Next Article in Journal
Plasmonic Titanium Nitride Tubes Decorated with Ru Nanoparticles as Photo-Thermal Catalyst for CO2 Methanation
Previous Article in Journal
Synthesis of Four Enantiomers of (1-Amino-3-Hydroxypropane-1,3-Diyl)Diphosphonic Acid as Diphosphonate Analogues of 4-Hydroxyglutamic Acid
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

An Insight into All Tested Small Molecules against Fusarium oxysporum f. sp. Albedinis: A Comparative Review

1
Laboratory of Inorganic Chemistry, Department of Chemistry, University of Helsinki, P.O. Box 55, FI-00014 Helsinki, Finland
2
Laboratoire de Bioressources, Biotechnologie, Ethnopharmacologie et Santé (LBBES), Department of Biology, Faculty of Sciences, University Mohamed Premier, Oujda 11022, Morocco
3
Chemistry Department, College of Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
4
Laboratory of Applied Chemistry and Environment (LCAE), Faculty of Sciences, University Mohammed Premier, Oujda 11022, Morocco
*
Authors to whom correspondence should be addressed.
Molecules 2022, 27(9), 2698; https://doi.org/10.3390/molecules27092698
Submission received: 6 February 2022 / Revised: 18 April 2022 / Accepted: 18 April 2022 / Published: 22 April 2022

Abstract

:
Bayoud disease affects date palms in North Africa and the Middle East, and many researchers have used various methods to fight it. One of those methods is the chemical use of synthetic compounds, which raises questions centred around the compounds and common features used to prepare targeted molecules. In this review, 100 compounds of tested small molecules, collected from 2002 to 2022 in Web of Sciences, were divided into ten different classes against the main cause of Bayoud disease pathogen Fusarium oxysporum f. sp. albedinis (F.o.a.) with structure–activity relationship (SAR) interpretations for pharmacophore site predictions as (δ···δ), where 12 compounds are the most efficient (one compound from each group). The compounds, i.e., (Z)-1-(1.5-Dimethyl-1H-pyrazole-3-yl)-3-hydroxy but-2-en-1-one 7, (Z)-3-(phenyl)-1-(1,5-dimethyl-1H-pyrazole-3-yl)-3-hydroxyprop-2-en-1-one 23, (Z)-1-(1,5-Dimethyl-1H-pyrazole-3-yl)-3-hydroxy-3-(pyridine-2-yl)prop-2-en-1-one 29, and 2,3-bis-[(2-hydroxy-2-phenyl)ethenyl]-6-nitro-quinoxaline 61, have antifungal pharmacophore sites (δ···δ) in common in N1---O4, whereas other compounds have only one δ pharmacophore site pushed by the donor effect of the substituents on the phenyl rings. This specificity interferes in the biological activity against F.o.a. Further understanding of mechanistic drug–target interactions on this subject is currently underway.

1. Introduction

Bayoud disease [1,2,3,4,5], caused by the telluric fungus pathogen Fusarium oxysporum f. sp. albedinis (F.o.a) [6,7,8,9], represents the leading dangerous agent of date palms cultivation, having killed more than 15 million Moroccan and Algerian date palm trees. Fungal infection causes significant implications, threatening date palms with high morbidity and mortality every year worldwide. Therefore, new antifungal inhibitors must be discovered urgently, especially those with new modes of action, low toxicity, and bioavailability, and are effective for responsive and drug-resistant fungi [10,11,12,13,14,15]. Due to their biological activity and chemical properties in recent years, fused heterocyclic compounds containing bridgehead nitrogen or oxygen donor atoms have drawn further interest. Indeed, several classes are reported in this review as pyrazole- and imidazole-based derivatives [16] presented in different biomolecules, such as histidine [17], histamine [18], and natural products [19]; this is an exciting building block [20]. Specifically, in recent decades, 4,5-diarylpyrazoles [21] and 2,5-diarylimidazoles [22] have gained interesting recognition as possible biomolecules in the field of drug development. Many biological and pharmacological properties are related to these structures [23]. βKeto-enol compounds [24,25,26,27] are found in many natural products as coumarin derivatives and play an important role in medicine and in the development of coordination chemistry as stable complexes. Imidazothiazole derivatives [28,29,30] are attractive nitrogen-containing heterocyclic ring-like histidine, biotin, nucleic acid, purine, etc., and have a broad spectrum of biological and pharmacological diverse activities.
Pyrazolic compounds [31] have established widespread potential biological activities, such as anti-inflammatory [32,33,34], antianxiety [35], antipyretic [36], antimicrobial [37,38,39,40], antiviral [41], antitumor [42,43,44], anticonvulsant [36,45,46,47], etc. Quinoxalines [48] are polyfunctionalized compounds with interesting biological activities, such as anti-human immunodeficiency virus (anti-HIV) and antidiabetic agents. Benzimidazole-1,2,3-triazole hybrid molecules [49] are hybrid compounds consisting of benzimidazole and 1,2,3-triazole, where both of them have a broad range of biological activities. N,N′-bipyrazole piperazine derivatives [50] are established as polypharmacological mixed ligands with several biological activities reported in the literature [51,52,53,54]. Meanwhile, Schiff base derivatives [53] have different biological functions, such as anti-inflammatory [55], antifungal [56], and antibacterial effects [57], and are commonly used as carriers of catalysts [58], optical chemical receptors [59], thermo-stable products [60], agents of metal complexion [61], inhibitors of corrosion [62], and stabilizers of polymers [63].

2. Pyrazole- and Imidazole-Based Derivatives

After some modifications, the agar diffusion approach is used for the antifungal analysis of pyrazole- and imidazole-based derivatives. In short, after isolation and preparation of the Fusarium fungus, the sterilized solution of the six compounds tested (16) in dimethyl sulfoxide (DMSO) is mixed with the potato dextrose agar (PDA) medium as an emulsifier at different concentrations using the method mentioned in the literature [16]. These compounds were synthesized by Takfaoui et al. using direct diarylation of pyrazoles and imidazoles with aryl halides, using palladium as the catalyst, DMAc as the solvent, and CsOAc as the base [64,65].
Using a non-linear regression algorithm curve of the concentration/percentage of inhibition, the half-maximal inhibitory concentration (IC50) was measured using Graphpad Prism software. DMSO-distilled water mixture was used as the negative control; no recognized antibiotic can specifically treat this infection.
The IC50 values are given in (Table 1) below. In the pyrazole derivatives, compound 4 (IC50 = 99.1 μg/mL) has the best fungus inhibition of all the tested compounds, where it contains p-C6H4 groups on the phenyl rings as an electron-donating character, and the high toxicity effect of the phenyl groups on the F.o.a. Furthermore, compound 1 (IC50 = 110.9 μg/mL), presenting m-CF3 groups on both phenyl rings, displays good activity close to that of compound 4. However, the following compound is from the imidazole series (compound 5) containing p-Cl groups on phenyl rings with an IC50 value equal to 114.7 μg/mL. The substitution of the phenyl rings by formyl (COH) groups (compound 6) is highly unfavorable for inhibitory potency [16].

3. β-Keto-enol Derivatives

a
β-Keto-enol Pyridine and Furan Derivatives
Using the agar diffusion process, we determined the in vitro antifungal ability of 11 compounds (717) against the pathogenic fungus (F.o.a). The synthetic route of the target compounds (717) was carried out following Claisen condensation under mild conditions [24,26,67,68,69,70,71,72,73,74]. Using the protocol described in the literature [27], the percentages of inhibition and semi-maximal inhibitory concentration (IC50) were measured and estimated using the inhibition percentage non-linear regression equation, while benomyl was used as a positive control (Table 2).
As presented in Table 2, the fungal activity of 7 is very substantial, though it decreases slightly in the case of 10 because of ethoxy phenyl groups, which commonly have pharmacophore sites (δ···δ+), as presented in Figure 1, due to their physicochemical properties and their ability to penetrate the envelope of fungal cells and enter their cellular place of action, thus displaying more excellent activity in [27].
b
(Z)-3(3-bromophenyl)-1-(1,5-dimethyl-1H-pyrazol-3yl)-3-hydroxyprop-2-en-1-one derivatives
The agar diffusion technique was tested for in vitro antifungal function (ADT), where the literature reported the protocol details [7]. The optical density values were measured for each culture at 625 nm, and the inhibition percentage (%) is expressed as (D0 − Dx)/D0 × 100. D0 is the diameter of the mycelial growth of the culture witness, and Dx is the diameter of the mycelial growth (Table 3). The target biomolecules 1823 based on βketo-enol and pyrazole entities and pyridine were prepared using a one-pot in situ condensation method, similar to the procedures in the literature [24].
As presented in Table 3, only compounds 22 and 23 reach values close to the standard (benomyl), as they belong to the same family. Such variations depend on the radical group attached to the fragment of pyrazole keto-enol, where compound 23 has a phenyl ring attached instead of the methyl group in compound 22. In addition, numerous molecular improvements are currently being made to these compounds as antifungal candidates [25].
c
β-Keto-enol pyrazolic compounds
The in vitro antifungal potential of ten prepared βKeto-enol pyrazolic compounds against the pathogen F.o.a was determined by the agar diffusion technique reported in the literature [26], and the half-maximal inhibitory concentration (IC50) was determined using a non-linear regression algorithm of the concentration-inhibition percentage graph, with benomyl used as a positive control. In addition, the target biomolecules 2430 based on βketo-enol and pyrazole entities were prepared by a one-pot in situ condensation method, which is similar to the procedures given in the literature [24].
On the other hand, most of these molecules demonstrate potent antifungal action against F.o.a, as seen in Table 4. These were based on the structure–activity relationships (S.A.R.s). Where a stimulating effect is exerted against F.o.a of the substitution pattern, we found compound 28 in the 3-thiophene group. In contrast, compound 30 with the 2-naphthalene group led the same inhibition percentage of 94% as the benomyl fungicide, while the best antifungal activity was found for compound 29 containing the 2-pyridine group IC50 of 60.84 μg/m. The existence of the R substituent should be further exploited [8] to evaluate the S.A.R.s for this novel class of antifungal agents.

