CH3); 13C NMR (75 MHz, CDCl3) δ [M++Na].
169.13 (s), 137.42 (s), 133.75 (s), 124.93 (s), 120.70 (s), 102.29 (s),
62.14 (s), 31.68 (s), 24.37 (s), 15.23 (s); MS (ESI): m/z = found 376.06 4.2.6.14. 4-((4-((4-Bromobenzyl)selanyl)phenyl)amino)-4-oxobutanoic
[M++2Na]; calcd. 331.07 [M+]; HRMS calcd. for C14H21NO3Se [M+]: acid (6a). Compound 62 was prepared following procedure IV from 2c
331.0687, found 331.06812 [M+]. (344 mg, 1 mmol) and succinic anhydride (100 mg, 1 mmol). Its
formation was monitored by TLC chloroform: methanol = 8:1,
4.2.6.10. N-(4-(Benzylselanyl)phenyl)acetamide (4b). Compound 4b Rf = 0.36, purified by column silica gel chromatography with
was prepared following procedure II from 4-(benzylselanyl)aniline 2b chloroform: methanol = 6:1. White solid; Yield: 396.9 mg (90%); mp
(263 mg, 1 mmol) and acetic anhydride (2.0 equivalents). The progress 258–260 °C. 1H NMR (300 MHz, DMSO) δ 12.13 (s, 1H, COOH), 10.00
of the product formation was followed by TLC petroleum ether: (s, 1H, NH), 7.50 (d, J = 8.7 Hz, 2H, Ar-H), 7.45–7.33 (m, 4H, Ar-H),
EtOAc = 8:1, Rf = 0.42, purified by column silica gel 7.17–7.12 (m, 2H, Ar-H), 4.14 (s, 2H, SeCH2), 2.54–2.51 (m, 4H, 2CH2);
chromatography with petroleum ether: EtOAc = 8:1.5. White solid; 13
C NMR (75 MHz, DMSO) δ 174.24, 170.64, 139.23, 134.26, 131.57,
Yield: 280.6 mg (92%); mp 180–182 °C. 1H NMR (300 MHz, DMSOd6) δ 131.32, 122.77, 120.09, 119.97, 31.53, 30.84, 29.20; MS (ESI): m/
6.83–6.55 (m, 4H, Ar-H), 6.49–6.41 (m, 4H, Ar-H), 4.11 (s, 2H, SeCH2), z = found 463.99 [M++1]; calcd. 440.95 [M+]; HRMS calcd. for
1.39 (s, 3H, CH3); 13C NMR (75 MHz, MeOD) δ 170.32, 139.06, 138.17, C17H16BrNO3Se [M++Na]: 463.93710, found 463.93534 [M++Na].

53
S. Shaaban et al. Bioorganic Chemistry 80 (2018) 43–56

4.2.6.15. 4-((4-((3-Methyl-1,4-dioxo-1,4-dihydronaphthalen-2-yl) Rf = 0.36, purified by column silica gel chromatography with


selanyl)phenyl)amino)-4-oxobutanoic acid (6b). Compound 6b was petroleum ether: EtOAc = 6:1. Brown solid; Yield: 323 mg (76%); mp
prepared following procedure IV from 2d (344 mg, 1 mmol)and 224–226 °C. 1H NMR (300 MHz, CDCl3) δ 8.07–7.95 (m, 2H, Ar-H),
succinic anhydride (100 mg, 1 mmol). Its formation was monitored by 7.68–7.59 (m, 2H, Ar-H), 7.57–7.51 (m, 2H, Ar-H), 7.21–7.15 (m, 2H,
TLC chloroform: methanol = 8:1, Rf = 0.36, purified by column silica Ar-H), 2.81 (s, 4H, 2CH2), 2.21 (s, 3H, CH3); 13C NMR (75 MHz, CDCl3)
gel chromatography with chloroform: methanol = 6:1. Brown solid; δ 182.45, 181.23, 175.76, 150.72, 145.96, 133.76, 133.67, 132.18,
Yield: 411.99 mg (93%); mp 208–210 °C. 1H NMR (300 MHz, DMSO) δ 131.99, 131.50, 130.17, 127.25, 127.05, 126.80, 28.37, 18.24; MS
12.11 (s, 1H, COOH), 10.04 (s, 1H, NH), 8.04–7.92 (m, 2H, Ar-H), (ESI): m/z = found 447.38 [M++Na]; calcd. 425.02 [M+]; HRMS
7.87–7.77 (m, 2H, Ar-H), 7.56–7.45 (m, 4H, Ar-H), 2.52 (dd, J = 6.8, calcd. for C21H15NO4Se [M++1]: 448.00585, found 448.00451
2.5 Hz, 4H, 2CH2), 2.07 (s, 3H, CH3); 13C NMR (75 MHz, DMSO) δ [M++Na].
182.33, 181.60, 173.27, 170.45, 149.63, 146.08, 139.38, 134.33,
134.04, 132.19, 126.97, 120.27, 51.82, 31.35, 28.87, 17.96; MS 4.2.6.20. (E)-Methyl 4-((4-((4-bromobenzyl)selanyl)phenyl)amino)-4-
(ESI): m/z = found 466.05 [M++Na]; calcd. 443.03 [M+]; HRMS oxobut-2-enoate (7a). Compound 7a was prepared following
calcd. for C21H17NO5Se [M++Na]: 466.01642, found 466.01484 procedure VI from 5a (439 mg, 1 mmol) in methanol (10 ml) and
[M++Na]. conc.H2SO4 (200 µl). The progress of the product formation was
followed by TLC petroleum ether: EtOAc = 8:1, Rf = 0.43, purified by
4.2.6.16. (Z)-5-((4-((4-Bromobenzyl)selanyl)phenyl)imino)-1H-pyrrol- column silica gel chromatography with petroleum ether: EtOAc = 6:1.
