Next Article in Journal
Prediction of pH Value of Aqueous Acidic and Basic Deep Eutectic Solvent Using COSMO-RS σ Profiles’ Molecular Descriptors
Next Article in Special Issue
Antileishmanial Anthracene Endoperoxides: Efficacy In Vitro, Mechanisms and Structure-Activity Relationships
Previous Article in Journal
Performance Improvement of NIR Spectral Pattern Recognition from Three Compensation Models’ Voting and Multi-Modal Fusion
Previous Article in Special Issue
Structure-Based Bioisosterism Design, Synthesis, Biological Evaluation and In Silico Studies of Benzamide Analogs as Potential Anthelmintics
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Evaluation of Anthelmintic and Anti-Inflammatory Activity of 1,2,4-Triazole Derivatives

by
Renata Paprocka
1,*,†,
Przemysław Kołodziej
2,*,†,
Małgorzata Wiese-Szadkowska
3,†,
Anna Helmin-Basa
3 and
Anna Bogucka-Kocka
2
1
Department of Organic Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Jurasza Str. 2, 85-089 Bydgoszcz, Poland
2
Chair and Department of Biology and Genetics, Faculty of Pharmacy, Medical University in Lublin, Chodźki Str. 4A, 20-093 Lublin, Poland
3
Department of Immunology, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, M. Curie-Sklodowska Str. 9, 85-094 Bydgoszcz, Poland
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Molecules 2022, 27(14), 4488; https://doi.org/10.3390/molecules27144488
Submission received: 10 June 2022 / Revised: 8 July 2022 / Accepted: 11 July 2022 / Published: 13 July 2022
(This article belongs to the Special Issue Chemistry of Antiparasitic Drugs)

Abstract

:
Parasitic diseases, caused by intestinal helminths, remain a very serious problem in both human and veterinary medicine. While searching for new nematicides we examined a series of 1,2,4-triazole derivatives 922, obtained during reactions of N3-substituted amidrazones with itaconic anhydride. Two groups of compounds, 916 and 1722, differed in the position of the double bond on the methacrylic acid moiety. The toxicity of derivatives 922 and the anti-inflammatory activity of 12 and 1922 were studied on peripheral blood mononuclear cells (PBMC). Antiproliferative activity of compounds 12 and 1922 was tested cytometrically in PBMC cultures stimulated by phytohemagglutinin. The influence of derivatives 12 and 1922 on the TNF-α, IL-6, IL-10 and IFN-γ production was determined by ELISA in lipopolysaccharide-stimulated PBMC cultures. Anthelmintic activity of compounds 1022 was studied in the Rhabditis sp. nematodes model. Most compounds (1122) proved to be non-toxic to human PBMC. Derivatives 1922 showed anti-inflammatory activity by inhibiting the proliferation of lymphocytes. Moreover, compounds 12 and 1922 significantly reduced the production of TNF-α and derivatives 1921 decreased the level of INF-γ. The strongest anti-inflammatory activity was observed for compound 21. Compounds 12 and 14 demonstrated anthelmintic activity higher than albendazole and may become promising candidates for anthelmintic drugs.

1. Introduction

Parasitic diseases, including intestinal parasitic infections, are caused by various species of parasites, and constitute a serious health and socioeconomic problem. Globally, it is estimated that infections caused by intestinal parasites affect 3.5 billion people, 450 million of whom are symptomatic and have health problems as a direct result of the disease. Nematodes are largely responsible for these diseases, including Ascaris lumbricoides, Trichiuris trichiuria, Ancylostoma duodenale and Strongyloides stercoralis [1,2,3,4,5,6]. Diseases caused by intestinal nematodes can cause, among others, abdominal pain, dehydration, weight loss, anemia, intestinal obstruction, disorders of mental and physical development, malabsorption, inflammation and in severe cases, if left untreated, these can even lead to death [6,7,8].
Currently, a narrow group of drugs is used in the treatment of parasitic diseases caused by helminths, mainly albendazole, mebendazole and semi-synthetic ivermectin. Anthelmintic agents are characterized by a variety of structures and mechanisms of action, which makes it difficult to develop a simple route in the search for new drugs [9,10,11,12,13,14,15,16]. Although there is an urgent need for new antiparasitic drugs, including anthelmintics, a new generation of drugs is still not available. This lack of investment in research is mainly caused by the fact that there is no interest from the pharmaceutical industry, as most parasitic diseases occur in poor countries and there is a high risk of no return on investment [17]. However, an increase in helminths’ resistance to available drugs has been observed in recent years [18,19,20]. Therefore, it is important to conduct scientific research using both natural and synthetic chemical compounds to obtain new and effective anthelmintic drugs. Many plants have drawn attention as a potential source of anthelmintic substances [9,10,21,22]. The anthelmintic activity of synthetic chemical compounds has also been demonstrated, including that of thiosemicarbazide derivatives [23,24]. Our research is in line with the trend moving towards discovering new synthetic anthelmintic compounds.
Currently, research concerning the bioactivity of triazole derivatives is very popular. Many different biological activities of 1,2,4-triazole derivatives have been described in the literature, e.g., antibacterial, antifungal, antiviral and antitumor [25,26,27,28,29]. However, there are very few reports on the anthelmintic activity of triazole derivatives and these are mainly studies conducted on earthworms, not nematodes specifically [30,31,32].
Nematodes of the genus Rhabditis sp. are free-living organisms found mainly in organic debris and soil [33]. However, there have been reports in the literature that this nematode can cause infections in humans, e.g., of the urinary tract, outer ear canal and digestive system [33,34,35,36,37,38,39,40].
In our previous publication, we described some 1,2,4-triazole derivatives which possessed methacrylic acid moieties, promising anti-inflammatory activity and low toxicity towards human lymphocytes [41]. Continuing our study on amidrazone derivatives, we took up the idea that compounds possessing a 1,2,4-triazole ring similar to (but containing one extra nitrogen atom) the diazole moiety present in anthelmintic drugs like albendazole or mebendazole could also be active against nematodes. The aim of this study was to determine the anthelmintic and anti-inflammatory activities of newly synthesized and already known 1,2,4-triazole derivatives and the anti-inflammatory activity of the new compounds.