4. Imidazothiazole Derivatives

The synthesis of various types of imidazothiazoles 3135 is potentially helpful for developing biologically active heterocycles. The synthetic methods are practical and straightforward and are conceivably applicable to analogous heterocyclic systems possessing nitrogen and sulfur [30,75,76,77,78,79,80,81,82]. The antifungal action of five imidazothiazole derivatives 3135 is carried out on an F.o.a using the concentrations C1, C2, C3, C4, and C5 as 5.0, 1.0, 0.2, 0.05, and 0.01 mg/mL, respectively. Each compound was prepared at various concentrations in the potato dextrose agar (PDA) before the fungus was cultured using the protocol described in the literature [28]. The IC50 was calculated using the linear regression equation between the normal logarithm concentrations and growth inhibition percentages.
From Table 5, the antifungal test of the five imidazothiazole derivatives tested against F.o.a. at five different concentrations acted differently, while all the molecules showed interesting results. Indeed, the best antifungal activity is found for compound 33 due to three methyl substituents on the ortho and para positions of the phenyl ring with IC50 not exceeding 20.00 μg/mL [28].

5. Pyrazolic Compounds

Monopyrazolic heterocyclic compounds 3655 were prepared in excellent yields by condensing one equivalent of hydroxymethylpyrazole with one equivalent of primary amines [83,84,85]. The antifungal behavior, as defined in the literature, was calculated by the agar diffusion technique [31], with the linear regression equation between the normal logarithm of the concentrations and the growth inhibition percentages calculated at the half-maximal inhibitory concentration (IC50).
The pyrazolic derivatives 50, 51, and 5355 were screened in vitro for their antifungal potential against F.o.a and collected in Table 6, where compounds 50 and 55 showed an excellent efficacy of IC50 = 86 μM and 168 μM, respectively, arguably due to the presence of the two phenyl moieties. Due to the (-Br) group, which is an essential source of electronegativity, compound 53 showed a moderate potential with an IC50 = 284 μM. The two other pyrazoles tested demonstrated low antifungal function [31].

6. Quinoxalines

A variety of 2,3-bifunctionalized quinoxalines (5661) have been prepared by the condensation of 1,6-disubstituted-hexan-1,3,4,6-tetraones with o-phenylenediamine, (R,R)-1,2-diaminocyclohexane, and p-nitro-o-phenylenediamine [86,87,88]. The antifungal activity of six prepared quinoxaline compounds’ antifungal activity was measured against F.o.a, as described in the method in the literature [48].
Based on Table 7, the most effective inhibitor is nitroquinoxaline 61, which produces 51% inhibition of the growth of Fusarium at a concentration of 72 mg/L due to its small nitro groups that disturb the cell membrane, with some intracellular target and electron-withdrawing solid group. At the same time, compounds 56, 60, and 59 are less effective but produce appreciable growth inhibition at comparable concentrations [48].

7. Benzimidazole-1,2,3-triazole Hybrid Molecules

A series of hybrid molecules 6269 was prepared by condensing 4-(trimethylsilylethynyl)benzaldehyde with substituted o-phenylenediamines. These, in turn, were reacted with 2-(azidomethoxy)ethyl acetate in a Cu alkyne–azide cycloaddition (CuAAC) to generate the 1,2,3-triazole pharmacophore under microwave assistance [89,90,91,92].
The eight new benzimidazole-1,2,3-triazole hybrid molecules were tested against F.o.a using the method described in the literature [49], and their linear growth and sporulation inhibitory rates are presented in Table 8.
Based on Table 8, all compounds were tested at a 20 mg/mL concentration, with Pelt, a systemic fungicide and benzimidazole precursor (70% of methyl thiophanate), as the positive control. Compound 66 shows a significantly increased rate with (17.01 and 30.62%) (p < 0.05) against F.o.a, which uniquely holds a CF3 group fixed to the benzimidazole core, a lipophilic group known to modulate absorption and metabolism, and may explain the enhanced activity [49].

8. N,N′-Bipyrazole Piperazine Derivatives

Novel bipyrazoles 7073 possessing piperazine or a mimed piperazine ring spacer were prepared in a one-step reaction in excellent yields. First, it condensed two hydroxymethylpyrazole derivatives with one equivalent of cyclic and acyclic piperazine [93,94,95,96].
As stated in the literature, in vitro antibacterial and antifungal activity is tested by the agar diffusion technique [50] using pathogenic strains of F.o.a. In contrast, streptomycin was used in the antibacterial assay as a reference compound for quality reasons. Therefore, the minimal concentration of inhibition (M.I.C.) is the lowest concentration of the tested compound that has inhibited the development of the micro-organism.
As presented in Table 9, four tested compounds showed differential anti-proliferative activity against F.o.a, as the best M.I.C. value was found for compound 71 of 5 μg/mL. These results are explained by the piperazine ring spacer and the carboxylate moiety at the three-position of the pyrazole rings that considerably increases the antifungal activity [50].

9. Bipyrazolic Tripodal Derivatives

A series of novel bipyrazolic tripodal derivatives 7481 was prepared in one step, with good and excellent yields. Then, one equivalent of the appropriate amine derivatives was added to a solution of two equivalents of the substituted hydroxymethylpyrazole in acetonitrile, and the mixture was continued under stirring at room temperature for 4–5 days. Finally, the crude material was evaporated, washed with water and CH2Cl2, and purified by silica gel column flash chromatography to give the target product 7481 [52].
The eight compounds containing bipyrazolic tripod derivatives are tested in vitro for their efficacy against Fusarium oxysporum f. Isolated from a date palm with vascular fusariosis, F.o.a was used as the protocol described in the literature [52]. The minimum inhibition concentration (M.I.C.) is the lowest dose of the compound that can inhibit micro-organism development.
From data in Table 10, the presence of the methyl as electron donor groups on the pyrazole rings increased the antifungal activity for compounds 74, 76, 78, and 80, but has a counter effect on the phenyl ring, e.g., in the case of compounds 80 and 81 which have M.I.C. values of 40 and 80 μg/mL, respectively. Additionally, nitro substituent as an electron-withdrawing group for compound 79 increased its effect compared with compound 77 [52].

10. Schiff Base Derivatives

Twelve new Schiff base derivatives are prepared using the condensation reaction of different amino-substituted compounds (such as aniline, pyridine-2-amine, o-toluidine, 2-nitrobenzenamine, 4-aminophenol, and 3-aminopropanol) and substituted aldehydes (such as nicotinaldehyde, o,m,p-nitrobenzaldehyde, and picolinaldehyde) in ethanol with acetic acid as a catalyst [53].
The agar diffusion technique against Fusarium oxysporum f evaluated the in vitro antifungal activities of all the new Schiff base derivative compounds, including F.o.a fungus, as described earlier [53]. In the presence of a concentration of the tested compound over the mycelium diameter of the reference culture multiplied by 100, it is found that the inhibition proportion of a molecule is proportional to the ratio of the mycelium diameter of the culture. Therefore, the minimal concentration of inhibition (M.I.C.) is the lowest dose of the compound, which inhibited the growth of the microorganism when the mixture (DMSO/EtOH + distilled water) is used as a negative control without any standard reference drug.
On the contrary, based on their M.I.C. values in Table 11, the in vitro antifungal assay findings showed that most of the screened ligands exhibited high to moderate activity against F.o.a. The maximum activity was 0.02 µg/mL, shown by compound 84, followed by compounds 87, 88, and 93 with M.I.C. values equal to 0.04 µg/mL, while compound 83 showed the most negligible M.I.C. value of 0.9 µg/mL. Other products also have numerous activities, with M.I.C.s varying from 0.08 µg/mL for compound 92 to 0.30 µg/mL for compound 86. Comparing both the structures of 83 and 84, it can be inferred that the presence at the ortho position of the phenyl ring of a strong electron-withdrawing group, such as nitro moiety (NO2), is very appropriate for increasing antifungal efficiency; the presence of an electron donation group, such as methyl moiety (CH3) for antifungal action, is unfavorable in the period. Unfortunately, though, the correct variables that influence the antifungal ability of these derivatives are difficult to ascertain with these early investigations. Further investigations using other models and techniques are essential for this [53].