2(5H)-one (9a). Compound 9a was prepared following procedure V Brown solid; Yield: 389.58 mg (86%); mp 190–192 °C. 1H NMR
from 5a (439 mg, 1 mmol), acetic anhydride (3 ml) and 100 mg of (300 MHz, DMSO) δ 11.05 (s, 1H, NH), 7.59 (d, J = 8.6 Hz, 2H, Ar-
sodium acetate. The progress of the product formation was followed by H), 7.46–7.34 (m, 4H, Ar-H), 7.08–6.97 (m, 2H, Ar-H), 6.46 (d,
TLC petroleum ether: EtOAc = 8:1, Rf = 0.45, purified by column silica J = 13.4 Hz, 1H, CH=), 6.26 (d, J = 13.4 Hz, 1H, =CH), 4.02 (s, 2H,
gel chromatography with petroleum ether: EtOAc = 6:1. Yellow solid; SeCH2), 3.88 (s, 3H, OCH3); 13C NMR (75 MHz, DMSO) δ 162.60,
Yield: 324.17 mg (77%), mp 185–187 °C. 1H NMR (300 MHz, CDCl3) δ 156.62, 135.86, 133.28, 132.95, 130.56, 126.72, 125.69, 120.22,
7.47–7.29 (m, 4H, Ar-H), 7.25–7.14 (m, 2H, Ar-H), 7.04–6.96 (m, 2H, 119.87, 115.87, 115.76, 48.12, 27.25; MS (ESI): m/z = found451.91
Ar-H), 6.76 (d, J = 5.5 Hz, 1H, CH=), 6.62 (d, J = 5.5 Hz, 1H, CH=), [M+]; calcd. 452.95 [M+]; HRMS calcd. for C18H16BrNO3Se [M++1]:
4.01 (s, 2H, SeCH2); 13C NMR (75 MHz, CDCl3) δ 166.94, 150.34, 475.93710, found 475.93486 [M++Na].
143.16, 142.77, 137.50, 134.29, 134.02, 131.59, 130.49, 127.93,
125.94, 120.83, 31.52; MS (ESI): m/z = found 474.92 [M++CH3OH 4.2.6.21. (Z)-Methyl 4-((4-((3-methyl-1,4-dioxo-1,4-dihydronaphthalen-
+Na]; calcd. 420.92 [M+]; HRMS calcd. for C17H12BrNO2Se [M++1]: 2-yl)selanyl)phenyl)amino)-4-oxobut-2-enoate (7b). Compound 7b was
475.93688, found 475.93544 [M++CH3OH+Na]. prepared following procedure VI from 5b (441 mg, 1 mmol) in
methanol (10 ml) and conc.H2SO4 (200 µl). The progress of the
4.2.6.17. (E)-2-Methyl-3-((4-((5-oxofuran-2(5H)-ylidene)amino)phenyl) product formation was followed by TLC petroleum ether:
selanyl)naphthalene-1,4-dione (9b). Compound 9b was prepared EtOAc = 8:1, Rf = 0.36, purified by column silica gel
following procedure V from 5b (441 mg, 1 mmol), acetic anhydride chromatography with petroleum ether: EtOAc = 6:1. Yellow solid;
(3 ml) and 100 mg of sodium acetate. The progress of the product Yield: 382.2 mg (84%); mp 208–210 °C. 1H NMR (300 MHz, CDCl3) δ
formation was followed by TLC petroleum ether: EtOAc = 8:1, 10.98 (s, 1H, NH), 8.05–7.92 (m, 2H, Ar-H), 7.66–7.57 (m, 2H, Ar-H),
Rf = 0.46, purified by column silica gel chromatography with 7.54 (d, J = 8.7 Hz, 2H, Ar-H), 7.48–7.41 (m, 2H, Ar-H), 6.35 (dd,
petroleum ether: EtOAc = 6:1. Brown solid; Yield: 317.25 mg (75%); J = 13.4, 3.3 Hz, 1H, CH=), 6.20–6.12 (dd, J = 13.4, 3.3 Hz, 1H,
mp 200–202 °C. 1H NMR (300 MHz, CDCl3) δ 8.09–7.94 (m, 2H, Ar-H), =CH), 3.78 (s, 3H, OCH3), 2.12 (s, 3H, CH3); 13C NMR (75 MHz,
7.69–7.59 (m, 2H, Ar-H), 7.57–7.42 (m, 2H, Ar-H), 7.35–7.20 (m, 2H, CDCl3) δ 181.41, 180.63, 166.33, 160.39, 148.36, 146.05, 139.42,
Ar-H), 6.78 (d, J = 5.5 Hz, 1H, CH=), 6.67 (d, J = 5.5 Hz, 1H, CH=), 136.89, 133.68, 132.68, 132.49, 131.26, 131.04, 126.06, 125.73,
2.23 (s, 3H, CH3); 13C NMR (75 MHz, CDCl3) δ 182.44, 181.31, 169.11, 124.08, 123.46, 119.79, 51.87, 16.68; MS (ESI): m/z = found 478.04
150.53, 146.11, 134.29, 133.89, 133.80, 133.66, 132.20, 132.00, [M++Na]; calcd. 455.03 [M+]; HRMS calcd. for C22H17NO5Se
127.23, 126.81, 126.42, 121.59, 18.17; MS (ESI): m/z = found [M++1]: 478.01642, found 478.01544 [M++Na].
