Next Article in Journal
Unveiling the Multifaceted Capabilities of Endophytic Aspergillus flavus Isolated from Annona squamosa Fruit Peels against Staphylococcus Isolates and HCoV 229E—In Vitro and In Silico Investigations
Previous Article in Journal
Enhanced Anti-Inflammatory Activity of Tilianin Based on the Novel Amorphous Nanocrystals
Previous Article in Special Issue
The Synthesis, In Vitro Bio-Evaluation, and In Silico Molecular Docking Studies of Pyrazoline–Thiazole Hybrid Analogues as Promising Anti-α-Glucosidase and Anti-Urease Agents
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Exploring the Potential Biological Activities of Pyrazole-Based Schiff Bases as Anti-Diabetic, Anti-Alzheimer’s, Anti-Inflammatory, and Cytotoxic Agents: In Vitro Studies with Computational Predictions

by
Ahmed M. Naglah
1,*,
Abdulrahman A. Almehizia
1,
Asma S. Al-Wasidi
2,
Amirah Senaitan Alharbi
3,
Mohammed H. Alqarni
4,
Ashraf S. Hassan
5,* and
Wael M. Aboulthana
6
1
Drug Exploration and Development Chair (DEDC), Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
2
Department of Chemistry, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh 11671, Saudi Arabia
3
King Khalid Hospital, King Saud University Medical City, P.O. Box 7805, Riyadh 11472, Saudi Arabia
4
Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
5
Organometallic and Organometalloid Chemistry Department, National Research Centre, Dokki 12622, Cairo, Egypt
6
Biochemistry Department, Biotechnology Research Institute, National Research Centre, Dokki 12622, Cairo, Egypt
*
Authors to whom correspondence should be addressed.
Pharmaceuticals 2024, 17(5), 655; https://doi.org/10.3390/ph17050655
Submission received: 22 April 2024 / Revised: 14 May 2024 / Accepted: 15 May 2024 / Published: 17 May 2024
(This article belongs to the Special Issue Pyrazole and Thiazole Derivatives in Medicinal Chemistry)

Abstract

:
In this innovative research, we aim to reveal pyrazole-based Schiff bases as new multi-target agents. In this context, we re-synthesized three sets of pyrazole-based Schiff bases, 5af, 6af, and 7af, to evaluate their biological applications. The data from in vitro biological assays (including antioxidant and scavenging activities, anti-diabetes, anti-Alzheimer’s, and anti-inflammatory properties) of the pyrazole-based Schiff bases 5af, 6af, and 7af showed that the six pyrazole-based Schiff bases 5a, 5d, 5e, 5f, 7a, and 7f possess the highest biological properties among the compounds evaluated. The cytotoxicity against lung (A549) and colon (Caco-2) human cancer types, as well as normal lung (WI-38) cell lines, was evaluated. The data from the cytotoxicity investigation demonstrated that the three Schiff bases 5d, 5e, and 7a are active against lung (A549) cells, while the two Schiff bases 5e and 7a exhibited the highest cytotoxicity towards colon (Caco-2) cells. Additionally, the enzymatic activities against caspase-3 and Bcl-2 of the six pyrazole-based Schiff bases 5a, 5d, 5e, 5f, 7a, and 7f were evaluated. Furthermore, we assessed the in silico absorption, distribution, metabolism, and toxicity (ADMT) properties of the more potent pyrazole-based Schiff bases. After modifying the structures of the six pyrazole-based Schiff bases, we plan to further extend the studies in the future.