11. Amino Acids Pyrazole Compounds

The functional pyrazolyl derivatives 94100 were prepared by condensing two equivalents of (3,5-dimethyl-1H-pyrazole-1-yl)methanol with one equivalent of amino acid ester hydrochloride derivatives (commercially available) in anhydrous solvents. All reactions were carried out at room temperature under stirring conditions for 4 to 6 days in an inert atmosphere [42,97,98,99,100,101,102,103,104,105,106].
The activities of the pyrazole compound amino acids and the agar techniques determined 94100. The yeast of the F.o.a was isolated from a date palm touched by the vascular Fusarium prepared in a PDA medium at 37 g/L [54].
Based on Table 12, compared to blank culture, the inhibition rates of F.o.a development ranged from 0 to 480 mg/L for ester hydrochloride amino acids or their tripodal pyrazolic homologs. Inhibition activity against the growth of F.o.a. was shown by the various compounds studied, except 94 and 95. However, the rate of this inhibition changes from one molecule to another. Compound 98 has the best antifungal activity due to methyl substituents as electron donor groups in methyl alaninate (alanine ester) as the amino acid; these products’ structural and electronic diversity affected their biological activities. Further developments on this subject are currently in progress in order to understand their mechanistic interactions [54].

12. Comparison Using Structure–Activity Relationship

To understand this structure–activity relationship and the modes of action of these new biologically active molecules, we can carry out a theoretical study with bioinformatics molecular modeling (DFT, Docking, and ADME-Tox studies) after studying the mechanism of the reaction using conceptual DFT [107,108]. As a result, we obtained various prospective targeted drugs as inhibitors for Bayoud disease (Figure 2).
As presented in Figure 2, compounds 7, 23, 29, and 61 have the antifungal pharmacophore sites (δ···δ) in common in N1---O4, whereas other compounds have only one δ pharmacophore site pushed by the donor effect of the substituents on the phenyl rings; this specificity interferes in the biological activity against F.o.a.

13. Conclusions

This review uses 100 compounds of tested small molecules divided into ten classes against Fusarium oxysporum f. sp. albedinis (F.o.a). First, compound 4 (IC50 = 99.1 μg/mL) has the best fungus inhibition over all the pyrazole and imidazole derivatives, containing electron-donating character as para phenyl substituents. Furthermore, it is displays high toxicity in the phenyl groups on the F.o.a. Second, from βketo-enol derivatives, compounds 7, 23, 29, and 61 have the antifungal pharmacophore sites (δ···δ) in common in N1---O4, whereas other compounds have only one δ pharmacophore site pushed by the donor effect of the substituents on the phenyl rings; this specificity interferes in the biological activity against F.o.a. Moreover, these products’ structural and electronic diversity can affect their biological activities. Further developments on this subject are currently in progress to better understand their mechanistic interactions.

Funding

The review was funded by ANPMA/CNRST/UMP 2020–2023 Project, and Taif University Researchers Supporting Project TURSP2020/91, Taif, Saudi Arabia.