478.03 [M++CH3OH+Na]; calcd. 423.00 [M+]; HRMS calcd. for
C21H13NO4Se [M+]: 420.92, found 477.9328 [M++CH3OH+Na]. 4.2.6.22. Methyl 4-((4-((4-bromobenzyl)selanyl)phenyl)amino)-4-
oxobutanoate (8a). Compound 8a was prepared following
4.2.6.18. 1-(4-((4-Bromobenzyl)selanyl)phenyl)pyrrolidine-2,5-dione procedure VI from 4-((4-((4-bromobenzyl)selanyl)phenyl)amino)-4-
(10a). Compound 10a was prepared following procedure V from 6a oxobutanoic acid 6a (441 mg, 1 mmol) in methanol (10 ml) and
(441 mg, 1 mmol), acetic anhydride (3 ml) and 100 mg of sodium conc.H2SO4 (200 µl). The progress of the product formation was
acetate. The progress of the product formation was followed by TLC followed by TLC petroleum ether: EtOAc = 8:1, Rf = 0.41, purified
petroleum ether: EtOAc = 8:1, Rf = 0.44, purified by column silica gel by column silica gel chromatography with petroleum ether:
chromatography with petroleum ether: EtOAc = 6:1. White solid; Yield: EtOAc = 6:1. White solid; Yield: 395.85 mg (87%); mp 185–187 °C.
321.48 mg (76%); mp 205–207 °C. 1H NMR (300 MHz, CDCl3) δ 1
H NMR (300 MHz, CDCl3) δ 7.64 (s, 1H, NH), 7.37–7.20 (m, 6H, Ar-
7.49–7.41 (m, 2H, Ar-H), 7.34–7.26 (m, 2H, Ar-H), 7.15–7.08 (m, 2H, H), 6.97–6.87 (m, 2H, Ar-H), 3.92 (s, 2H, SeCH2), 3.64 (s, 3H, OCH3),
Ar-H), 7.06–6.98 (m, 2H, Ar-H), 4.03 (s, 2H, SeCH2), 2.82 (s, 4H, 2.73–2.53 (m, 4H, 2CH2); 13C NMR (75 MHz, CDCl3) δ 173.67,
2CH2); 13C NMR (75 MHz, CDCl3) δ 174.87, 136.31, 132.99, 130.62, 169.75, 138.01, 137.74, 135.34, 131.45, 130.45, 124.10, 120.60,
130.17, 129.82, 129.51, 125.85, 119.87, 30.52, 27.39; MS (ESI): m/ 120.17, 52.05, 32.16, 32.00, 29.19; MS (ESI): m/z = found 477.98
z = found 445.93 [M++Na]; calcd. 422.94 [M+]; HRMS calcd. for [M++Na]; calcd. 454.96 [M+]; HRMS calcd. for C18H18BrNO3Se
C17H14BrNO2Se [M++1]: 445.92653, found 445.92580 [M++Na]. [M++1]: 477.95275, found 477.95070 [M++1].

4.2.6.19. 1-(4-((3-Methyl-1,4-dioxo-1,4-dihydronaphthalen-2-yl)selanyl) Acknowledgements


phenyl)pyrrolidine-2,5-dione (10b). Compound 10b was prepared
following procedure V from 6b (443 mg, 1 mmol), acetic anhydride This work was supported by the “Ministère des Affaires Etrangères:
(3 ml) and 100 mg of sodium acetate. The progress of the product the Embassy of France in Egypt-Institut Français en Egypte” and the
formation was followed by TLC petroleum ether: EtOAc = 8:1, “Science and Technology Development Fund” by according to a

54
S. Shaaban et al. Bioorganic Chemistry 80 (2018) 43–56

postdoctoral fellowship to S. Shaaban. The authors thank the Conseil (c) S. Kumar, J. Yan, J.F. Poon, V.P. Singh, X. Lu, O.M. Karlsson, L. Engman,
Régional de Bourgogne (France) and the Ministère de l’Enseignement et S. Kumar, Multifunctional antioxidants: regenerable radical-trapping and hydro-
peroxide-decomposing ebselenols, Angew. Chem. Int. Ed. 7 (2016) 3793–3797.
de la Recherche (France) for their financial supports. The authors thank [17] B.R. Cardoso, B.R. Roberts, A.I. Bush, D.J. Hare, Selenium, selenoproteins and
also the Egyptian Ministry of Higher Education and Mansoura neurodegenerative diseases, Metallomics 7 (2015) 1213–1228.
University for financial support. This article is based upon work from [18] H. Balaban, M. Nazıroğlu, K. Demirci, İ.S. Övey, The protective role of selenium on
scopolamine-induced memory impairment, oxidative stress, and apoptosis in aged
COST Action CA16112-NutRedOx (Personalized Nutrition in aging society: rats: the involvement of TRPM2 and TRPV1 channels, Mol. Neurobiol. 1 (2017)
redox control of major age-related diseases), supported by COST 2852–2868.
(European Cooperation in Science and Technology). [19] J.E. Yeo, J.H. Kim, S.K. Kang, Selenium attenuates ROS-mediated apoptotic cell
death of injured spinal cord through prevention of mitochondria dysfunction; in
vitro and in vivo study, Cell. Physiol. Biochem. 21 (2008) 225–238.