Graphical Abstract

1. Introduction

Nowadays, various studies correlated to drug design focus on creating agents with a broad spectrum of physiological effects against diverse diseases. Various human diseases such as cancer, diabetes, Alzheimer’s disease, and inflammation have risks and relationships between them from varied causative factors and complications. Therefore, scientific cooperation is necessary to produce a molecule that possesses multiple activities. This strategy is called one drug–multiple targets or multi-target agents [1,2,3,4,5,6]. A literature survey revealed that many studies are based on this strategy. For example, the study by Loganathan and his team in 2024 referred to anthraquinone-connected coumarin derivatives that may act as multi-target agents [7]. Additionally, Almehizia and his colleagues’ research indicated that pyrazolo[1, 5-a]pyrimidine derivatives possess various biological activities against cancer, diabetes, Alzheimer’s disease, and inflammation. Therefore, they were suggested as multi-target agents [8].
Schiff bases are well known for their various biological effects [9], and numerous studies have established this [10,11,12,13]. In 2024, Çavuş et al. prepared carbohydrazide-based Schiff base A, which exhibits an antibacterial effect against Bacillus cereus, interacts with plasmid DNA, and has cytotoxic effects on HT-29 cell lines [14]. The study by Abdel-Baky et al. proved that chitosan–quinoline Schiff base derivative B possesses antibacterial and antioxidant effects and has high competence as an anti-diabetic agent through the inhibition of α-amylase and α-glucosidase enzymes [15]. The cooperation of Koçyiğit with his co-workers proved that a chalcone derivative incorporating Schiff base C acts as an acetylcholinesterase (AChE) inhibitor for treating Alzheimer’s disease [16]. In 2018, Hanif et al. prepared a 1, 2, 4-triazole-5(4H)-thione based-Schiff base with ferrocenyl group D, which acts as a potent anti-inflammatory and antioxidant agent [17] (Figure 1).
The pyrazole scaffold has received considerable attention because of its promising biological applications [18,19,20,21]. For examples, Mortada et al. (2024) synthesized a pyrazole-triazole derivative, E, that displayed powerful inhibition of α-glucosidase and α-amylase enzymes. Additionally, a pyrazole-triazole derivative showed an excellent antioxidant activity and radical scavenging abilities [22]. In 2023, Alkahtani et al. prepared a pyrazole derivative, F, (also an example of a pyrazole-based Schiff base) that acts as a multi-target agent against the α-amylase enzyme (an anti-diabetic agent), AChE enzyme (an anti-Alzheimer’s agent), and displays influential antioxidant properties [23]. From our previous work, the prepared isatin-pyrazole derivative G showed high antioxidant, anti-diabetes, anti-Alzheimer’s, and anti-arthritis properties [24] (Figure 1).
Based on the findings obtained from the previous studies mentioned above, such as the relationships between various human diseases, the biological effects of Schiff bases and the pyrazole scaffold, our research on multi-target agents, and the synthesis of biologically active heterocyclic compounds [25,26,27,28,29,30], we have been encouraged to re-synthesize three sets of pyrazole-based Schiff bases, 5af, 6af, and 7af, from our previous works to evaluate their biological applications, including the antioxidant and scavenging activities, the anti-diabetes, anti-Alzheimer’s, anti-inflammatory, cytotoxicity, and enzymatic properties. Our goal is to find new candidates with multiple biological applications. Additionally, we assessed the in silico ADMT properties of the more potent pyrazole-based Schiff bases.

2. Results

2.1. Chemistry

The various starting materials, 5-aminopyrazoles 1af [31,32], 4-(piperidin-1-yl)benzaldehyde (2) [33], 5-chloro-3-methyl-1-phenyl-1H-pyrazole-4-carbaldehyde (3) [34], and 1,5-dimethyl-3-oxo-2-phenyl-2,3-dihydro-1H-pyrazole-4-carbaldehyde (4), were used as the basic materials for re-synthesizing the three sets of pyrazole-based Schiff bases. Here, we re-synthesized three sets of pyrazole-based Schiff bases (5af, 6af, and 7af) via the reaction of 1af with 2, 3, and 4, respectively, according to the literature procedures mentioned in our previous works [35,36] (Scheme 1).

2.2. Biological Evaluations

2.2.1. The Antioxidant Activity

The antioxidant and scavenging activities of the pyrazole-based Schiff bases 5af, 6af, and 7af were assessed at equal concentrations (Table 1). It was found that the compounds 5a, 5d, 5e, 5f, 7a, and 7f exhibited higher total antioxidant capacities (TACs) and iron-reducing power (IRP) than the other pyrazole-based Schiff bases. The compounds 5b and 7e showed the lowest TACs (15.26 ± 0.06 and 15.68 ± 0.06 mg gallic acid/g, respectively) and IRP (10.01 ± 0.02 and 10.29 ± 0.02 µg/mL, respectively).
Regarding the scavenging activity, it was noticed that the compounds 5a, 5d, 5e, 5f, 7a, and 7f had lower IC50 values for 1,1-diphenyl-2-picryl-hydrazyl (DPPH, 26.43 ± 0.01, 23.98 ± 0.01, 23.71 ± 0.01, 22.83 ± 0.01, 24.94 ± 0.01, and 21.37 ± 0.01 µM, respectively) and higher inhibition percentages against the 2, 2‘-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) (ABTS radical, 75.46 ± 0.05, 78.18 ± 0.05, 76.87 ± 0.05, 76.74 ± 0.05, 75.59 ± 0.05, and 78.05 ± 0.05%, respectively) compared to the other pyrazole-based Schiff bases. Furthermore, the highest IC50 values for DPPH were noticed with the compounds 5b and 7e (42.45 ± 0.02 and 51.98 ± 0.03 µM, respectively), and they showed the lowest inhibition percentages against the ABTS radical (22.03 ± 0.01 and 22.65 ± 0.01%, respectively).