2. Results

2.1. The Synthesis of 922

1,2,4-triazole derivatives 916 were obtained following previously established procedures via the reaction of amidrazones 1– 8 [42] with itaconic anhydride [41]. Consequently, isomerization of compounds 914 was undertaken in alkalic solution, leading to derivatives 1722 (Scheme 1). Considerably higher yields of isomerization were obtained for compounds 20 and 21, which may suggest that a 4-pyridine ring, found in the R1 position, facilitates this reaction. In contrast, no isomerization products were obtained following the reactions of 1,2,4-triazole derivatives 15 and 16, which may suggest that the presence of a 4-nitrophenyl substituent hinders this reaction. Structures of new compounds were confirmed by 1H NMR, 13C NMR and MS and their purities by elemental analyses.
The main structural difference between derivatives 914 and 1722 was the presence of a methylidene CH2= moiety attached to propanoic acid in compounds 914 and the presence of a methyl (CH3) group attached to (E)-propenoic acid in compounds 1722. Derivatives 1722 were also more thermodynamically stable than compounds 914 which is beneficial for pharmacological use. The influence of this structural difference on anthelmintic and anti-inflammatory activity was the subject of further biological research.

2.2. Biological Assays In Vitro

Human PBMC represented a tissue source in the biological part of the presented studies. Interestingly, PBMC are widely used as a physiological model for immunological research of parasitic infections, such as those caused by Toxoplasma gondii and others [43]. PBMC are routinely isolated from blood samples (as we describe previously in [44]) and then used in several different tests to determine the toxicity, anti-proliferative activity and anti-inflammatory activity of studied compounds. We used the cytometric method to evaluate the proliferation response to phytohemagglutinin (PHA) and immunoezymatic tests to verify the levels of proinflammatory cytokines after lipopolysaccharide (LPS) stimulation. The cytokines from the early immune response that were tested include: TNF-α, IL-6 and IL-10. Additionally, we verified the level of cytokines from adaptive immunity—namely, IFN-γ. IFN-γ is a very important cytokine which takes part in immune responses against parasites [45].

2.2.1. Toxicity of Compounds 922

Most of the studied 1,2,4-triazole derivatives 1122 were characterized by low toxicity towards PBMC at a concentration range up to 100 µg/mL (the range of toxicity at the highest compound concentration was 6.60–28.96% of apoptotic and necrotic cells, Figure S21, Supplementary Materials). Only derivatives 9 and 10 possessed a higher toxicity (only 52.7 and 43.40% of viable cells in 24 h PBMC culture, respectively). To compare the results, we also prepared the PBMC culture with a nonsteroidal anti-inflammatory drug—ibuprofen (the reference control); the toxicity level was 29.53% of apoptotic and necrotic cells [41]. It should be noted that all compounds that had a higher toxicity level than 30% were excluded from our research. In our previous study, PBMC cultures treated with compounds 911 and 1318 showed 71.04–82.39% viable cells [41]. The percentage of viable cells for new compounds 12, 1922 was in the range of 90.56–93.40%, which is higher than the control culture containing the same concentration of DMSO (87.27%). For comparison, five by six 1H-pyrrole-2,5-dione derivatives (obtained from the same amidrazones 18 as compounds 1122) showed about 79–92% of viable cells [46]. In general, derivatives 1722 were less toxic than 916 while compounds 10 and 18 possessing phenyl and 2-pyridine rings were the most toxic within those two groups.

2.2.2. Antiproliferative Activity of 12 and 1922

Flow cytometry assay was used to find the cytotoxic potential of compounds upon proliferation of PHA-induced PBMC. Compounds 19, 21 and IBU (reference drug) were inhibitory at a concentration of 50 µg/mL. All compounds (12, 1922) as well as IBU inhibited the proliferation of PBMC at a concentration of 100 µg/mL (the strongest inhibitory effect was observed for IBU and compound 19 causing 77.0% and 62.4% of inhibition, respectively; Figure 1). Significant inhibition of PBMC proliferation was also observed for previously studied compounds 13 and 1718 (nearly 100% at a dose of 100 µg/mL and 80% at 50 µg/mL) [41]. None of the compounds (12 and 1922) inhibited PBMC proliferation at the lowest concentration of 10 µg/mL.

2.2.3. The Influence of Compounds 12 and 1922 on Cytokine Production

The influence of newly synthetized compounds and IBU was studied on a PBMC model. PBMC were induced by LPS (an endotoxin from gram-negative bacteria used to elicit an inflammatory response in vitro). Stimulating PBMC with LPS produces a high level of pro-inflammatory cytokines such as TNF-α, IL-6, IFN-γ and anti-inflammatory cytokines IL-10 [47].

2.2.4. TNF-α

IBU significantly reduced the level of TNF-α in PBMC stimulated by LPS at all used doses (by about 99%). Compounds 12 and 1922 significantly inhibited the production of TNF-α only at a concentration of 100 µg/mL (Figure 2). The strongest activity was observed for 21 (about 84% of inhibition), while compounds 19, 20 and 22 caused about 71–74% of inhibition. Inhibitory effects were also observed for derivatives 2021 at concentrations of 50 µg/mL. Compound 12 inhibited the production of TNF-α only at the highest dose (about 50% of inhibition). Derivatives 13 and 1718 revealed stronger suppression of TNF-α production, especially in low doses (nearly 100% at 10 µg/mL, about 75% at 50 µg/mL and about 50% at 100 µg/mL) [41].

2.2.5. IL-6

Compounds 12 and 1922 had no significant effect on IL-6 production with the exception of compound 21, which elevated IL-6 production at a concentration of 100 µg/mL (Figure 3). This result is in agreement with our previous studies—compounds 911 and 1318 demonstrated no significant influence on IL-6 production [41].

2.2.6. IL-10

In the case of LPS-induced IL-10 production, neither of the compounds significantly increased the release of this cytokine. However, an inhibition of IL-10 production was observed at a concentration of 100 µg/mL for 12, 19, 20, 22 (about 71–83% inhibition) and IBU (92.3% inhibition) as well as for 22 at a concentration of 50 µg/mL (Figure 4). It is noteworthy that compound 21 inhibited the production of IL-10 the least.

2.2.7. INF-γ

Newly synthetized compound 19 (high dose), 20 (low dose) and 21 (middle and high dose) significantly reduced the production of IFN-γ (Figure 5). Interestingly, using a similar model, researchers have shown that this factor is very important for the transmission of some parasites, because it increases the expression of an adhesion marker such as intercellular adhesion molecule-1 (ICAM-1) [48]. Thus, it can be concluded that the reduction of IFN-γ production is beneficial in counteracting parasitic infections.

2.2.8. Anthelmintic Activity of Compounds 1022

Compound 12, which possesses 4-pyridyl and phenyl substituents (at R1 and R2 positions, respectively), demonstrated the highest anthelmintic activity with a lethal concentration LC50 (the amount of an ingested substance that kills 50 percent of a test sample) of 2.475 ± 0.283 µg/µL (Figure 6 and Figure 7). Moreover, compound 14, bearing 2-pyridyl and 4-methylphenyl substituents, was characterized by anthelmintic activity (LC50 = 6.550 ± 0.866 µg/µL). Both compounds were more effective than albendazole (LC50 = 19.24 µg/µL) [49]. Derivative 18 possessed significant anthelmintic activity only at a dose of 1.1 µg/µL (Figure 7). Compounds 10, 11, 13, 1517 and 1922 were characterized by a lack of anthelmintic activity.