Acknowledgments

The authors thank the (ANPMA/CNRST/UMP 2020–2023 Project: Formulations fongiques, insecticide ou acaricides d’huiles essentielles des plantes aromatiques et médicinales et de leurs extraits aqueux) for their support. M.H.A expresses thanks to the Researchers Supporting Project TURSP2020/91 of Taif University, Taif, Saudi Arabia.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ibrahim, E.B.; Mohamed, M.; Rafik, B. Bayoud disease of date palm in Algeria: History, epidemiology and integrated disease management. Afr. J. Biotechnol. 2015, 14, 542–550. [Google Scholar] [CrossRef] [Green Version]
  2. Bouissil, S.; Guérin, C.; Roche, J.; Dubessay, P.; Alaoui-Talibi, E.; Pierre, G.; Michaud, P.; Mouzeyar, S.; Delattre, C.; El Modafar, C. Induction of Defense Gene Expression and the Resistance of Date Palm to Fusarium oxysporum f. sp. Albedinis in Response to Alginate Extracted from Bifurcaria bifurcata. Mar. Drugs 2022, 20, 88. [Google Scholar] [PubMed]
  3. M’Hammed, E.; Fatiha, D.; Ayada, D.; Said, B.; Mohamed, K. Catalytic and synthesis of new compound based on geranium oil. Egyp. J. Chem. 2021, 64, 7341–7346. [Google Scholar] [CrossRef]
  4. Belaidi, H.; Toumi-Benali, F.; Benzohra, I.E. Biocontrol of bayoud disease (Fusarium oxysporum f. sp. albedinis) on deglet-nour variety of date palm (Phoenix dactylifera L.) in south western oases of Algeria. Agric. Sci. Digest 2021, 41, 450–454. [Google Scholar] [CrossRef]
  5. Komeil, D.A.; Abdalla, M.Y.; El-Bebany, A.F.; Basyony, A.B.A. Resistance marker detection in ten date palm cultivars to the wilt pathogen, Fusarium oxysporum. Asian J. Plant Sci. 2021, 20, 363–369. [Google Scholar] [CrossRef]
  6. Tantaoui, A.; Ouinten, M.; Geiger, J.P.; Fernandez, D. Characterization of a single clonal lineage of Fusarium oxysporum f. sp. albedinis causing Bayoud disease of date palm in Morocco. Phytopathology 1996, 86, 787–792. [Google Scholar] [CrossRef] [Green Version]
  7. Khalil, N.M.; Ali, H.M.; Ibrahim, A.E. Biochemical Activity of Propolis Alcoholic Extracts against Fusarium oxysporum hm89. Egyp. J. Bot. 2022, 62, 197–212. [Google Scholar] [CrossRef]
  8. Shalaby, M.G.; Al-Hossainy, A.F.; Abo-Zeid, A.M.; Mobark, H.; Mahmoud, Y.A.G. Synthesis, characterization, physicochemical properties, and in-vitro anti-bacterial evaluation for doped Fe-Fusarium. J. Mol. Str. 2022, 1259, 132643. [Google Scholar] [CrossRef]
  9. Hallasgo, A.M.; Hauser, C.; Steinkellner, S.; Hage-Ahmed, K. Single and coinoculation of Serendipita herbamans with arbuscular mycorrhizal fungi reduces Fusarium wilt in tomato and slows disease progression in the long-term. Biol. Control 2022, 168, 104876. [Google Scholar] [CrossRef]
  10. Sedra, M.; Lazrek, B.H. Fusarium oxysporum f. sp. Albedinis Toxin Characterization and Use for Selection of Resistant Date Palm to Bayoud Disease; Springer: Cham, Switzerland, 2011; pp. 253–270. [Google Scholar]
  11. Picot, S.; Beugnet, F.; Leboucher, G.; Bienvenu, A.-L. Drug resistant parasites and fungi from a one-health perspective: A global concern that needs transdisciplinary stewardship programs. One Health 2022, 14, 100368. [Google Scholar] [CrossRef]
  12. Bankaitis, V.A.; Tripathi, A.; Chen, X.-R.; Igumenova, T.I. New strategies for combating fungal infections: Inhibiting inositol lipid signaling by targeting Sec14 phosphatidylinositol transfer proteins. Adv. Biol. Reg. 2022, 84, 100891. [Google Scholar] [CrossRef] [PubMed]
  13. Alarjani, K.M.; Huessien, D.; Rasheed, R.A.; Kalaiyarasi, M. Green synthesis of silver nanoparticles by Pisum sativum L. (pea) pod against multidrug resistant foodborne pathogens. J. King Saud Univ. Sci. 2022, 34, 101897. [Google Scholar] [CrossRef]
  14. Green, R.M.; Bicker, K.L. Development of an Anti-Biofilm Screening Technique Leads to the Discovery of a Peptoid with Efficacy against Candida albicans. ACS Infect. Diseas. 2022, 8, 310–320. [Google Scholar] [CrossRef] [PubMed]
  15. Vanzolini, T.; Bruschi, M.; Rinaldi, A.C.; Magnani, M.; Fraternale, A. Multitalented Synthetic Antimicrobial Peptides and Their Antibacterial, Antifungal and Antiviral Mechanisms. Int. J. Mol. Sci. 2022, 23, 545. [Google Scholar] [CrossRef] [PubMed]
  16. Abrigach, F.; Rokni, Y.; Takfaoui, A.; Khoutoul, M.; Doucet, H.; Asehraou, A.; Touzani, R. In vitro screening, homology modeling and molecular docking studies of some pyrazole and imidazole derivatives. Biomed. Pharmacother. 2018, 103, 653–661. [Google Scholar] [CrossRef] [PubMed]
  17. Madany, N.M.K.; Shehata, M.R.; Mohamed, A.S. Ovothiol—A isolated from sea urchin eggs suppress oxidative stress, inflammation, and dyslipidemia resulted in restoration of liver activity in cholestatic rats. Biointerf. Res. Appl. Chem. 2022, 12, 8152–8162. [Google Scholar]
  18. Bulut, M.; Çelebi Sezer, Y.; Ceylan, M.M.; Alwazeer, D.; Koyuncu, M. Hydrogen-rich water can reduce the formation of biogenic amines in butter. Food Chem. 2022, 384, 132613. [Google Scholar] [CrossRef]
  19. Dantas, T.S.; de Oliveira, A.M.; Ferreira, M.R.A.; Soares, L.A.L. Therapeutic potential of croton blanchetianus for the treatment of gastric ulcers: A brief review. Biointerf. Res. Appl. Chem. 2022, 12, 8219–8230. [Google Scholar]
  20. Pakornchote, T.; Ektarawong, A.; Sukserm, A.; Pinsook, U.; Bovornratanaraks, T. Presence and absence of intrinsic magnetism in graphitic carbon nitrides designed through C–N–H building blocks. Sci. Rep. 2022, 12, 2343. [Google Scholar] [CrossRef]
  21. Teng, Q.-H.; Sun, G.-X.; Luo, S.-Y.; Wang, K.; Liang, F.-P. Design, syntheses and antitumor activities evaluation of 1,5-diaryl substituted pyrazole secnidazole ester derivatives. J. Heter. Chem. 2021, 58, 1656–1664. [Google Scholar] [CrossRef]
  22. Yang, Z.; Fang, Y.; Kim, J.-M.; Lee, K.-T.; Park, H. Synthesis of halogenated 1,5-diarylimidazoles and their inhibitory effects on lps-induced pge2 production in raw 264.7 cells. Molecules 2021, 26, 6093. [Google Scholar] [CrossRef] [PubMed]
  23. Mohamady, S.; Kralt, B.; Samwel, S.K.; Taylor, S.D. Efficient One-Pot, Two-Component Modular Synthesis of 3,5-Disubstituted Pyrazoles. ACS Omega 2018, 3, 15566–15574. [Google Scholar] [CrossRef] [PubMed]
  24. Radi, S.; Tighadouini, S.; Feron, O.; Riant, O.; Bouakka, M.; Benabbes, R.; Mabkhot, Y.N. Synthesis of Novel beta-Keto-Enol Derivatives Tethered Pyrazole, Pyridine and Furan as New Potential Antifungal and Anti-Breast Cancer Agents. Molecules 2015, 20, 20186–20194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Tighadouini, S.; Benabbes, R.; Tillard, M.; Eddike, D.; Haboubi, K.; Karrouchi, K.; Radi, S. Synthesis, crystal structure, DFT studies and biological activity of (Z)-3-(3-bromophenyl)-1-(1,5-dimethyl-1H-pyrazol-3-yl)-3-hydroxyprop-2-en-1-one. Chem. Cent. J. 2018, 12, 122. [Google Scholar] [CrossRef] [PubMed]
  26. Tighadouini, S.; Radi, S.; Abrigach, F.; Benabbes, R.; Eddike, D.; Tillard, M. Novel β-keto-enol Pyrazolic Compounds as Potent Antifungal Agents. Design, Synthesis, Crystal Structure, DFT, Homology Modeling, and Docking Studies. J. Chem. Inf. Model. 2019, 59, 1398–1409. [Google Scholar] [CrossRef]
  27. Tighadouini, S.; Radi, S.; Benabbes, R.; Youssoufi, M.H.; Shityakov, S.; El Massaoudi, M.; Garcia, Y. Synthesis, Biochemical Characterization, and Theoretical Studies of Novel beta-Keto-enol Pyridine and Furan Derivatives as Potent Antifungal Agents. ACS Omega 2020, 5, 17743–17752. [Google Scholar] [CrossRef]