2.2.2. The Anti-Diabetic Activity

The anti-diabetic activities of the pyrazole-based Schiff bases 5af, 6af, and 7af were assessed in the present study by determining their inhibitory effects against α-amylase and α-glucosidase (Figure 2, see the Supplementary Materials Table S1). It was observed that the pyrazole-based Schiff bases 5a, 5d, 5e, 5f, 7a, and 7f possessed a higher inhibitory effect on α-amylase (31.24 ± 0.05, 32.37 ± 0.05, 31.83 ± 0.05, 31.77 ± 0.05, 31.30 ± 0.05, 32.31 ± 0.05%, respectively) and α-glucosidase (26.87 ± 0.05, 27.84 ± 0.05, 27.37 ± 0.05, 27.33 ± 0.05, 26.91 ± 0.05, and 27.79 ± 0.05%, respectively) compared to the efficiency of acarbose against α-amylase (76.58 ± 0.01%) and α-glucosidase activities (53.94 ± 0.01%) at equal concentrations. Regarding the compounds 5b and 7e, it was found that they have the lowest inhibition percentages against α-amylase (9.12 ± 0.01 and 9.38 ± 0.02%, respectively) and α-glucosidase (7.85 ± 0.01 and 8.07 ± 0.02%, respectively).

2.2.3. The Anti-Alzheimer’s Activity

The anti-Alzheimer’s activities of the pyrazole-based Schiff bases 5af, 6af, and 7af were assessed by determining their inhibitory effects against AChE (Figure 3, see the Supplementary Materials Table S2). It was found that the two pyrazole-based Schiff bases 7f and 5d possessed a higher inhibitory effect on AChE (62.11 ± 0.04 and 62.00 ± 0.04%, respectively) compared to the efficiency of donepezil (70.32 ± 0.04%) at the same concentration. On the contrary, the compounds 5b and 7e exhibited the lowest inhibition percentages against AChE (12.28 ± 0.01 and 12.62 ± 0.01%, respectively).

2.2.4. The Anti-Inflammatory Activity

The anti-inflammatory activities of the pyrazole-based Schiff bases 5af, 6af, and 7af were evaluated by determining their inhibitory effects against protein denaturation and a proteinase enzyme (Table 2, see the Supplementary Materials Figure S1). It was found that the pyrazole-based Schiff bases 5a, 5d, 5e, 5f, 7a, and 7f possessed a higher inhibitory effect on protein denaturation (26.83 ± 0.03, 27.79 ± 0.03, 27.33 ± 0.03, 27.28 ± 0.03, 26.87 ± 0.03, and 27.75 ± 0.03%, respectively) and the activity of the proteinase enzyme (23.33 ± 0.03, 24.17 ± 0.03, 23.76 ± 0.03, 23.72 ± 0.03, 23.37 ± 0.03, and 24.13 ± 0.03%, respectively) compared to the efficiency of diclofenac sodium against both protein denaturation and the proteinase enzyme (49.08 ± 0.01 and 46.11 ± 0.02%, respectively) at equal concentrations. On the contrary, the lowest inhibitory activities were observed with the compounds 5b and 7e, which inhibited both protein denaturation by 7.83 ± 0.01 and 8.05 ± 0.01%, respectively, and the activity of the proteinase enzyme by 6.81 ± 0.01 and 4.81 ± 0.01%, respectively.