3. Discussion

Continuing our previous research on 1,2,4-triazole derivatives obtained during the reaction between N3-substituted amidrazones and itaconic anhydride [41], we examined two groups of compounds, 916 and 1722, differing in the position of the double bond in the side chain of methylacrylic acid. Anthelmintic and anti-inflammatory activities of obtained compounds were studied to determine the influence of their structure on their biological properties.
Derivatives 1122 were not toxic to human PBMC at a concentration of 100 µg/mL in a physiological model. In our previous study, derivatives 13 and 1718 revealed the strongest anti-inflammatory activity among compounds 11 and 1318 by inhibiting both TNF-α production and proliferation of PBMC [41]. As a continuation of this work, we extended the research on the anti-inflammatory activity of derivatives 12 and 1922. Compounds 12 and 1922 (at concentration of 100 µg/mL) and derivatives 2021 (50 µg/mL) significantly inhibited production of TNF-α in LPS-stimulated PBMC (but were less effective than the IBU, Figure 2). Neither of the studied compounds lowered the level of IL-6 (Figure 3). All compounds (12 and 1922, except for 21) showed a similar inhibitory effect on the synthesis of anti-inflammatory IL-10 as IBU (Figure 4). Compounds 1922 (at a concentration of 100 µg/mL) and derivatives 19 and 21 (50 µg/mL) significantly inhibited lymphocyte proliferation in a similar way to reference IBU (Figure 1). Comparing the results with earlier studies, we confirmed that derivatives 1722 (which possess a methyl group in propenoic acid moiety) have stronger antiproliferative properties in PHA-induced PBMC cultures than compounds 1116 (which contain a CH2= group in a propanoic acid moiety) [41].
We suspected that compound 21, which contains the same 4-pyridyl and 4-methyphenyl substituents as 13 (the strongest anti-inflammatory agent among 11 and 1316), and the favorable position of the double bond will be the most active derivative in the current study. In fact, compound 21 was the only one among derivatives 1922 which inhibited lymphocyte proliferation and the production of pro-inflammatory cytokines (TNF-α and IFN-γ) at two doses: 50 µg/mL and 100 µg/mL in mitogen-stimulated PBMC cultures. Moreover, derivative 21 was the only one among compounds 1722 that did not significantly decrease the production of the anti-inflammatory cytokine IL-10 at all studied concentrations. Summing up, compound 21 has shown the most promising properties among derivatives 1922 and it is worth future research as anti-inflammatory agent.
Afterwards, we studied the anthelmintic potential of compounds 1122 on Rhabditis sp. nematodes. Our research model was similar to the Caenorhabditis elegans often used in research [50]. Rhabditis sp. nematodes were used, among others, to determine the anthelmintic activity of the new rhodanine derivatives of cinnamaldehyde [49], naphthalimide—conjugates of boron clusters [51]—and thiosemicarbazide derivatives [24].
According to our knowledge, there are only few reports in the literature on the anthelmintic activity of 1,2,4-triazole derivatives. In previous research on Indian worm Pheritima postuma activity of 1,2,4-triazole, the strongest anthelmintic activity showed derivatives possessing 2,4-dichlorophenyl [52], 4-chlorophenyl or 4-nitrophenyl moieties [53]. The diversity of substituents present in the most active compounds indicates various possibilities for the modification of potential nematicidal agents.
Among 1,2,4-triazole derivatives 1122 we studied in the Rhabditis model, compound 12 showed about eight times greater nematocidal activity than albendazole and compound 14 was approximately three times more active than the reference drug. When analyzing the results of the anthelmintic activity, we have also been able to detect a relationship between the structure and antiparasitic potential of studied compounds. The derivatives which possess a methylidene group CH2=, like 12 and 14, showed stronger anthelmintic activity than compounds obtained by their isomerization (20 and 22). Among the second group of 1,2,4-triazole derivatives (1722), only one compound 18 at a single dose 1.1 µg/µL possessed significant anthelmintic activity (Figure 7c), which indicates a lower nematocidal potential of derivatives 1722. We observed that compounds possessing a 4-nitrophenyl (15 and 16) or 2-pyridine moiety at the R2 position (17) were devoid of anthelmintic activity. However, the phenyl substituent at the R2 position could be connected with the anthelmintic activity of compounds 12 and 18. The influence of the substituent at the R1 position on anthelmintic activity is not so clear, however, the presence of a 2- or 4-pyridyl substituent at the R1 position in conjunction with the phenyl or 4-methylphenyl substituent at R2 seems to be important.
In our previous study, compound 14 revealed low acute toxicity in mice (LC50 value of 1000 mg/kg i.p.), lack of neurotoxic activity and weak effects on the central nervous system (CNS) of mice [25]. It may suggest favorable pharmacological properties of a whole group of related 1,2,4-triazole derivatives (1122). Moreover, 1,2,4-triazole derivatives 911 and 1318 demonstrated moderate antimicrobial activity (MIC ≥ 100 µg/mL) [25]. This may suggest neutrality to the human intestinal flora of studied compounds 922.
It’s worth noting that the 1,2,4-triazole ring present in studied compounds 1122 may interact with the same molecular target as albendazole (colchicine binding side). Our reference anthelmintic drug has three nitrogen atoms in a similar configuration to the compounds we studied. The structural advantage of compound 12 above other compounds could be the 4-pyridyl substituent (R1), which would detach with a similar purpose to the sulfur atom present in albendazole. The phenyl substituent in the R2 position provides better lipophilicity than the compounds 9, 1517 having an additional nitrogen atom or nitro group, none of which showed nematocidal activity. Another important element of the structure of derivative 12 seems to be the presence of the CH2= group in a methacrylic acid moiety also present in the second nematocidal compound 14.
A limited number of publications on the anthelmintic properties of triazoles indicate that further research is required. The next scientific step will be to conduct research in experimental in vivo models and to determine the molecular mechanism of action of 1,2,4-triazole derivatives 12 and 14.

4. Materials and Methods

All reagents and solvents were acquired from Sigma-Aldrich (Burlington, MA, USA) or Avantor Performance Materials Poland S.A (Gliwice, Poland). 1H NMR and 13C NMR spectra were recorded on Bruker Avance spectrophotometers (300, 400 or 700 MHz) in DMSO-d6 using TMS as an internal standard. Elemental analyses were performed on a Vario MACRO CHN analyzer (ELEMENTAR Analysensysteme GmbH, Langenselbold, Germany). Melting points were determined on a Mel TEMP 1002D apparatus and are given uncorrected. All reactions were controlled by TLC chromatography.