Appendix A. Supplementary material [20] L. Savarino, D. Granchi, G. Ciapetti, E. Cenni, G. Ravaglia, P. Forti, F. Maioli,
R. Mattioli, Serum concentrations of zinc and selenium in elderly people: results in
healthy nonagenarians/centenarians, Exp. Gerontol. 36 (2001) 327–339.
Supplementary data associated with this article can be found, in the
[21] J. Gu, J.E. Royland, R.C. Wiggins, G.W. Konat, Selenium is required for normal
online version, at https://doi.org/10.1016/j.bioorg.2018.05.019. upregulation of myelin genes in differentiating oligodendrocytes, J. Neurosci. Res.
15 (47) (1997) 626–635.
References [22] P.A. Eccleston, D.H. Silberberg, The differentiation of oligodendrocytes in a serum-
free hormone-supplementedmedium, Brain. Res. 318 (1) (1984) 1–9.
[23] J.E. Bottenstein, Growth requirements in vitro of oligodendrocyte cell lines and
[1] (a) I. Coman, G. Barbin, P. Charles, B. Zalc, C. Lubetzki, Axonal signals in central neonatal rat brain oligodendrocytes, Proc. Natl Acad. Sci. USA 83 (6) (1986)
nervous system myelination demyelination remyelination, J. Neurol. Sci. 233 1955–1959.
(2005) 67–71; [24] (a) D.C. Mahan, Effect of organic and inorganic selenium sources and levels on sow
(b) V.A. Pavlov, K.J. Tracey, Neural ron of immunity: molecular mechanisms and colostrum and milk selenium content, J. Anim. Sci. 78 (2000) 100–105;
clinical translation, Nat. Neurosci. 20 (2017) 156–166. (b) L. Sancineto, A. Mariotti, L. Bagnoli, F. Marini, J. Desantis, N. Iraci, C. Santi,
[2] Y. Liu, J. Zhou, Oligodendrocytes in neurodegenerative diseases, Front. Biol. 8 C. Pannecouque, O. Tabarrini, Design and synthesis of diselenobisbenzamides
(2013) 127–133. (DISeBAs) as nucleocapsid protein 7 (NCp7) inhibitors with anti-HIV activity, J.
[3] (a) M. Bradl, H. Lassmann, Oligodendrocytes: biology and pathology, Acta Med. Chem. 10 (2015) 9601–9614.
Neuropathol. 119 (2009) 37–53; [25] (a) S. Pinton, C.A. Brüning, C.E. Sartori Oliveira, M. Prigol, C.W. Nogueira,
(b) R. Van den Berg, C.C. Hoogenraad, R.Q. Hintzen, Axonal transport deficits in Therapeutic effect of organoselenium dietary supplementation in a sporadic de-
multiple sclerosis: spiraling into the abyss, Acta Neuropathol. 134 (2017) 1–14. mentia of Alzheimer's Type model in rats, J. Nutr. Biochem. 24 (2013) 311–317;
[4] B. El Waly, M. Macchi, M. Cayre, P. Durbec, Oligodendrogenesis in the normal and (b) M.C. Kahya, M. Nazıroğlu, İ.S. Övey, Modulation of diabetes-induced oxidative
pathological central nervous system, Front. Neurosci. 8 (2014) 145. stress, apoptosis, and Ca2+ entry through TRPM2 and TRPV1 channels in dorsal
[5] (a) G. Lizard, O. Rouaud, J. Demarquoy, M. Cherkaoui-Malki, L. Iuliano, Potential root ganglion and hippocampus of diabetic rats by melatonin and selenium, Mol.
roles of peroxisomes in alzheimer's disease and in dementia of the Alzheimer's Type, Neurobiol. (2017) 12345–12360.
J. Alzheimers Dis. 29 (2012) 241–254; [26] E.C. Stangherlin, C. Luchese, S. Pinton, J.B.T. Rocha, C.W. Nogueira, Sub-chronical
(b) R.L. Doty, Olfactory dysfunction in neurodegenerative diseases: is there a exposure to diphenyl diselenide enhances acquisition and retention of spatial
common pathological substrate? Lancet Neurol. 16 (2017) 478–488. memory in rats, Brain Res. 1201 (2008) 106–113.
[6] N. Edgar, E. Sibille, A putative functional role for oligodendrocytes in mood reg- [27] S. Shaaban, A. Negm, M.A. Sobh, L.A. Wessjohann, Expeditious entry to functio-
ulation, Transl. Psychiatry. 2 (2012) e109. nalized pseudo-peptidic organoselenide redox modulators via sequential Ugi/SN
[7] M. Simons, K.A. Nave, Oligodendrocytes: myelination and axonal support, Cold. methodology, Anticancer Agents Med. Chem. 16 (5) (2016) 621–632.
Spring. Harb. Perspect. Biol. 22 (8) (2015) a020479. [28] (a) S. Shaaban, E.H. Gaffer, M. Alshahd, S.S. Elmorsy, Cytotoxic symmetrical
[8] J. Vamecq, A. Dessein, M. Fontaine, G. Briand, L. Porchet Norbert, P. Andreolotti, thiazolediselenides with increased selectivity against MCF-7 breast cancer cells, Int.
M. Cherkaoui-Malki, Mitochondrial dysfunction and lipid homeostasis, Curr. Drug. J. Res. Develp. Pharm & Life Sci 4 (2015) 1654–1668;
Metab 13 (2012) 1388–1400. (b) W.S. Hamama, M.A. Ismail, M. Soliman, S. Shaaban, H.H. Zoorob, Behavior of
[9] R.J. Mailloux, Teaching the fundamentals of electron transfer reactions in mi- 2-iminothiazolidin-4-one with different reagents, J. Het. Chem. 48 (5) (2011)
tochondria and the production and detection of reactive oxygen species, Redox. 1169–1174.