  28. Koudad, M.; El Hamouti, C.; Elaatiaoui, A.; Dadou, S.; Oussaid, A.; Abrigach, F.; Pilet, G.; Benchat, N.; Allali, M. Synthesis, crystal structure, antimicrobial activity and docking studies of new imidazothiazole derivatives. J. Iran. Chem. Soc. 2019, 17, 297–306. [Google Scholar] [CrossRef]
  29. Kassab, R.M.; Gomha, S.M.; Muhammad, Z.A.; El-Khouly, A.S. Synthesis, biological profile, and molecular docking of some new bis-imidazole fused templates and investigation of their cytotoxic potential as anti-tubercular and/or anticancer prototypes. Med. Chem. 2021, 17, 875–886. [Google Scholar] [CrossRef]
  30. Haddad, Y.; Remes, M.; Adam, V.; Heger, Z. Toward structure-based drug design against the epidermal growth factor receptor (EGFR). Drug Discov. Today 2021, 26, 289–295. [Google Scholar] [CrossRef]
  31. Abrigach, F.; Karzazi, Y.; Benabbes, R.; El Youbi, M.; Khoutoul, M.; Taibi, N.; Karzazi, N.; Benchat, N.; Bouakka, M.; Saalaoui, E.; et al. Synthesis, biological screening, P.O.M., and 3D-QSAR analyses of some novel pyrazolic compounds. Med. Chem. Res. 2017, 26, 1784–1795. [Google Scholar] [CrossRef]
  32. Bandgar, B.P.; Gawande, S.S.; Bodade, R.G.; Gawande, N.M.; Khobragade, C.N. Synthesis and biological evaluation of a novel series of pyrazole chalcones as anti-inflammatory, antioxidant and antimicrobial agents. Bioorg. Med. Chem. 2009, 17, 8168–8173. [Google Scholar] [CrossRef] [PubMed]
  33. Alegaon, S.G.; Alagawadi, K.R.; Garg, M.K.; Dushyant, K.; Vinod, D. 1,3,4-Trisubstituted pyrazole analogues as promising anti-inflammatory agents. Bioorg. Chem. 2014, 54, 51–59. [Google Scholar] [CrossRef] [PubMed]
  34. Li, Y.R.; Li, C.; Liu, J.C.; Guo, M.; Zhang, T.Y.; Sun, L.P.; Zheng, C.J.; Piao, H.R. Synthesis and biological evaluation of 1,3-diaryl pyrazole derivatives as potential antibacterial and anti-inflammatory agents. Bioorg. Med. Chem. Lett. 2015, 25, 5052–5057. [Google Scholar] [CrossRef] [PubMed]
  35. Khan, M.F.; Alam, M.M.; Verma, G.; Akhtar, W.; Akhter, M.; Shaquiquzzaman, M. The therapeutic voyage of pyrazole and its analogs: A review. Eur. J. Med. Chem. 2016, 120, 170–201. [Google Scholar] [CrossRef]
  36. Alam, J.; Alam, O.; Alam, P.; Naim, M.J. A Review on Pyrazole chemical entity and Biological Activity. Inter. J. Pharm. Sci. Res. 2015, 6, 1433–1442. [Google Scholar]
  37. Chovatia, P.T.; Akabari, J.D.; Kachhadia, P.K.; Zalavadia, P.D.; Joshi, H.S. Synthesis and selective antitubercular and antimicrobial inhibitory activity of 1-acetyl-3,5-diphenyl-4,5-dihydro-(1h)-pyrazole derivatives. J. Serb. Chem. Soc. 2006, 71, 713–720. [Google Scholar] [CrossRef]
  38. Khanage, S.G.; Mohite, P.B.; Pandhare, R.B.; Raju, S.A. Microwave Assisted Synthesis of 1-[5-(Substituted Aryl)-1H-Pyrazol-3-yl]-3,5-Diphenyl-1H-1,2,4-Triazole as Antinociceptive and Antimicrobial Agents. Adv. Pharm. Bull. 2014, 4, 105–112. [Google Scholar]
  39. Hamada, N.M.M.; Abdo, N.Y.M. Synthesis, characterization, antimicrobial screening and free-radical scavenging activity of some novel substituted pyrazoles. Molecules 2015, 20, 10468–10486. [Google Scholar] [CrossRef] [Green Version]
  40. Elshaier, Y.A.; Barakat, A.; Al-Qahtany, B.M.; Al-Majid, A.M.; Al-Agamy, M.H. Synthesis of Pyrazole-Thiobarbituric Acid Derivatives: Antimicrobial Activity and Docking Studies. Molecules 2016, 21, 1337. [Google Scholar] [CrossRef] [Green Version]
  41. Kucukguzel, S.G.; Coskun, I.; Aydin, S.; Aktay, G.; Gursoy, S.; Cevik, O.; Ozakpinar, O.B.; Ozsavci, D.; Sener, A.; Kaushik-Basu, N.; et al. Synthesis and characterization of celecoxib derivatives as possible anti-inflammatory, analgesic, antioxidant, anticancer and anti-HCV agents. Molecules 2013, 18, 3595–3614. [Google Scholar] [CrossRef] [Green Version]
  42. El Kodadi, M.; Benamar, M.; Ibrahim, B.; Zyad, A.; Malek, F.; Touzani, R.; Ramdani, A.; Melhaoui, A. New synthesis of two tridentate bipyrazolic compounds and their cytotoxic activity tumor cell lines. Nat. Prod. Res. 2007, 21, 947–952. [Google Scholar] [CrossRef] [PubMed]
  43. Insuasty, B.; Tigreros, A.; Orozco, F.; Quiroga, J.; Abonía, R.; Nogueras, M.; Sanchez, A.; Cobo, J. Synthesis of novel pyrazolic analogues of chalcones and their 3-aryl-4-(3-aryl-4,5-dihydro-1H-pyrazol-5-yl)-1-phenyl-1H-pyrazole derivatives as potential antitumor agents. Bioorg. Med. Chem. 2010, 18, 4965–4974. [Google Scholar] [CrossRef] [PubMed]
  44. Yahyi, A.; Et-Touhami, A.; Yahyaoui, R.; Touzani, R. Synthesis, Characterization by Means of I.R., 1H, 13C-N.M.R. and Biological Investigations on New Diorganotin Carboxylic Acid Derivatives. Lett. Drug Des. Discov. 2010, 7, 534–540. [Google Scholar] [CrossRef]
  45. Ajay Kumar, K.; Jayaroopa, P. Pyrazoles: Synthetic strategies and their pharmaceutical applications-an overview. Int. J. Pharm. Tech. Res. 2013, 5, 1473–1486. [Google Scholar]
  46. Kucukguzel, S.G.; Senkardes, S. Recent advances in bioactive pyrazoles. Eur. J. Med. Chem. 2015, 97, 786–815. [Google Scholar] [CrossRef]
  47. Küçükgüzel, Ş.G.; Çıkla-Süzgün, P. Recent advances bioactive 1,2,4-triazole-3-thiones. Eur. J. Med. Chem. 2015, 97, 830–870. [Google Scholar] [CrossRef]
  48. Waring, M.J.; Ben-Hadda, T.; Kotchevar, A.T.; Ramdani, A.; Touzani, R.; El Kadiri, S.; Hakkou, A.; Bouakka, M.; Ellis, T. 2,3-Bifunctionalized quinoxalines: Synthesis, DNA interactions and evaluation of anticancer, anti-tuberculosis and antifungal activity. Molecules 2002, 7, 641–656. [Google Scholar] [CrossRef]
  49. Ouahrouch, A.; Ighachane, H.; Taourirte, M.; Engels, J.W.; Sedra, M.H.; Lazrek, H.B. Benzimidazole-1,2,3-triazole hybrid molecules: Synthesis and evaluation for antibacterial/antifungal activity. Arch. Pharm. 2014, 347, 748–755. [Google Scholar] [CrossRef] [Green Version]
  50. Radi, S.; Toubi, Y.; Hakkou, A.; Souna, F.; Himri, I.; Bouakka, M. Synthesis, Antibacterial and Antifungal Activities of Novel N,N’-bipyrazole Piperazine Derivatives. Lett. Drug Des. Discov. 2012, 9, 853–857. [Google Scholar] [CrossRef]
  51. Klenc, J.; Raux, E.; Barnes, S.; Sullivan, S.; Duszynska, B.; Bojarski, A.J.; Strekowski, L. Synthesis of 4-Substituted 2-(4-Methylpiperazino) pyrimidines and Quinazoline Analogs as Serotonin 5-HT 2A Receptor Ligands. J. Heter. Chem. 2009, 46, 1259–1265. [Google Scholar]
  52. Radi, S.; Toubi, Y.; Hamdani, I.; Hakkou, A.; Souna, F.; Himri, I.; Bouakka, M. Synthesis, antibacterial and antifungal activities of some new bipyrazolic tripodal derivatives. Res. J. Chem. Sci. 2012, 2, 40–44. [Google Scholar]
  53. Toubi, Y.; Abrigach, F.; Radi, S.; Souna, F.; Hakkou, A.; Alsayari, A.; Muhsinah, A.B.; Mabkhot, Y.N. Synthesis, antimicrobial screening, homology modeling, and molecular docking studies of a new series of Schiff base derivatives as prospective fungal inhibitor candidates. Molecules 2019, 24, 3250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Boussalah, N.; Touzani, R.; Souna, F.; Himri, I.; Bouakka, M.; Hakkou, A.; Ghalem, S.; El Kadiri, S. Antifungal activities of amino acid ester functional pyrazolyl compounds against Fusarium oxysporum f. sp. albedinis and Saccharomyces cerevisiae yeast. J. Saudi Chem. Soc. 2013, 17, 17–21. [Google Scholar] [CrossRef] [Green Version]