4.1. General Methods of Synthesis

Compounds 922 were synthesized according to established general procedures [41]:
Amidrazones 18 were obtained from corresponding thioamides and 80% aqueous hydrazine hydrate solution [42]. 0.01 mol of amidrazones 18 and 0.01 mol (1.12 g) of itaconic anhydride were dissolved in 100 mL of anhydrous diethyl ether and left in room temperature for 7 days. Obtained crude solids 916 were filtered off and purified by crystallization from a methanol−water mixture (1:1) [41,54]. Compounds 1722 were obtained by heating 0.25 g of derivatives 914 at boiling point with 20 mL of 2% NaOH aqueous solution for 2 h. After cooling down, the reaction mixture was filtered off. A 1% hydrochloric acid solution was added dropwise to the obtained filtrate for as long as precipitation was observed. Obtained crude solids of 1722 were filtered off and purified by crystallization from water or from water with the addition of methanol [41,54]. The characteristics of the new compounds 12 and 1922 are available in the Supplementary Materials (1H NMR and 13C NMR spectral data on Figures S1–S10, HRMS data on Figures S11–S15, HPLC data on Figures S16–S20). Compounds 911 and 1318 were characterized in our previous publications [41,54,55].

4.2. Biological Assays In Vitro

4.2.1. Peripheral Blood Mononuclear Cells Preparation

Peripheral blood mononuclear cells (PBMC) were isolated immediately from fresh blood as previously described [43]. After isolation, PBMC were used to conduct experiments which assessed toxicity of compounds and their impact on proliferation and cytokine production.
The cell suspensions (1 × 106 cells/mL) in culture medium (5% fetal bovine serum—FBS—Euroclone SpA, Pero, Ml, Italy, in RPMI 1640—Biomed Lublin, Lublin, Poland) were added to 5 mL tubes (Falcon® Round Bottom Polystyrene Tubes, Corning, NY, USA) in all experiments. Compounds 922 and ibuprofen (IBU) were initially dissolved in dimethyl sulfoxide (DMSO, Sigma-Aldrich) and afterwards, were added in appropriate amounts to the cell culture. Final concentrations of compounds and IBU in the cultures were 10, 50 and 100 μg/mL in all biological assays. The maximum concentration of DMSO in the individual assay was lower than 0.5%. These concentrations allowed us to avoid cell culture toxicity. The ibuprofen was selected as the reafference medicine generally used as nonsteroidal anti-inflammatory drugs (positive control). Additionally, control samples contained DMSO in the highest used dose (negative control) were prepared.

4.2.2. Toxicity of Compounds 922

Obtained PBMC were subjected to a 24 h culture (37 °C at 5% CO2 atmosphere) with the studied compounds. After stimulation, the cells were washed once (centrifuged at 400× g at 4 °C for 5 min) with PBS (phosphate-buffered saline; Biomed Lublin, Lublin, Poland) and then used for staining with fluorescein isothiocyanate (FITC)-labeled Annexin V and propidium iodide (Annexin V Apoptosis Detection Kit I, Becton Dickinson, Franklin Lakes, NJ, USA). This procedure has been described previously [43].

4.2.3. Antiproliferative Activity of 12 and 1922

Lymphocyte proliferation was evaluated by BD Horizon™ Violet Proliferation Dye 450 (VPD450, BD Pharmingen, San Diego, CA, USA). This dye is used for flow cytometric monitoring of cell division. VPD450-stained cells were cultured for 72 h with the mitogen, phytohemagglutinin (PHA, Sigma-Aldrich, 1 µg/mL, positive control), and/or increasing concentrations of compounds 12 and 1922 in DMSO (10, 50 and 100 μg/mL). Control samples contained DMSO in the highest used dose (negative control). Thereafter, culture tubes were centrifuged at 400× g at room temperature for 5 min and washed once in PBS. The cell palette was suspended in PBS and analyzed by flow cytometry (FACSCanto II flow cytometer, Becton Dickinson, Franklin Lakes, NJ, USA). Twenty thousand events were collected and analyzed with FlowJo software (v 7.6.1, Tree Star, Ashland, OR, USA).

4.2.4. The Influence of 12 and 1922 on Cytokine Production

The in vitro effect on cytokine production was measured by the enzyme-linked immunosorbent assay (ELISA). The assay was conducted as described earlier [41]. PBMC were cultured with lipopolysaccharide (LPS, from E. coli, O55:B5, Sigma-Aldrich, 1 μg/mL, positive control) and/or increasing concentrations of compounds. Cytokine levels (TNF-α, IL-6, IL-10 and IFN-γ) were measured by means of commercially available ELISA kits (DuoSet, BD Bioscience, San Diego, CA, USA) according to the manufacturer’s instructions. The samples were analyzed with iEMS Reader MF (Labsystems, Vanta, Finland). The contents of the analyzed cytokines were calculated by Ascent Software AscSW26 (London, UK).

4.2.5. Data Analysis

Data was analyzed in Statistica 13.3 software (StatSoft, Cracow, Poland) and graphed in Excel 2016. All p-values represent nonparametric Mann−Whitney U test.

4.2.6. Anthelmintic Activity of 1022

Due to their very low solubility in water, test compounds 1022 were suspended in 60% glycerol. To obtain a homogeneous emulsion, sonication was performed using an ultrasonic homogenizer (Hielscher Ultrasound Technology). The culturing of nematodes and the methodology of testing nematocidal properties was developed by the Department of Biology and Genetics of the Medical University of Lublin (patent No. PL232918, Bogucka-Kocka A., Kołodziej P., 2019). The culturing was carried out for 4–5 days at room temperature. All development stages of the nematodes were observed.
The nematodes were then eluted with 0.6% NaCl into new, sterile 24-well plates. The tested derivatives were added to the nematode cultures in experimentally selected 5 concentrations: 0.2; 1.1; 3.3; 5.5; 11.1 µg/µL.
The anthelmintic activity was carried out according to the protocol described previously [24,38,48,50]. ANOVA and the Tukey test (GraphPad Prism 5.02 software, San Diego, CA, USA) was used for statistical analysis. Differences with p < 0.05 *, p < 0.01 ** and p < 0.001 *** were considered statistically significant [48,51].