Biol. 4 (2015) 381–398. [29] S. Shaaban, E.H. Gaffer, Y. Jabar, S.S. Elmorsy, Cytotoxic naphthalene based-
[10] (a) A.M. Pisoschi, A. Pop, The role of antioxidants in the chemistry of oxidative symmetrical diselenides with increased selectivity against MCF-7 breast cancer
stress, Eur. J. Med. Chem. 97 (2015) 55–74; cells, Int. J. Pharm. 5 (2015) 738–746.
(b) V.S.S.S. Sajja, H. Nora, J.V.V. Pamela, Role of glia in memory deficits following [30] S. Shaaban, A. Negm, M.A. Sobh, L.A. Wessjohann, Organoselenocyanates and
traumatic brain injury: biomarkers of glia dysfunction, Front. Integr. Neurosci. 10 symmetrical diselenides redox modulators: design, synthesis and biological eva-
(2016) 7. luation, Eur. J. Med. Chem. 97 (2015) 190–201.
[11] E.K. Wirth, M. Conrad, J. Winterer, C. Wozny, B.A. Carlson, S. Roth, D. Schmitz, [31] V. Nascimento, E.E. Alberto, D.W. Tondo, D. Dambrowski, M.R. Detty, F. Nome,
G.W. Bornkamm, V. Coppola, L. Tessarollo, L. Schomburg, J. Köhrle, D.L. Hatfield, A.L. Braga, GPx-Like activity of selenides and selenoxides: experimental evidence
U. Schweizer, Neuronal selenoprotein expression is required for interneuron de- for the involvement of hydroxy perhydroxy selenane as the active species, J. Am.
velopment and prevents seizures and neurodegeneration, FASEB J. 24 (2009) Chem. Soc. 9 (2011) 138–141.
844–852. [32] S. Shaaban, F. Sasse, T. Burkholz, C. Jacob, Sulfur, selenium and tellurium pseu-
[12] L.V. Papp, J. Lu, A. Holmgren, K.K. Khanna, From selenium to selenoproteins: dopeptides: synthesis and biological evaluation, Bioorg. Med. Chem. 22 (2014)
synthesis, identity, and their role in human health, Antioxid. Redox Signal. 9 (2007) 3610–3619.
775–806. [33] S. Mecklenburg, S. Shaaban, L.A. Ba, T. Burkholz, T. Schneider, B. Diesel,
[13] (a) H. Vural, H. Demirin, Y. Kara, I. Eren, N. Delibas, Alterations of plasma mag- A.K. Kiemer, A. Röseler, K. Becker, J. Reichrath, Exploring synthetic avenues for the
nesium, copper, zinc, iron and selenium concentrations and some related ery- effective synthesis of selenium-and tellurium-containing multifunctional redox
throcyte antioxidant enzyme activities in patients with Alzheimer’s disease, J. Trace agents, Org. Biomol. Chem. 7 (2009) 4753–4762.
Elem. Med. Biol. 24 (2010) 169–173; [34] S. Shabaan, L.A. Ba, M. Abbas, T. Burkholz, A. Denkert, A. Gohr, L.A. Wessjohann,
(b) H.J. Reich, J.H. Robert, Why nature chose selenium, ACS. Chem. Biol. 4 (2016) F. Sasse, W. Weber, C. Jacob, Multicomponent reactions for the synthesis of mul-
821–841. tifunctional agents with activity against cancer cells, Chem. Commun. (Camb) 31
[14] (a) C. Berr, A. Nicole, J. Godin, I. Ceballos-Picot, M. Thevenin, J.F. Dartigues, (2009) 4702–4704.
A. Alperovitch, Selenium and oxygen-metabolizing enzymes in elderly community [35] S. Shaaban, Synthesis and Biological Activity of Multifunctional Sensor/Effector
residents: a pilot epidemiological study, J. Am. Geriatr. Soc. 41 (1993) 143–148; Catalysts, Dissertation, 2011.
(b) K. Arai, T. Takei, M. Okumura, S. Watanabe, Y. Amagai, Y. Asahina, [36] S. Shaaban, R. Diestel, B. Hinkelmann, Y. Muthukumar, R.P. Verma, F. Sasse,
L. Moroder, H. Hojo, K. Inaba, M. Iwaoka, Preparation of selenoinsulin as a long- C. Jacob, Novel peptidomimetic compounds containing redox active chalcogens and
lasting insulin analogue, Angew. Chem. Int. Ed. 56 (2017) 5522–5526; quinones as potential anticancer agents, Eur. J. Med. Chem. 58 (2012) 192–205.
(c) D. de Souza, O.M. Douglas, N. Fernanda, S. Eduarda, F.C. Vinícius, S. Fabiana, [37] A. Toshimitsu, S. Uemura, Organic selenium and tellurium compounds, organic
S.S. Rafael, et al., New organochalcogen multitarget drug: synthesis and antioxidant selenocyanates, tellurocyanates and related compounds, Patai's Chem. Funct
and antitumoral activities of chalcogenozidovudine derivatives, J. Med. Chem. 58 Groups, John Wiley & Sons Ltd. 2 (14) (1987) 541–590.