  55. Xing, A.; Zeng, D.; Chen, Z. Synthesis, crystal structure and antioxidant activity of butylphenol Schiff bases: Experimental and DFT study. J. Mol. Str. 2022, 1253, 132209. [Google Scholar] [CrossRef]
  56. Aljamali, N.M.; Jawad, S.F. Preparation, spectral characterization, thermal study, and antifungal assay of (Formazane-mefenamic acid)–derivatives. Egyp. J. Chem. 2022, 65, 449–457. [Google Scholar] [CrossRef]
  57. Jambulingam, M.; AnandaThangadurai, S.; Vijayabaskaran, M. Designing and Synthesis of Some Transition Metal Complexes Derived from Schiff Bases for Anti-Bacterial Activity. J. Med. Chem. Sci. 2022, 5, 10–18. [Google Scholar]
  58. Mokhtari, P.; Mohammadnezhad, G. Anti-cancer properties and catalytic oxidation of sulfides based on vanadium (V) complexes of unprotected sugar-based Schiff-base ligands. Polyhedron 2022, 215, 115655. [Google Scholar] [CrossRef]
  59. Murakami, M.; Kouyama, T. Crystal structure of squid rhodopsin. Nature 2008, 453, 363–367. [Google Scholar] [CrossRef]
  60. Saavedra, C.P.; Encinas, M.V.; Araya, M.A.; Pichuantes, S.E.; Vásquez, C.C. Biochemical characterization of a thermostable cysteine synthase from Geobacillus stearothermophilus V. Biochimie 2004, 86, 481–485. [Google Scholar] [CrossRef]
  61. Levitskiy, O.A.; Aglamazova, O.I.; Grishin, Y.K.; Nefedov, S.E.; Magdesieva, T.V. Corey-Chaykovsky cyclopropanation of dehydroalanine in the Ni(II) coordination environment: Electrochemical vs. chemical activation. Electrochim. Acta 2022, 409, 139980. [Google Scholar] [CrossRef]
  62. Shenoy, K.V.; Venugopal, P.P.; Reena Kumari, P.D.; Chakraborty, D. Anti-corrosion investigation of a new nitro veratraldehyde substituted imidazopyridine derivative Schiff base on mild steel surface in hydrochloric acid medium: Experimental, computational, surface morphological analysis. Mater. Chem. Phys. 2022, 281, 125855. [Google Scholar] [CrossRef]
  63. Hajri, A.K.; Jamoussi, B.; Albalawi, A.E.; Alhawiti, O.H.N.; Alsharif, A.A. Designing of modified ion-imprinted chitosan particles for selective removal of mercury (II) ions. Carbohydr. Polym. 2022, 286, 119207. [Google Scholar] [CrossRef] [PubMed]
  64. Takfaoui, A.; Zhao, L.; Touzani, R.; Soulé, J.-F.; Dixneuf, P.H.; Doucet, H. One pot Pd(OAc)2-catalysed 2,5-diarylation of imidazoles derivatives. Tetrahedron 2014, 70, 8316–8323. [Google Scholar] [CrossRef]
  65. Takfaoui, A.; Zhao, L.; Touzani, R.; Dixneuf, P.H.; Doucet, H. Palladium-catalysed direct diarylations of pyrazoles with aryl bromides: A one step access to 4,5-diarylpyrazoles. Tetrahedron Lett. 2014, 55, 1697–1701. [Google Scholar] [CrossRef]
  66. Wu, Z.B.; Park, H.Y.; Xie, D.W.; Yang, J.X.; Hou, S.T.; Shahzad, N.; Kim, C.K.; Yang, S. Synthesis, biological evaluation, and 3D-QSAR studies of N- (Substituted pyridine-4-yl)-1-(substituted phenyl)-5-trifluoromethyl- 1H- pyrazole-4-carboxamide derivatives as potential succinate dehydrogenase inhibitors. J. Agric. Food Chem. 2021, 69, 1214–1223. [Google Scholar] [CrossRef]
  67. Tighadouini, S.; Radi, S.; Garcia, Y. Selective chemical adsorption of Cd(II) on silica covalently decorated with a β-ketoenol-thiophene-furan receptor. Mol. Syst. Des. Eng. 2020, 5, 1037–1047. [Google Scholar] [CrossRef]
  68. Radi, S.; Tighadouini, S.; Bacquet, M.; Degoutin, S.; Dacquin, J.-P.; Eddike, D.; Tillard, M.; Mabkhot, Y.N. β-Keto-enol Tethered Pyridine and Thiophene: Synthesis, Crystal Structure Determination and Its Organic Immobilization on Silica for Efficient Solid-Liquid Extraction of Heavy Metals. Molecules 2016, 21, 888. [Google Scholar] [CrossRef] [Green Version]
  69. Radi, S.; Tighadouini, S.; Eddike, D.; Tillard, M.; Mabkhot, Y.N. Crystal Structure of (Z)-1-(1,5-dimethyl-1H-pyrazol-3-yl)-3-hydroxy-3-(4-methoxyphenyl)prop-2-en-1-one, C15H16N2O3. Z. Kristallogr. New Cryst. Struct. 2017, 232, 199–200. [Google Scholar] [CrossRef] [Green Version]
  70. Radi, S.; Tighadouini, S.; Eddike, D.; Tillard, M.; Mabkhot, Y.N. Crystal Structure of (Z)-1-(1,5-dimethyl-1H-pyrazol-3-yl)-3-hydroxy-3-phenylprop-2-en-1-one, C14H14N2O2. Z. Kristallogr. New Cryst. Struct. 2017, 232, 201–202. [Google Scholar]
  71. Radi, S.; Tighadouini, S.; Eddike, D.; Tillard, M.; Mabkhot, Y.N. Crystal Structure of (Z)-1-(1,5-dimethyl-1H-pyrazol-3-yl)-3-(4-ethoxyphenyl)-3-hydroxyprop-2-en-1-one, C16H18N2O3. Z. Kristallogr. New Cryst. Struct. 2017, 232, 207–208. [Google Scholar] [CrossRef] [Green Version]
  72. Radi, S.; Tighadouini, S.; Eddike, D.; Tillard, M.; Mabkhot, Y.N. Crystal Structure of (Z)-1-(1,5-dimethyl-1H-pyrazol-3-yl)-3-hydroxy-3-(p-toly)prop-2-en-1-one, C15H16N2O2. Z. Kristallogr. New Cryst. Struct. 2017, 232, 209–210. [Google Scholar] [CrossRef]
  73. Radi, S.; Tighadouini, S.; Eddike, D.; Tillard, M.; Mabkhot, Y.N. Crystal Structure of (Z)-3-hydroxy-3-(4-methoxyphenyl)-1-(pyridin-2-yl)prop-2-en-1-one, C15H13NO3. Z. Kristallogr. New Cryst. Struct. 2017, 232, 235–236. [Google Scholar] [CrossRef] [Green Version]
  74. Tighadouini, S.; Radi, S.; Ferbinteanu, M.; Garcia, Y. Highly Selective Removal of Pb(II) by a Pyridylpyrazole-β-ketoenol Receptor Covalently Bonded onto the Silica Surface. ACS Omega 2019, 4, 3954–3964. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Peng, Y.-H.; Liao, F.-Y.; Tseng, C.-T.; Kuppusamy, R.; Li, A.S.; Chen, C.H.; Fan, Y.S.; Wang, S.Y.; Wu, M.H.; Hsueh, C.C.; et al. Unique Sulfur-Aromatic Interactions Contribute to the Binding of Potent Imidazothiazole Indoleamine 2,3-Dioxygenase Inhibitors. J. Med. Chem. 2020, 63, 1642–1659. [Google Scholar] [CrossRef]
  76. Adole, V.A.; Jagdale, B.S.; Pawar, T.B.; Sagane, A.A. Ultrasound promoted stereoselective synthesis of 2,3-dihydrobenzofuran appended chalcones at ambient temperature. S. Afr. J. Chem. 2020, 73, 35–43. [Google Scholar] [CrossRef]
  77. Morigi, R.; Vitali, B.; Prata, C.; Palomino, R.A.N.; Graziadio, A.; Locatelli, A.; Rambald, M.; Leoni, A. Investigation on the effects of antimicrobial imidazo[2,1-b]Thiazole derivatives on the genitourinary microflora. Med. Chem. 2018, 14, 311–319. [Google Scholar] [CrossRef]
  78. Abdel-Wahab, B.F.; Khidre, R.E.; Awad, G.E.A. Design and Synthesis of Novel 6-(5-Methyl-1H-1,2,3-triazol-4-yl)-5-[(2-(thiazol-2-yl)hydrazono)methyl]imidazo[2,1-b]thiazoles as Antimicrobial Agents. J. Heter. Chem. 2017, 54, 489–494. [Google Scholar] [CrossRef]
  79. Jallapally, A.; Addla, D.; Yogeeswari, P.; Sriram, D.; Kantevari, S. 2-Butyl-4-chloroimidazole based substituted piperazine-thiosemicarbazone hybrids as potent inhibitors of Mycobacterium tuberculosis. Bioorg. Med. Chem. Let. 2014, 24, 5520–5524. [Google Scholar] [CrossRef]
  80. Rani, N.; Sharma, A.; Gupta, G.K.; Singh, R. Imidazoles as potential antifungal agents: A review. Med. Chem. 2013, 13, 1626–1655. [Google Scholar] [CrossRef]
  81. Mohamed, H.A.; Abdel-Wahab, B.F. Synthetic access to imidazo[2,1-b]thiazoles. J. Sulf. Chem. 2012, 33, 589–604. [Google Scholar] [CrossRef]
  82. Abdel-Wahab, B.F.; Mohamed, H.A. Imidazobenzothiazoles: Synthesis and application. J. Sulf. Chem. 2012, 33, 335–349. [Google Scholar] [CrossRef]
  83. Touzani, R.; Ramdani, A.; Ben-Hadda, T.; El Kadiri, S.; Maury, O.; Le Bozec, H.; Dixneuf, P.H. Efficient synthesis of new nitrogen donor containing tripods under microwave irradiation and without solvent. Synth. Commun. 2001, 31, 1315–1321. [Google Scholar] [CrossRef]