5. Conclusions

Some biological properties of the 1,2,4-triazole derivatives 922 were experimentally evaluated. Most compounds (1122) proved to be non-toxic to human PBMC. In anti-inflammatory studies, derivatives 1922 showed higher activity than derivative 12 and previously tested compounds 11, 1416. The most active compound, 21, acts in a number of ways: inhibiting the proliferation of PBMC and reducing the production of TNF-α and INF-γ in mitogen stimulated PBMC cultures. In general, the propenoic acid derivatives 1722 had a stronger anti-inflammatory potential than compounds 1116 differing in position of the double bond in the side chain.
Owing to their high anthelmintic potential, which was proved to be higher than that of the recommended drug albendazole, we have selected compounds 12 and 14 as potential candidates for further research. Derivative 12 also showed anti-inflammatory activity (by inhibiting TNF-α production), which constitutes an additional advantage. Interestingly, the clinically used anthelmintic drug ivermectin also has an anti-inflammatory effect [56]. Thanks to their anthelmintic and anti-inflammatory properties, the selected compounds may, in the future, be used in the treatment of parasitic diseases caused by intestinal nematodes in humans.

6. Patents

Part of the results of the work were used to prepare the Polish patent application No. P.434865.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/molecules27144488/s1, Figure S1: 1H NMR spectrum of compound 12 (in DMSO-d6). Figure S2: 1H NMR spectrum of compound 19 (in DMSO-d6). Figure S3: 1H NMR spectrum of compound 20 (in DMSO-d6). Figure S4: 1H NMR spectrum of compound 21 (in DMSO-d6). Figure S5: 1H NMR spectrum of compound 22 (in DMSO-d6). Figure S6. 13C NMR spectrum of compound 12 (in DMSO-d6). Figure S7. 13C NMR spectrum of compound 19 (in DMSO-d6). Figure S8. 13C NMR spectrum of compound 20 (in DMSO-d6). Figure S9. 13C NMR spectrum of compound 21 (in DMSO-d6). Figure S10. 13C NMR spectrum of compound 22 (in DMSO-d6). Figure S11. HRMS data of compound 12. Figure S12. HRMS data of compound 19. Figure S13. HRMS data of compound 20. Figure S14. HRMS data of compound 21. Figure S15. HRMS data of compound 22. Figure S16. HPLC data of compound 12. Figure S17. HPLC data of compound 19. Figure S18. HPLC data of compound 20. Figure S19. HPLC data of compound 21. Figure S20. HPLC data of compound 22. Figure S21: Cell apoptosis of compounds 921 measured by flow cytometry.

Author Contributions

Conceptualization, R.P.; methodology, M.W.-S., A.B.-K. and P.K.; software, A.H.-B.; validation, A.H.-B., M.W.-S. and P.K.; formal analysis, R.P.; investigation, R.P., P.K. and M.W.-S.; resources, R.P.; writing—original draft preparation, R.P., P.K. and M.W.-S.; writing—review and editing, A.B.-K., A.H.-B., P.K. and M.W.-S.; visualization, R.P.; supervision R.P., P.K., M.W.-S. and A.B.-K.; project administration, R.P.; funding acquisition, A.B.-K., R.P. and P.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Statutory Funds of the Medical University of Lublin provided by the Polish Ministry of Science and Higher Education, grant number DS43 AB-K and National Science Centre, Poland, No. 2017/01/X/NZ6/01001. This work has been supported by Nicolaus Copernicus University, Collegium Medicum (PDB and IDUB IDE HS-2020 Renata Paprocka).

Institutional Review Board Statement

Experiments using peripheral blood mononuclear cells (PBMC) ware conducted according to the guidelines of the Declaration of Helsinki and were approved by the Collegium Medicum of Nicolaus Copernicus University Bioethical Commission (KB 39/2019).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