(2015) 3329–3339. [38] J.C. Guillemin, Organic selenocyanates: synthesis, characterization and uses in
[15] E. Sofic, K.W. Lange, K. Jellinger, P. Riederer, Reduced and oxidized glutathione in chemistry and biology, Curr. Org. Chem. 15 (2011) 1670–1687.
the substantia nigra of patients with Parkinson's disease, Neurosci. Lett. 142 (1992) [39] A.C. Feutz, D. Pham-Dinh, B. Allinquant, M. Miehe, M.S. Ghandour, An im-
128–130. mortalized jimpy oligodendrocyte cell line: defects in cell cycle and cAMP pathway,
[16] (a) A.K. Sharma, S. Amin, Post SELECT: selenium on trial, Future Med. Chem. 5 Glia 34 (2001) 241–252.
(2013) 163–174; [40] (a) S. Shaaban, B.F. Abdel-Wahab, Groebke-Blackburn-Bienayme multicomponent
(b) M. Nazıroğlu, M. Salina, P. Laszlo, Nanoparticles as potential clinical ther- reaction: emerging chemistry for drug discovery, Mol. Divers. 20 (1) (2016)
apeutic agents in Alzheimer’s disease: focus on selenium nanoparticles, Expert. Rev. 233–254;
Clin. Pharmacol. 10 (2017) 773–782; (b) M. Giustiniano, A. Basso, V. Mercalli, A. Massarotti, E. Novellino, G.C. Tron,

55
S. Shaaban et al. Bioorganic Chemistry 80 (2018) 43–56

J. Zhu, To each his own: isonitriles for all flavors. functionalized isocyanides as selenium supply, Carcinogenesis 33 (2012) 620–628;
valuable tools in organic synthesis, Chem. Soc. Rev. 46 (2017) 1295–1357. (b) S. Attacha, D. Solbach, K. Bela, A. Moseler, S. Wagner, M. Schwarzländer,
[41] K.P. Haval, S.B. Mhaske, N.P. Argade, Cyanuric chloride: decent dehydrating agent I. Aller, S.J. Müller, A.J. Meyer, Glutathione peroxidase-like enzymes cover five
for an exclusive and efficient synthesis of kinetically controlled isomaleimides, distinct cell compartments and membrane surfaces in arabidopsis thaliana, Plant.
Tetrahedron 62 (2006) 937–942. Cell. Environ. 40 (2017) 1281–1295.
[42] E.J. Alvarez-Manzaneda, R. Chahboun, E. Cabrera Torres, E. Alvarez, R. Alvarez- [62] M.J. Parnham, H. Sies, The early research and development of ebselen, Biochem.
Manzaneda, A. Haidour, J. Ramos, Triphenylphosphine–iodine: an efficient reagent Pharmacol. 86 (2013) 1248–1253.
for the regioselective dehydration of tertiary alcohols, Tetrahedron Lett. 45 (2004) [63] X. Huang, X. Liu, Q. Luo, J. Liu, J. Shen, Artificial selenoenzymes: designed and
4453–4455. redesigned, Chem. Soc. Rev. 40 (2011) 1171–1184.
[43] W. Hertler, E. Corey, Notes. A novel preparation of isonitriles, J. Org. Chem. 23 [64] L.A. Wessjohann, A. Schneider, M. Abbas, W. Brandt, Selenium in chemistry and
(1958) 1221–1222. biochemistry in comparison to sulfur, Biol. Chem. 388 (2007) 997–1006.
[44] A. Porcheddu, G. Giacomelli, M. Salaris, Microwave-assisted synthesis of isonitriles: [65] H. Safayhi, G. Tiegs, A. Wendel, A novel biologically active seleno-organic
a general simple methodology, J. Org. Chem. 70 (2005) 2361–2363. compound–V. Inhibition by ebselen (PZ 51) of rat peritoneal neutrophil lipox-
[45] S. Burckhardt, Methyl N-(Triethylammonium-Sulfonyl)Carbamate: “Burgess ygenase, Biochem. Pharmacol. 34 (1985) 2691–2694.
Reagent”, Synlett 2000 (2001) 559. [66] A.S. Hodage, P.P. Phadnis, A. Wadawale, K.I. Priyadarsini, V.K. Jain, Synthesis,
[46] A. Zarrouk, A. Vejux, T. Nury, H.I. El Hajj, M. Haddad, M. Cherkaoui-Malki, characterization and structures of 2-(3,5-dimethylpyrazol-1-Yl)Ethylseleno deriva-
J. Riedinger, M. Hammami, G. Lizard, Induction of mitochondrial changes asso- tives and their probable glutathione peroxidase (GPx) like activity, Org. Biomol.
ciated with oxidative stress on very long chain fatty acids (C22:0, C24:0, Or C26:0)- Chem. 9 (2011) 2992–2998.
treated human neuronal cells (SK-NB-E), Oxid. Med. Cell. Longev. 2012 (2012) [67] B.K. Sarma, D. Manna, M. Minoura, G. Mugesh, Synthesis, structure, spirocycliza-
623257. tion mechanism, and glutathione peroxidase-like antioxidant activity of stable
[47] M. Baarine, K. Ragot, E.C. Genin, H. El Hajj, D. Trompier, P. Andreoletti, spirodiazaselenurane and spirodiazatellurane, J. Am. Chem. Soc. 132 (2010)
M.S. Ghandour, F. Menetrier, M. Cherkaoui-Malki, S. Savary, G. Lizard, Peroxisomal 5364–5374.
and mitochondrial status of two murine oligodendrocytic cell lines (158N, 158JP): [68] F. Kumakura, B. Mishra, K.I. Priyadarsini, M. Iwaoka, A Water-soluble cyclic sele-
potential models for the study of peroxisomal disorders associated with dysmyeli- nide with enhanced glutathione peroxidase-like catalytic activities, Eur. J. Org.
nation processes, J. Neurochem. 111 (2009) 119–131. Chem. 2010 (2010) 440–445.