  84. Lamsayah, M.; Khoutoul, M.; Abrigach, F.; Oussaid, A.; Touzani, R. Selective liquid-liquid extraction of fe(II) and cd(II) using n,n’-pyrazole bidentate ligands with theoretical study investigations. Separ. Sci. Tech. 2015, 50, 2170–2176. [Google Scholar]
  85. Khoutoul, M.; Abrigach, F.; Zarrouk, A.; Benchat, N.-E.; Lamsayah, M.; Touzani, R. New nitrogen-donnor pyrazole ligands for excellent liquid-liquid extraction of Fe2+ ions from aqueous solution, with theoretical study. Res. Chem. Interm. 2015, 41, 3319–3334. [Google Scholar] [CrossRef]
  86. Touzani, R.; Ben-Hadda, T.; El Kadiri, S.; Ramdani, A.; Maury, O.; Le Bozec, H.; Toupet, L.; Dixneuf, P.H. Solution, solid state structure and fluorescence studies of 2,3-functiolized quinoxalines: Evidebce for π-delocalized keto-enamine form with N-H..O intramolecular hydrogen bonds. New J. Chem. 2001, 25, 391–395. [Google Scholar] [CrossRef]
  87. Daoud, A.; Cheknane, A.; Touzani, R.; Hilal, H.S.; Boulouiz, A. Simulation of the Electrochemical Properties of Dye-Sensitized Solar Cells Based on Quinoxaline Dyes: Effects of Hydroxyl Group Numbers and Positions. J. Electr. Mater. 2021, 50, 5656–5663. [Google Scholar] [CrossRef]
  88. Bouabdallah, I.; Zidane, I.; Touzani, R.; Hacht, B.; Ramdani, A. Quinoxalines and tetraketones for metal cations extraction. Arkivoc 2006, 10, 77–81. [Google Scholar] [CrossRef] [Green Version]
  89. Tornøe, C.W.; Christensen, C.; Meldal, M. Peptidotriazoles on Solid Phase:  [1,2,3]-Triazoles by Regiospecific Copper(I)-Catalyzed 1,3-Dipolar Cycloadditions of Terminal Alkynes to Azides. J. Org. Chem. 2002, 67, 3057–3064. [Google Scholar] [CrossRef]
  90. Rostovtsev, V.V.; Green, L.G.; Fokin, V.V.; Sharpless, K.B. A stepwise huisgen cycloaddition process: Copper(I)-catalyzed regioselective “ligation” of azides and terminal alkynes. Angew. Chem. Int. Ed. 2002, 41, 2565–2599. [Google Scholar] [CrossRef]
  91. Meldal, M.; Tornøe, C.W. Cu-Catalyzed Azide–Alkyne Cycloaddition. Chem. Rev. 2008, 108, 2952–3015. [Google Scholar] [CrossRef]
  92. Pradere, U.; Roy, V.; Mc Brayer, T.R.; Schinazi, R.F.; Agrofoglio, L.A. Preparation of ribavirin analogues by copper- and ruthenium-catalyzed azide-alkyne 1,3-dipolar cycloaddition. Tetrahedron 2008, 64, 9044–9051. [Google Scholar] [CrossRef] [PubMed]
  93. Al-Ghorbani, M.; Gouda, M.A.; Baashen, M.; Alharbi, O.; Almalki, F.A.; Ranganatha, L.V. Piperazine Heterocycles as Potential Anticancer Agents: A Review. Pharm. Chem. J. 2022. [Google Scholar] [CrossRef]
  94. Sanka, B.M.; Tadesse, D.M.; Bedada, E.T.; Mengesha, E.T.; Babu, G.N. Design, synthesis, biological screening and molecular docking studies of novel multifunctional 1,4-di (aryl/heteroaryl) substituted piperazine derivatives as potential antitubercular and antimicrobial agents. Bioorg. Chem. 2021, 119, 105568. [Google Scholar] [CrossRef] [PubMed]
  95. Desai, N.C.; Rupala, Y.M.; Khasiya, A.G.; Shah, K.N.; Pandit, U.P.; Khedkar, V.M. Synthesis, biological evaluation, and molecular docking study of thiophene-, piperazine-, and thiazolidinone-based hybrids as potential antimicrobial agents. J. Heter. Chem. 2021, 59, 75–87. [Google Scholar] [CrossRef]
  96. Harit, T.; Malek, F.; Ameduri, B. Fluorinated polymers based on pyrazole groups for fuel cell membranes. Eur. Polym. J. 2016, 79, 72–81. [Google Scholar] [CrossRef]
  97. Garbacia, S.; Hillairet, C.; Touzani, R.; Lavastre, O. New nitrogen-rich tripodal molecules based on bis(pyrazol-1-ylmethyl)amines with substituents modulating steric hindrances and electron density of donor sites. Collect. Czech. Chem. Commun. 2005, 70, 34–40. [Google Scholar] [CrossRef]
  98. Roh, S.-G.; Park, Y.-C.; Park, D.-K.; Kim, T.-J.; Jeong, J.H. Synthesis and characterization of a Zn(II) complex of a pyrazole-based ligand bearing a chiral L-alaninemethylester. Polyhedron 2001, 20, 1961–1965. [Google Scholar] [CrossRef]
  99. Scarpellini, M.; Wu, A.J.; Kampk, J.W.; Pecoraro, V.L. Corroborative models of the cobalt(II) inhibited Fe/Mn superoxide dismutases. Inorg. Chem. 2005, 44, 5001–5010. [Google Scholar] [CrossRef]
  100. Bouabdallah, I.; Touzani, R.; Zidane, I.; Ramdani, A. Synthesis of new tripodal ligand: N,N-bis[(1,5-dimethylpyrazol-3-yl)methyl]benzylamine. Catecholase activity of two series of tripodal ligands with some copper (II) salts. Catal. Commun. 2007, 8, 707–712. [Google Scholar] [CrossRef]
  101. Touzani, R.; Garbacia, S.; Lavastre, O.; Yadav, V.K.; Carboni, B. Efficient solution phase combinatorial access to a library of pyrazole- and triazole-containing compounds. J. Comb. Chem. 2003, 5, 375–378. [Google Scholar] [CrossRef]
  102. Touzani, R.; Vasapollo, G.; Scorrano, S.; Del Sole, R.; Manera, M.G.; Rella, R.; El Kadiri, S. New complexes based on tridentate bispyrazole ligand for optical gas sensing. Mater. Chem. Phys. 2011, 126, 375–380. [Google Scholar] [CrossRef]
  103. Boussalah, N.; Touzani, R.; Bouabdallah, I.; El Kadiri, S.; Ghalem, S. Oxidation catalytic properties of new amino acid based on pyrazole tripodal ligands. Int. J. Acad. Res. 2009, 1, 137–143. [Google Scholar]
  104. Boussalah, N.; Touzani, R.; Bouabdallah, I.; El Kadiri, S.; Ghalem, S. Synthesis, structure and catalytic properties of tripodal amino-acid derivatized pyrazole-based ligands. J. Mol. Catal. A Chem. 2009, 306, 113–117. [Google Scholar] [CrossRef]
  105. Spadoni, G.; Balsamini, C.; Bedini, A.; Duranti, E.; Tontini, A. Short synthesis of tryptophane and β-carboline derivatives by reaction of indoles with N-(diphenylmethylene)-α,β-didehydroamino acid esters. J. Heterocycl. Chem. 1992, 29, 305–309. [Google Scholar] [CrossRef]
  106. Zumbuehl, A.; Stano, P.; Sohrmann, M.; Dietiker, R.; Peter, M.; Carreira, E.M. Synthesis and investigation of tryptophan—Amphotericin B conjugates. ChemBioChem 2009, 10, 1617–1620. [Google Scholar] [CrossRef]
  107. Qian, A.; Zheng, Y.; Wang, R.; Wei, J.; Cui, Y.; Cao, X.; Yang, Y. Design, synthesis, and structure-activity relationship studies of novel tetrazole antifungal agents with potent activity, broad antifungal spectrum and high selectivity. Bioorg. Med. Chem. Lett. 2017, 28, 344–350. [Google Scholar] [CrossRef]
  108. Xiao, Z.P.; Ma, T.W.; Liao, M.L.; Feng, Y.T.; Peng, X.C.; Li, J.L.; Li, Z.P.; Wu, Y.; Luo, Q.; Deng, Y.; et al. Tyrosyl-tRNA synthetase inhibitors as antibacterial agents: Synthesis, molecular docking and structure-activity relationship analysis of 3-aryl-4-arylaminofuran-2(5H)-ones. Eur. J. Med. Chem. 2011, 46, 4904–4914. [Google Scholar] [CrossRef]
Figure 1. Antibacterial and antifungal pharmacophore sites for compound 7.
Figure 1. Antibacterial and antifungal pharmacophore sites for compound 7.
Molecules 27 02698 g001
Figure 2. Chemical structure of the best active compounds from the group.
Figure 2. Chemical structure of the best active compounds from the group.
Molecules 27 02698 g002
Table 1. IC50 values of the tested pyrazole- and imidazole-based derivatives tested against F.o.a.
Table 1. IC50 values of the tested pyrazole- and imidazole-based derivatives tested against F.o.a.
ID.StructureIC50
μg/mLμM
1 Molecules 27 02698 i001110.9299.4
2 Molecules 27 02698 i002153.2538.8