Data is available from the authors.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hajare, S.T.; Gobena, R.K.; Chauhan, N.M.; Erniso, F. Prevalence of Intestinal Parasite Infections and Their Associated Factors among Food Handlers Working in Selected Catering Establishments from Bule Hora, Ethiopia. BioMed Res. Int. 2021, 2021, 6669742. [Google Scholar] [CrossRef] [PubMed]
  2. Tigabu, A.; Taye, S.; Aynalem, M.; Adane, K. Prevalence and associated factors of intestinal parasitic infections among patients attending Shahura Health Center, Northwest Ethiopia. BMC Res. Notes 2019, 12, 333. [Google Scholar] [CrossRef] [PubMed]
  3. Kiani, H.; Haghighi, A.; Salehi, R.; Azargashb, E. Distribution and risk factors associated with intestinal parasite infections among children with gastrointestinal disorders. Gastroenterol. Hepatol. Bed Bench. 2016, 9, S80–S87. [Google Scholar]
  4. Bertoncello, C.; Amoruso, I.; Fonzo, M.; Zanzot, D.; Baldo, V.; Buja, A.; Baldovin, T. Epidemiology of intestinal parasitic diseases in Nepalese children: Chiara Bertoncello. Eur. J. Public Health 2017, 27, ckx186.146. [Google Scholar] [CrossRef] [Green Version]
  5. Haque, R. Human intestinal parasites. J. Health Popul. Nutr. 2007, 25, 387–391. [Google Scholar]
  6. Hechenbleikner, E.M.; McQuade, J.A. Parasitic colitis. Clin. Colon Rectal Surg. 2015, 28, 79–86. [Google Scholar] [CrossRef] [Green Version]
  7. Feleke, D.G.; Alemu, Y.; Bisetegn, H.; Mekonnen, M.; Yemanebrhane, N. Intestinal parasitic infections and associated factors among street dwellers and prison inmates: A systematic review and meta-analysis. PLoS ONE 2021, 16, e0255641. [Google Scholar] [CrossRef]
  8. Faria, C.P.; Zanini, G.M.; Dias, G.S.; da Silva, S.; de Freitas, M.B.; Almendra, R.; Santana, P.; Sousa, M.D. Geospatial distribution of intestinal parasitic infections in Rio de Janeiro (Brazil) and its association with social determinants. PLoS Negl. Trop. Dis. 2017, 11, e0005445. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Romero-Benavides, J.C.; Ruano, A.L.; Silva-Rivas, R.; Castillo-Veintimilla, P.; Vivanco-Jaramillo, S.; Bailon-Moscoso, N. Medicinal plants used as anthelmintics: Ethnomedical, pharmacological, and phytochemical studies. Eur. J. Med. Chem. 2017, 129, 209–217. [Google Scholar] [CrossRef]
  10. Jayawardene, K.L.T.D.; Palombo, E.A.; Boag, P.R. Natural Products Are a Promising Source for Anthelmintic Drug Discovery. Biomolecules 2021, 11, 1457. [Google Scholar] [CrossRef]
  11. Nixon, S.A.; Welz, C.; Woods, D.J.; Costa-Junior, L.; Zamanian, M.; Martin, R.J. Where are all the anthelmintics? Challenges and opportunities on the path to new anthelmintics. Int. J. Parasitol. Drugs Drug Res. 2020, 14, 8–16. [Google Scholar] [CrossRef] [PubMed]
  12. Laing, R.; Gillan, V.; Devaney, E. Ivermectin—Old Drug, New Tricks? Trends Parasitol. 2017, 33, 463–472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Partridge, F.A.; Forman, R.; Bataille, C.J.R.; Wynne, G.M.; Nick, M.; Russell, A.J.; Else, K.J.; Sattelle, D.B. Anthelmintic drug discovery: Target identification, screening methods and the role of open science. Beilstein J. Org. Chem. 2020, 16, 1203–1224. [Google Scholar] [CrossRef] [PubMed]
  14. Campbell, S.; Soman-Faulkner, K. Antiparasitic Drugs. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2021. Available online: https://www.ncbi.nlm.nih.gov/books/NBK544251 (accessed on 24 February 2022).
  15. Kappagoda, S.; Singh, U.; Blackburn, B.G. Antiparasitic therapy. Mayo Clin. Proc. 2011, 86, 561–583. [Google Scholar] [CrossRef] [Green Version]
  16. Holden-Dye, L.; Walker, R.J. Anthelmintic drugs. WormBook 2007, 2, 1–13. [Google Scholar] [CrossRef]
  17. Pink, R.; Hudson, A.; Mouriès, M.-A.; Bendig, M. Opportunities and challenges in antiparasitic drug discovery. Nat. Rev. Drug Discov. 2005, 4, 727–740. [Google Scholar] [CrossRef]
  18. Geerts, S.; Gryseels, B. Drug resistance in human helminths: Current situation and lessons from livestock. Clin. Microbiol. Rev. 2000, 13, 207–222. [Google Scholar] [CrossRef]
  19. Geerts, S.; Gryseels, B. Anthelmintic resistance in human helminths: A review. Trop. Med. Int. Health. 2001, 6, 915–921. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Fissiha, W.; Kinde, M.Z. Anthelmintic Resistance and Its Mechanism: A Review. Infect. Drug Resist. 2021, 15, 5403–5410. [Google Scholar] [CrossRef]
  21. Grzybek, M.; Kukula-Koch, W.; Strachecka, A.; Jaworska, A.; Phiri, A.M.; Paleolog, J.; Tomczuk, K. Evaluation of Anthelmintic Activity and Composition of Pumpkin (Cucurbita pepo L.) Seed Extracts-In Vitro and in Vivo Studies. Int. J. Mol. Sci. 2016, 17, 1456. [Google Scholar] [CrossRef]
  22. Mravčáková, D.; Komáromyová, M.; Babják, M.; Urda Dolinská, M.; Königová, A.; Petrič, D.; Čobanová, K.; Ślusarczyk, S.; Cieslak, A.; Várady, M.; et al. Anthelmintic Activity of Wormwood (Artemisia absinthium L.) and Mallow (Malva sylvestris L.) against Haemonchuscontortus in Sheep. Animals 2020, 10, 219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Alfaro-Fuentes, I.; Castro-Ramírez, R.; Ortiz-Pastrana, N.; Medina-Guerrero, R.M.; Soler-Jiménez, L.C.; Martínez-Rodríguez, I.; Betancourt-Lozano, M.; Ibarra-Castro, L.; Barba-Behrens, N.; Fajer-Ávila, E.J. Novel antihelmintic activity of tinidazole coordination compounds. Relevance of the metal ion and structural properties. J. Inorg. Biochem. 2017, 176, 159–167. [Google Scholar] [CrossRef] [PubMed]
  24. Dziduch, K.; Kołodziej, P.; Paneth, A.; Bogucka-Kocka, A.; Wujec, M. Synthesis and Anthelmintic Activity of New Thiosemicarbazide Derivatives—A Preliminary Study. Molecules 2020, 25, 2770. [Google Scholar] [CrossRef]
  25. Paprocka, R.; Modzelewska-Banachiewicz, B.; Kutkowska, J.; Pawłowski, K.; Piątkowska-Chmiel, I.; Jagiełło-Wójtowicz, E. Antibacterial and central nervous system activity of (4,5-diaryl-4H-1,2,4-triazol-3-yl) methacrylic acid derivatives. Acta Pol. Pharm. 2017, 74, 289–292. [Google Scholar]
  26. Kosikowska, U.; Andrzejczuk, S.; Plech, T.; Malm, A. Inhibitory Effect of 1,2,4-Triazole-Ciprofloxacin Hybrids on Haemophilus Parainfluenzae and Haemophilus Influenzae Biofilm Formation in Vitro under Stationary Conditions. Res. Microbiol. 2016, 167, 647–654. [Google Scholar] [CrossRef] [PubMed]
  27. Jiang, Y.; Zhang, J.; Cao, Y.; Chai, X.; Zou, Y.; Wu, Q.; Zhang, D.; Jiang, Y.; Sun, Q. Synthesis, in vitro evaluation and molecular docking studies of new triazole derivatives as antifungal agents. Bioorg. Med. Chem. Lett. 2011, 21, 4471–4475. [Google Scholar] [CrossRef] [PubMed]