[48] C.I. Ghanem, M.J. Pérez, J.E. Manautou, A.D. Mottino, Acetaminophen from liver to [69] Q. Wang, K. Cui, O. Espin-Garcia, D. Cheng, X. Qiu, Z. Chen, M. Moore,
brain: new insights into drug pharmacological action and toxicity, Pharmacol. Res. R.G. Bristow, W. Xu, S. Der, G. Liu, Resistance to bleomycin in cancer cell lines is
109 (2016) 119–131. characterized by prolonged doubling time, reduced DNA damage and evasion of
[49] S. Shaaban, M.A. Arafat, W.S. Hamama, Vistas in the domain of organo seleno- G2/M arrest and apoptosis, PLoS One 8 (2013) e82363.
cyanates, ARKIVOC (2014) 470–505. [70] A. Mira, E.M. Gimenez, A.D. Bolzan, M.S. Bianchi, D.M. Lopez-Larraza, Effect of
[50] H.E. Gaffer, S. Shaaban, N.A. Abed, E. Abdel-latif, Synthesis of some new symme- thiol compounds on bleomycin-induced DNA and chromosome damage in human
trical diselenide dyestuffs for dyeing polyester fabrics, Pigm. Resin Technol. 45 (6) cells, Arch. Environ. Occup. Health. 68 (2013) 107–116.
(2016) 431–438. [71] H. Abramovič, B. Grobin, N.P. Ulrih, B. Cigić, The methodology applied in DPPH,
[51] F.H. Fry, C. Jacob, Sensor/effector drug design with potential relevance to cancer, ABTS and Folin-Ciocalteau assays has a large influence on the determined anti-
Curr. Pharm. Des. 12 (2006) 4479–4499. oxidant potential, Acta. Chim. Slov. 64 (2017) 2.
[52] C.A. Collins, F.H. Fry, A.L. Holme, A. Yiakouvaki, A. Al-Qenaei, C. Pourzand, [72] A. Daina, O. Michielin, V. Zoete, SwissADME: a free web tool to evaluate phar-
C. Jacob, Towards multifunctional antioxidants: synthesis, electrochemistry, in macokinetics, drug-likeness and medicinal chemistry friendliness of small mole-
vitro and cell culture evaluation of compounds with ligand/catalytic properties, cules, Sci. Rep. 3 (7) (2017) 42717.
Org. Biomol. Chem. 3 (2005) 1541–1546. [73] M. Lazard, M. Dauplais, S. Blanquet, P. Plateau, Recent advances in the mechanism
[53] F.H. Fry, A.L. Holme, N.M. Giles, G.I. Giles, C. Collins, K. Holt, S. Pariagh, of selenoamino acids toxicity in eukaryotic cells, Biomol. Concepts 24 (8) (2017)
T. Gelbrich, M.B. Hursthouse, N.J. Gutowski, C. Jacob, Multifunctional redox cat- 93–104 2.
alysts as selective enhancers of oxidative stress, Org. Biomol. Chem. 3 (2005) [74] R.M. Rosa, R. Roesler, A.L. Braga, J. Saffi, J.A. Henriques, Pharmacology and tox-
2579–2587. icology of diphenyl diselenide in several biological models, Braz. J. Med. Biol. Res.
[54] N.M. Giles, N.J. Gutowski, G.I. Giles, C. Jacob, Redox catalysts as sensitisers to- 40 (10) (2007) 1287–1304.
wards oxidative stress, FEBS Lett. 535 (2003) 179–182. [75] S.W. May, Selenium-based pharmacological agents: an update, Expert. Opin.
[55] (a) D.A. Bass, J.W. Parce, L.R. Dechatelet, P. Szejda, M.C. Seeds, M. Thomas, Flow Investig. Drugs. 11 (9) (2002) 1261–1269.
cytometric studies of oxidative product formation by neutrophils: a graded response [76] K.P. Bhabak, G. Mugesh, Functional mimics of glutathione peroxidase: bioinspired
to membrane stimulation, J. Immunol. 130 (1983) 1910–1917; synthetic antioxidants, Acc. Chem. Res 16 (43) (2010) 1408–1419 11.
(b) G. Rothe, G. Valet, Flow cytometric analysis of respiratory burst activity in [77] S.A. Hitchcock, L.D. Pennington, Structure−brain exposure relationships, J. Med.
phagocytes with hydroethidine and 2',7'-dichlorofluorescin, J. Leukoc. Biol. 47 Chem. 49 (26) (2006) 7559–7583.
(1990) 440–448; [78] P. Ertl, B. Rohde, P. Selzer, Fast calculation of molecular polar surface area as a sum
(c) K.J. Smith, R. Kapoor, P.A. Felts, Demyelination: the role of reactive oxygen of fragment-based contributions and its application to the prediction of drug
and nitrogen species, Brain. Pathol. 9 (1) (1999) 69–92; transport properties, J. Med. Chem. 43 (20) (2000) 3714–3717.
(d) S. Dikalov, K.K. Griendling, D.G. Harrison, Measurement of reactive oxygen [79] J. Fernandes, C.R. Gattass, Topological polar surface area defines substrate trans-
species in cardiovascular studies, Hypertension 49 (2007) 717–727; port by multidrug resistance associated protein 1 (MRP1/ABCC1), J. Med. Chem. 52
(e) X.L. Xu, Z. Li, Catalytic Electrophilic Alkylation of P-Quinones via a Redox (4) (2009) 1214–1218.