3 Molecules 27 02698 i003165.1509.1
4 Molecules 27 02698 i00499.1256.4
5 Molecules 27 02698 i005114.7378.3
6 Molecules 27 02698 i006194.5667.1
Compared with literary works, we found that the pyrazole skeleton and its derivatives exhibited excellent inhibitory activity against Fusarium oxysporum [66].
Table 2. IC50 values of the tested βketo-enol pyridine and furan derivatives against F.o.a.
Table 2. IC50 values of the tested βketo-enol pyridine and furan derivatives against F.o.a.
IDStructureIC50
μg/mLμM
7 Molecules 27 02698 i00712.83
8 Molecules 27 02698 i008NSNS
9 Molecules 27 02698 i009NSNS
10 Molecules 27 02698 i01017
11 Molecules 27 02698 i01136
12 Molecules 27 02698 i012--
13 Molecules 27 02698 i013--
14 Molecules 27 02698 i014--
15 Molecules 27 02698 i015--
16 Molecules 27 02698 i016--
17 Molecules 27 02698 i017--
Table 3. Volume is withdrawn, a diameter of the strain and inhibition percentages of the tested (Z)-3(3-bromophenyl)-1-(1,5-dimethyl-1H-pyrazole-3yl)-3-hydroxyprop-2-en-1-one derivatives 1823 against F.o.a.
Table 3. Volume is withdrawn, a diameter of the strain and inhibition percentages of the tested (Z)-3(3-bromophenyl)-1-(1,5-dimethyl-1H-pyrazole-3yl)-3-hydroxyprop-2-en-1-one derivatives 1823 against F.o.a.
IDStructureVolume Is Withdrawn (μL)Diameter of the Strain in the Presence of the Drug (cm)Inhibition (%)
18 Molecules 27 02698 i01850
200
500
5.0
3.8
2.7
0
24
46
19 Molecules 27 02698 i01950
200
500
5.0
3.5
2.3
0
30
54
20 Molecules 27 02698 i02050
200
500
5.0
3.6
2.5
0
28
50
21 Molecules 27 02698 i02150
200
500
5.0
3.8
3.2
0
24
36
22 Molecules 27 02698 i02250
200
500
1.2
0.9
0.5
76
82
90
23 Molecules 27 02698 i02350
200
500
2.0
1.3
0.2
60
74
96
Benomyl Molecules 27 02698 i02450
200
500
2.3
1.1
0.3
54
78
94
Table 4. IC50 values of the tested βketo-enol pyrazolic derivatives against F.o.a.
Table 4. IC50 values of the tested βketo-enol pyrazolic derivatives against F.o.a.
IDStructureIC50
μg/mLμM
24 Molecules 27 02698 i025--
25 Molecules 27 02698 i026260.7471
26 Molecules 27 02698 i027--
27 Molecules 27 02698 i028--
28 Molecules 27 02698 i029193.3148.00
29 Molecules 27 02698 i03060.8414.80
30 Molecules 27 02698 i031181.3053.00
Table 5. IC50 values of the tested imidazothiazole derivatives against F.o.a.
Table 5. IC50 values of the tested imidazothiazole derivatives against F.o.a.
IDStructureIC50 (μg/mL)
31 Molecules 27 02698 i03250.00
32 Molecules 27 02698 i03370.00
33 Molecules 27 02698 i03420.00
34 Molecules 27 02698 i03560.00
35 Molecules 27 02698 i03650.00
Table 6. IC50 values of the tested pyrazolic compounds against F.o.a.
Table 6. IC50 values of the tested pyrazolic compounds against F.o.a.
IDStructureIC50 (μM)
36 Molecules 27 02698 i038-
37 Molecules 27 02698 i039751
38 Molecules 27 02698 i0402507
39 Molecules 27 02698 i042406
40 Molecules 27 02698 i043398
41 Molecules 27 02698 i045333
42 Molecules 27 02698 i0462755
43 Molecules 27 02698 i0482550
44 Molecules 27 02698 i0492486
45 Molecules 27 02698 i0502614
46 Molecules 27 02698 i0511223
47 Molecules 27 02698 i052697
48 Molecules 27 02698 i0532856
49 Molecules 27 02698 i0542322
50 Molecules 27 02698 i05586
51 Molecules 27 02698 i057662
52 Molecules 27 02698 i0582592
53 Molecules 27 02698 i060284
54 Molecules 27 02698 i061-
55 Molecules 27 02698 i063168
Table 7. Percent growth inhibition at different concentrations for quinoxaline compounds tested against F.o.a.
Table 7. Percent growth inhibition at different concentrations for quinoxaline compounds tested against F.o.a.
IDStructurePercent Growth Inhibition (Concentration, mg/L)
C1C2C3
56 Molecules 27 02698 i0649 (20)7 (40)22 (80)
57 Molecules 27 02698 i0669 (60)15 (120)15 (180)
58 Molecules 27 02698 i06717 (60)17 (120)19 (180)
59 Molecules 27 02698 i069213235 (180)
60 Molecules 27 02698 i07015 (34)31 (67)33 (134)
61 Molecules 27 02698 i07229 (18)31 (36)51 (72)
Table 8. Linear growth and inhibitory sporulation rates of benzimidazole-1,2,3-triazole hybrid molecules tested against F.o.a.
Table 8. Linear growth and inhibitory sporulation rates of benzimidazole-1,2,3-triazole hybrid molecules tested against F.o.a.
IDStructureLinear Growth-Inhibitory Rates (%)Sporulation Inhibitory Rates (%)
62 Molecules 27 02698 i0733.02 ± 0.96−5.85 ± 0.04
63 Molecules 27 02698 i075−1.59 ± 0.0516.36 ± 0.2
64 Molecules 27 02698 i0762.7 ± 0.16−34.79 ± 0.72
65 Molecules 27 02698 i078−0.16 ± 0.0221.94 ± 0.26
66 Molecules 27 02698 i07917.01 ± 0.9630.62 ± 0.5
67 Molecules 27 02698 i0812.3 ± 0.29−77.59 ± 2.64
68 Molecules 27 02698 i082−1.41 ± 0.3−61.05 ± 1.34
69 Molecules 27 02698 i084−14 ± 0.05−48.72 ± 2.35
Table 9. M.I.C. values of N,N′-bipyrazole piperazine derivatives tested against F.o.a.
Table 9. M.I.C. values of N,N′-bipyrazole piperazine derivatives tested against F.o.a.
IDStructureM.I.C.
μg/mLμM
70 Molecules 27 02698 i0851033.06
71 Molecules 27 02698 i087511.94
72 Molecules 27 02698 i0881032.85
73 Molecules 27 02698 i0902047.56
Table 10. M.I.C. values of bipyrazolic tripodal compounds tested against F.o.a.
Table 10. M.I.C. values of bipyrazolic tripodal compounds tested against F.o.a.
IDStructureM.I.C.
μg/mLμM
74 Molecules 27 02698 i0912.58.05
75 Molecules 27 02698 i093511.73
76 Molecules 27 02698 i0942.58.08
77 Molecules 27 02698 i0964094.7
78 Molecules 27 02698 i0972.57.05
79 Molecules 27 02698 i099510.63
80 Molecules 27 02698 i10040123.84
81 Molecules 27 02698 i10280182.14
Table 11. M.I.C. values of Schiff base derivatives compounds tested against F.o.a.
Table 11. M.I.C. values of Schiff base derivatives compounds tested against F.o.a.
IDStructureMIC (μg/mL)
82 Molecules 27 02698 i1030.10
83 Molecules 27 02698 i1050.90
84 Molecules 27 02698 i1060.02
85 Molecules 27 02698 i1080.25
86 Molecules 27 02698 i1090.30
87 Molecules 27 02698 i1110.04
88 Molecules 27 02698 i1120.04
89 Molecules 27 02698 i1140.12
90 Molecules 27 02698 i1150.25
91 Molecules 27 02698 i1160.20
92 Molecules 27 02698 i1170.08
93 Molecules 27 02698 i1180.04
Table 12. MIC values of amino acids pyrazole compound tested against F.o.a.
Table 12. MIC values of amino acids pyrazole compound tested against F.o.a.
IDStructureMIC (mg/L)
94 Molecules 27 02698 i119-
95 Molecules 27 02698 i120-
96 Molecules 27 02698 i12117
97 Molecules 27 02698 i12315
98 Molecules 27 02698 i1240.3
99 Molecules 27 02698 i12610
100 Molecules 27 02698 i1270.5
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kaddouri, Y.; Benabbes, R.; Ouahhoud, S.; Abdellattif, M.; Hammouti, B.; Touzani, R. An Insight into All Tested Small Molecules against Fusarium oxysporum f. sp. Albedinis: A Comparative Review. Molecules 2022, 27, 2698. https://doi.org/10.3390/molecules27092698

AMA Style

Kaddouri Y, Benabbes R, Ouahhoud S, Abdellattif M, Hammouti B, Touzani R. An Insight into All Tested Small Molecules against Fusarium oxysporum f. sp. Albedinis: A Comparative Review. Molecules. 2022; 27(9):2698. https://doi.org/10.3390/molecules27092698

Chicago/Turabian Style

Kaddouri, Yassine, Redouane Benabbes, Sabir Ouahhoud, Magda Abdellattif, Belkheir Hammouti, and Rachid Touzani. 2022. "An Insight into All Tested Small Molecules against Fusarium oxysporum f. sp. Albedinis: A Comparative Review" Molecules 27, no. 9: 2698. https://doi.org/10.3390/molecules27092698

Article Metrics

Back to TopTop