  28. El Azab, I.H.; El-Sheshtawy, H.S.; Bakr, R.B.; Elkanzi, N.A.A. New 1,2,3-Triazole-Containing Hybrids as Antitumor Candidates: Design, Click Reaction Synthesis, DFT Calculations, and Molecular Docking Study. Molecules 2021, 26, 708. [Google Scholar] [CrossRef]
  29. Wujec, M.; Plech, T.; Siwek, A.; Rajtar, B.; Polz-Dacewiczb, M. Synthesis and in vitro study of antiviral and virucidal activity of novel 2-[(4-methyl-4H-1,2,4-triazol-3-yl)sulfanyl]acetamide derivatives. J. Z. Nat. C 2011, 66, 333–339. [Google Scholar] [CrossRef]
  30. Gupta, J.K.; Mishra, P. Antimicrobial and anthelmintic activities of some newly synthesized triazoles. Asian J. Pharm. Clin. Res. 2017, 10, 139–145. [Google Scholar] [CrossRef]
  31. Sahoo, B.M.; Nagamounika, K.; Banik, B.K. Microwave-assisted synthesis of Schiff’s bases of 1,2,4-triazole derivatives and their anthelmintic activity. J. Indian Chem. Soc. 2018, 95, 1289–1294. [Google Scholar]
  32. Jayappa, M.K.D.; Dasappa, J.P.; Chandrashekar, K.R.; Sheik, S.; Chaluvaiah, K.; Naik, N. Synthesis, Antimicrobial and Anthelmintic Activity Studies of Some Novel Triazole Schiff and Mannich Bases. Pharma Chem. 2017, 9, 163–171. [Google Scholar]
  33. Teschner, M.; Würfel, W.; Sedlacek, L.; Suerbaum, S.; Tappe, D.; Hornef, M.W. Outer ear canal infection with Rhabditis sp. nematodes in a human. J. Clin. Microbiol. 2014, 52, 1793–1795. [Google Scholar] [CrossRef] [Green Version]
  34. Tehrani, M.F.; Sharifdini, M.; Zahabiun, F.; Latifi, R.; Kia, E.B. Molecular characterization of human isolates of Strongyloides stercoralis and Rhabditis spp. based on mitochondrial cytochrome c oxidase subunit 1 (cox1). BMC Infect. Dis. 2019, 19, 776. [Google Scholar] [CrossRef] [Green Version]
  35. Meamar, A.R.; Kia, E.B.; Zahabiun, F.; Jafari-Mehr, A.; Moghadam, A.; Sadjjadi, S.M. The occurrence of severe infections with Rhabditis axei in AIDS patients in Iran. J. Helminthol. 2007, 81, 351–352. [Google Scholar] [CrossRef]
  36. Campos, D.M.; Araujo, J.L.; Vieira, M.C.; Damasceno, F.; Barbosa, A.P. A case of parasitism by Rhabditis sp. in a child from Goiania, Goias. Brazil Rev. Soc. Bras. Med. Trop. 2002, 35, 519–522. [Google Scholar] [CrossRef] [Green Version]
  37. Ahn, Y.K.; Chung, P.R.; Lee, K.T. Rhabditis sp. infected cases in rural school children. Kisaengchunghak Chapchi 1985, 23, 1–6. [Google Scholar] [CrossRef]
  38. Ziaja-Sołtys, M.; Kołodziej, P.; Stefaniuk, D.; Matuszewska, A.; Jaszek, M.; Bogucka-Kocka, A. Low-Molecular-Weight Secondary Metabolites from Fungi: Cerrena unicolor as a New Proposal of an Effective Preparation against Rhabditis Nematodes. Molecules 2022, 27, 1660. [Google Scholar] [CrossRef] [PubMed]
  39. He, Y.X.; Jiang, H. 3 human cases of urinary tract infection with Rhabditis. J. Parasitol. Parasit. Dis. 1985, 3, 206–208. [Google Scholar]
  40. Ye, L.P.; Zhu, C.G.; Zhang, J.N. Two cases of Rhaditis axei infections in human disgestive system. Chin. J. Schistosomiasis Cont. 2002, 14, 187. [Google Scholar]
  41. Paprocka, R.; Wiese, M.; Eljaszewicz, A.; Helmin-Basa, A.; Gzella, A.; Modzelewska-Banachiewicz, B.; Michalkiewicz, J. Synthesis and anti-inflammatory activity of new 1,2,4-triazole derivatives. Bioorg. Med. Chem. Lett. 2015, 25, 2664–2667. [Google Scholar] [CrossRef]
  42. Modzelewska, B.; Pyra, E. Synthesis of N3-substituted amidrazones. Ann. UMCS Sec. AA 1996, 50, 111–116. [Google Scholar]
  43. Davila, J.A.A.; Rios, A.H.D.L. An Overview of Peripheral Blood Mononuclear Cells as a Model for Immunological Research of Toxoplasma gondii and Other Apicomplexan Parasites. Front. Cell. Infect. Microbiol. 2019, 9, 24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Paprocka, R.; Wiese-Szadkowska, M.; Helmin-Basa, A.; Mazur, L.; Kutkowska, J.; Michałkiewicz, J.; Modzelewska-Banachiewicz, B.; Pazderski, L. Synthesis and evaluation of new amidrazone-derived hydrazides as a potential anti-inflammatory agents. Mon. Chem. 2018, 149, 1493–1500. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Morales, M.A.G.; La Rosa, G.; Ludovisi, A.; Onori, A.M.; Pozio, E. Cytokine profile induced by Cryptosporidium antigen in peripheral blood mononuclear cells from immunocompetent and immunosuppressed persons with cryptosporidiosis. J. Infect. Dis. 1999, 179, 967–973. [Google Scholar]
  46. Paprocka, R.; Pazderski, L.; Mazur, L.; Wiese-Szadkowska, M.; Kutkowska, J.; Nowak, M.; Helmin-Basa, A. Synthesis and Structural Study of Amidrazone Derived Pyrrole-2,5-Dione Derivatives: Potential Anti-Inflammatory Agents. Molecules 2022, 27, 2891. [Google Scholar] [CrossRef]
  47. Yücel, G.; Zhao, Z.; El-Battrawy, I.; Lan, H.; Lang, S.; Li, X.; Buljubasic, F.; Zimmermann, W.H.; Cyganek, L.; Utikal, J.; et al. Lipopolysaccharides induced inflammatory responses and electrophysiological dysfunctions in human-induced pluripotent stem cell derived cardiomyocytes. Sci. Rep. 2017, 7, 2935. [Google Scholar] [CrossRef]
  48. Pfaff, A.W.; Georges, S.; Abou-Bacar, A.; Letscher-Bru, V.; Klein, J.; Mousli, M.; Candolfi, E. Toxoplasma gondii regulates ICAM-1 mediated monocyte adhesion to trophoblasts. Immunol. Cell Biol. 2005, 83, 483–489. [Google Scholar] [CrossRef]
  49. Tejchman, W.; Kołodziej, P.; Kalinowska-Tłuścik, J.; Nitek, W.; Żuchowski, G.; Bogucka-Kocka, A.; Żesławska, E. Discovery of Cinnamylidene Derivative of Rhodanine with High Anthelmintic Activity against Rhabditis sp. Molecules 2022, 27, 2155. [Google Scholar] [CrossRef]
  50. Kiontke, K.; Fitch, D.H. The phylogenetic relationships of Caenorhabditis and other rhabditids. WormBook 2005, 1, 11. [Google Scholar] [CrossRef] [Green Version]
  51. Bogucka-Kocka, A.; Kołodziej, P.; Makuch-Kocka, A.; Różycka, D.; Rykowski, S.; Nekvinda, J.; Grüner, B.; Olejniczak, A.B. Nematicidal activity of naphthalimide-boron cluster conjugates. Chem. Commun. 2022, 58, 2528–2531. [Google Scholar] [CrossRef]
  52. Kumar, S.P.; Sahoo, J. Anthelmintic Evaluation of Some Novel Synthesized 1, 2, 4-Triazole Moiety Clubbed with Benzimidazole Ring. Orient. J. Chem. 2014, 30, 211–217. [Google Scholar] [CrossRef] [Green Version]
  53. Khan, G.; Sreenivasa, S.; Govindaiah, S.; Chandramohan, V.; Shetty, P.R. Synthesis, biological screening, in silico study and fingerprint applications of novel 1,2,4-triazole derivatives. J. Heterocycl. Chem. 2020, 57, 2010–2023. [Google Scholar] [CrossRef]