Chain Reaction, Angew. Chem. Int. Ed. 129 (2017) 8308–8312; [80] C.A. Lipinski, F. Lombardo, B.W. Dominy, P.J. Feeney, Experimental and compu-
(f) A.A. Ensafi, H.R. Jamei, E. Heydari-Bafrooei, B. Rezaei, Electrochemical study tational approaches to estimate solubility and permeability in drug discovery and
of quinone redox cycling: a novel application of DNA-based biosensors for mon- development settings, Adv. Drug. Deliv. Rev. 46 (1–3) (2001) 3–26.
itoring biochemical reactions, Bioelectrochemistry 31 (2016) 15–22. [81] E.C. Browne, S. Parakh, L.F. Duncan, S.J. Langford, J.D. Atkin, B.M. Abbott, Efficacy
[56] M. Karlsson, T. Kurz, U.T. Brunk, S.E. Nilsson, C.I. Frennesson, What does the of peptide nucleic acid and selected conjugates against specific cellular pathologies
commonly used DCF test for oxidative stress really show? Biochem. J. 428 (2010) of amyotrophic lateral sclerosis, Bioorganic. Med. Chem. 1 (2016) 1520–1527.
183–190. [82] V.A. Kachanov, Y.O. Slabko, V.O. Baranova, V.E. Shilova, A.V. Kaminskii,
[57] M. Baarine, P. Andreoletti, A. Athias, T. Nury, A. Zarrouk, K. Ragot, A. Vejux, Triselenium dicyanide from malononitrile and selenium dioxide. One-pot synthesis
J.M. Riedinger, Z. Kattan, G. Bessede, D. Trompier, S. Savary, M. Cherkaoui-Malki, of selenocyanates, Tetrahedron Lett. 45 (2004) 4461–4463.
G. Lizard, Evidence of oxidative stress in very long chain fatty acid-treated oligo- [83] D. Plano, Y. Baquedano, D. Moreno-Mateos, M. Font, A. Jiménez-Ruiz, J.A. Palop,
dendrocytes and potentialization of ROS production using RNA interference-di- C. Sanmartín, Selenocyanates and diselenides: a new class of potent antileishmanial
rected knockdown of ABCD1 and ACOX1 peroxisomal proteins, Neuroscience 213 agents, Eur. J. Med. Chem. 46 (8) (2011) 3315–3323.
(2012) 1–18. [84] S. Shaaban, M.A. Arafat, H.E. Gaffer, W.S. Hamama, Synthesis and anti-tumor
[58] A. Vejux, E. Kahn, D. Dumas, G. Bessede, F. Menetrier, A. Athias, J.M. Riedinger, evaluation of novel organoselenocyanates and symmetrical diselenides dyestuffs,
F. Frouin, J.F. Stoltz, E. Ogier-Denis, A. Todd-Pokropek, G. Lizard, 7- Der. Pharma. Chemica. 6 (2014) 186–193.
Ketocholesterol favors lipid accumulation and colocalizes with Nile Red positive [85] T. Nury, A. Zarrouk, A. Vejux, M. Doria, J.M. Riedinger, R. Delage-Mourroux,
cytoplasmic structures formed during 7-ketocholesterol-induced apoptosis: analysis G. Lizard, Induction of oxiapoptophagy, a mixed mode of cell death associated with
by flow cytometry, FRET biphoton spectral imaging microscopy, and subcellular oxidative stress, apoptosis and autophagy, on 7-ketocholesterol-treated 158N
fractionation, Cytometry A. 64 (2005) 87–100. murine oligodendrocytes: impairment by alpha-tocopherol, Biochem. Biophys. Res.
[59] E. Kahn, M. Baarine, S. Pelloux, J.M. Riedinger, F. Frouin, Y. Tourneur, G. Lizard, Commun. 446 (2014) 714–719.
Iron nanoparticles increase 7-ketocholesterol-induced cell death, inflammation, and [86] D.E. Paglia, W.N. Valentine, Studies on the quantitative and qualitative character-
oxidation on murine cardiac HL1-NB cells, Int. J. Nanomedicine 5 (2010) 185–195. ization of erythrocyte glutathione peroxidase, J. Lab. Clin. Med. 70 (1967)
[60] S. Shaaban, A. Negm, M.A. Sobh, L.A. Wessjohann, Expeditious entry to functio- 158–169.
nalized pseudo-peptidic organoselenide redox modulators via sequential Ugi/SN [87] Bruker Bruker AXS Inc. APEX2, Madison, Wisconsin, USA, 2012.
methodology, Anticancer Agents Med. Chem. 16 (2016) 621–632. [88] O.V. Dolomanov, L.J. Bourhis, R.J. Gildea, J.A.K. Howard, H. Puschmann, OLEX2: a
[61] (a) S. Krehl, M. Loewinger, S. Florian, A.P. Kipp, A. Banning, L.A. Wessjohann, complete structure solution, refinement and analysis program, J. Appl. Cryst. 42
M.N. Brauer, R. Iori, R.S. Esworthy, F. Chu, R. Brigelius-Flohé, Glutathione perox- (2009) 339–341.
idase-2 and selenium decreased inflammation and tumors in a mouse model of [89] G. Sheldrick, SHELXT - integrated space-group and crystal-structure determination,
inflammation-associated carcinogenesis whereas sulforaphane effects differed with Acta Cryst. 71 (2015) 3–8.

56

You might also like