  54. Modzelewska-Banachiewicz, B.; Paprocka, R.; Mazur, L.; Saczewski, J.; Kutkowska, J.; Stepien, D.K.; Cyranski, M. Experimental and theoretical study on the reaction of N3-phenyl-(pyridin-2-yl)carbohydrazonamide with itaconic anhydride. J. Mol. Struct. 2012, 1022, 211–219. [Google Scholar] [CrossRef]
  55. Paprocka, R.; Modzelewska-Banachiewicz, B.; Gzella, A.K. 2-{[5-(Pyridin-4-yl)-4-p-tolyl-4H-1,2,4-triazol-3-yl]methyl}acrylic acid hemihydrate. Acta Cryst. 2014, E70, o95–o96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Martin, R.J.; Robertson, A.P.; Choudhary, S. Ivermectin: An Anthelmintic, an Insecticide, and Much More. Trends Parasitol. 2021, 37, 48–64. [Google Scholar] [CrossRef] [PubMed]
Scheme 1. The synthesis of compounds 922.
Scheme 1. The synthesis of compounds 922.
Molecules 27 04488 sch001
Figure 1. The effect of compounds 12, 1922 and IBU on the PHA-induced PBMC proliferation in 72 h cell cultures (the results are shown as percentage of positive control (PHA alone), with values expressed as medians from six independent experiments and interquartile ranges [Q1, Q3]); ** indicates a significant difference compared to positive control (PHA alone) at p < 0.01.
Figure 1. The effect of compounds 12, 1922 and IBU on the PHA-induced PBMC proliferation in 72 h cell cultures (the results are shown as percentage of positive control (PHA alone), with values expressed as medians from six independent experiments and interquartile ranges [Q1, Q3]); ** indicates a significant difference compared to positive control (PHA alone) at p < 0.01.
Molecules 27 04488 g001
Figure 2. The effect of compounds 12, 1922 and IBU on the LPS-induced production of TNF-α in PBMC cultures (the results are shown as percentage of positive control (LPS alone), with values expressed as medians from previous independent experiments and confidence intervals); * indicates a significant difference compared to the positive control (LPS alone) at p < 0.05.
Figure 2. The effect of compounds 12, 1922 and IBU on the LPS-induced production of TNF-α in PBMC cultures (the results are shown as percentage of positive control (LPS alone), with values expressed as medians from previous independent experiments and confidence intervals); * indicates a significant difference compared to the positive control (LPS alone) at p < 0.05.
Molecules 27 04488 g002
Figure 3. The effect of compounds 12, 1922 and IBU on the LPS-induced production of IL-6 in PBMC cultures (the results are shown as percentage of positive control (LPS alone), with values expressed as medians from five independent experiments and confidence intervals); ** indicates a significant difference compared to positive control (LPS alone) at p < 0.01.
Figure 3. The effect of compounds 12, 1922 and IBU on the LPS-induced production of IL-6 in PBMC cultures (the results are shown as percentage of positive control (LPS alone), with values expressed as medians from five independent experiments and confidence intervals); ** indicates a significant difference compared to positive control (LPS alone) at p < 0.01.
Molecules 27 04488 g003
Figure 4. The effect of compounds 12, 1922 and IBU on the LPS-induced production of IL-10 in PBMC cultures (the results are shown as percentage of positive control (LPS alone), with values expressed as medians from five independent experiments and confidence intervals); * indicates significant difference comparing to positive control (LPS alone) at 0.01 < p < 0.05; ** indicates a significant difference compared to positive control at p < 0.01.
Figure 4. The effect of compounds 12, 1922 and IBU on the LPS-induced production of IL-10 in PBMC cultures (the results are shown as percentage of positive control (LPS alone), with values expressed as medians from five independent experiments and confidence intervals); * indicates significant difference comparing to positive control (LPS alone) at 0.01 < p < 0.05; ** indicates a significant difference compared to positive control at p < 0.01.
Molecules 27 04488 g004
Figure 5. The effect of 12 and 1922 on the LPS-induced production of INF-γ in PBMC cultures (the results are shown as percentage of positive control (LPS alone), with values expressed as medians from previous independent experiments and confidence intervals); * indicates a significant difference compared to positive control (LPS alone) at p < 0.05.
Figure 5. The effect of 12 and 1922 on the LPS-induced production of INF-γ in PBMC cultures (the results are shown as percentage of positive control (LPS alone), with values expressed as medians from previous independent experiments and confidence intervals); * indicates a significant difference compared to positive control (LPS alone) at p < 0.05.
Molecules 27 04488 g005
Figure 6. An exemplary nematode culture of Rhabditis sp. after 24 h exposure to compound 12. (a) control (blue arrows—alive, mobile), (b) after treatment (red arrows—dead, non-mobiles).
Figure 6. An exemplary nematode culture of Rhabditis sp. after 24 h exposure to compound 12. (a) control (blue arrows—alive, mobile), (b) after treatment (red arrows—dead, non-mobiles).
Molecules 27 04488 g006aMolecules 27 04488 g006b
Figure 7. Viability (%) of Rhabditis sp. nematodes after 24 h exposure to increasing concentrations of the compounds (a) 12, (b) 14, (c) 18 and (d) albendazole [38,49] (p < 0.05 *, p < 0.01 **, p < 0.001 ***).
Figure 7. Viability (%) of Rhabditis sp. nematodes after 24 h exposure to increasing concentrations of the compounds (a) 12, (b) 14, (c) 18 and (d) albendazole [38,49] (p < 0.05 *, p < 0.01 **, p < 0.001 ***).
Molecules 27 04488 g007aMolecules 27 04488 g007b
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Paprocka, R.; Kołodziej, P.; Wiese-Szadkowska, M.; Helmin-Basa, A.; Bogucka-Kocka, A. Evaluation of Anthelmintic and Anti-Inflammatory Activity of 1,2,4-Triazole Derivatives. Molecules 2022, 27, 4488. https://doi.org/10.3390/molecules27144488

AMA Style

Paprocka R, Kołodziej P, Wiese-Szadkowska M, Helmin-Basa A, Bogucka-Kocka A. Evaluation of Anthelmintic and Anti-Inflammatory Activity of 1,2,4-Triazole Derivatives. Molecules. 2022; 27(14):4488. https://doi.org/10.3390/molecules27144488

Chicago/Turabian Style

Paprocka, Renata, Przemysław Kołodziej, Małgorzata Wiese-Szadkowska, Anna Helmin-Basa, and Anna Bogucka-Kocka. 2022. "Evaluation of Anthelmintic and Anti-Inflammatory Activity of 1,2,4-Triazole Derivatives" Molecules 27, no. 14: 4488. https://doi.org/10.3390/molecules27144488

Article Metrics

Back to TopTop