2.2.5. Cytotoxic Activity

Based on the other in vitro biological activities, it was found that 5a, 5d, 5e, 5f, 7a, and 7f exhibited the highest antioxidant, scavenging, anti-diabetic, anti-Alzheimer’s, and anti-inflammatory activities compared to the other pyrazole-based Schiff bases. Therefore, the cytotoxicities of these derivatives were evaluated against human lung (A549) and colon (Caco-2) cancer types as well as normal Caucasian fibroblast-like fetal lung (WI-38) cell lines (Table 3; see the Supplementary Materials Tables S3–S5).
It was found that the three pyrazole-based Schiff bases 5d, 5e, and 7a were very active and exhibited the highest cytotoxic activities against the growth of the A549 cell line, resulting in the most significant decreases in cell viability. This was evident from their lower IC50 values (48.61 ± 0.14, 47.74 ± 0.20, and 49.40 ± 0.18 μM, respectively) compared to the other pyrazole-based Schiff bases. Regarding the Caco-2 cell line, the two pyrazole-based Schiff bases 5e and 7a exhibited the highest cytotoxic activities with the lowest IC50 values (40.99 ± 0.20 and 42.42 ± 0.18 μM, respectively). These pyrazole-based Schiff bases showed higher cytotoxic activities compared to doxorubicin, which was used as a reference drug (IC50 = 54.94 ± 0.16 μM).
The WI-38 cell line used to reveal the deleterious effects showed that compounds 5d and 7a demonstrated higher IC50 values (731.72 ± 10.46 and 736.26 ± 7.95 μM, respectively), followed by 5e (IC50 = 648.12 ± 7.57 μM). This indicates the safety of these compounds for normal cells compared to the other pyrazole-based Schiff bases and doxorubicin, which was used as a reference drug (IC50 = 304.94 ± 4.72 μM).
The therapeutic indexes (TIs) of the pyrazole-based Schiff bases 5a, 5d, 5e, 5f, 7a, and 7f were determined to study their safety and efficacy (Table 3). The results demonstrated that all the pyrazole-based Schiff bases (5a, 5d, 5e, 5f, 7a, and 7f) possessed a therapeutic index higher than doxorubicin in the case of Caco-2 cells. However, in the case of A549 cells, all Schiff bases possessed a therapeutic index higher than doxorubicin except for two Schiff bases: 5a and 5f.

2.2.6. The Enzymatic Activity

The efficiencies of six pyrazole-based Schiff bases, 5a, 5d, 5e, 5f, 7a, and 7f, against caspase-3 and Bcl-2 were evaluated (Table 4).
It was observed that the two pyrazole-based Schiff bases 5d and 7a increased the activity of caspase-3 (300.73 ± 0.42 and 322.21 ± 0.45 pg/mL, respectively) while decreasing the Bcl-2 level (2.62 ± 0.01 and 2.81 ± 0.01 ng/mL, respectively) in the treated A549 cells compared to the untreated A549 cells. It showed approximately the same effect induced by doxorubicin on both the caspase-3 and Bcl-2 levels (330.80 ± 0.46 pg/mL and 2.55 ± 0.01 ng/mL, respectively). The pyrazole-based Schiff base 5a showed the lowest anti-proliferative effect, as indicated by the lower caspase-3 level (184.74 ± 0.26 pg/mL) and the higher Bcl-2 level (6.55 ± 0.03 ng/mL).
Regarding the Caco-2 cell lines, it was observed that the two pyrazole-based Schiff bases 5e and 7a elevated the activity of caspase-3 (315.45 ± 0.46 and 363.98 ± 0.54 pg/mL, respectively) while decreasing the Bcl-2 level (2.13 ± 0.02 and 2.46 ± 0.02 ng/mL, respectively) in the treated cells compared to the untreated ones. It showed approximately the same effect induced by doxorubicin on both the caspase-3 and Bcl-2 levels (363.98 ± 0.54 pg/mL and 2.55 ± 0.01 ng/mL, respectively). The pyrazole-based Schiff base 5a showed the lowest anti-proliferative effect, as indicated by the lower caspase-3 (208.68 ± 0.31 pg/mL) and the higher Bcl-2 levels (5.33 ± 0.05 ng/mL).

2.3. Computational Prediction (ADMT Properties)

Six derivatives of pyrazole-based Schiff bases (5a, 5d, 5e, 5f, 7a, and 7f) were analyzed using the pkCSM website https://biosig.lab.uq.edu.au/pkcsm/prediction (accessed on 15 March 2024) to evaluate their properties and safety as potential drug candidates. The data presented in Table 5 showed the absorptions, distributions, metabolisms, and toxicities of these compounds. Also, the ideal values for all properties were presented.
It was observed that the intestinal absorption (human) range of the six derivatives of pyrazole-based Schiff bases (5a, 5d, 5e, 5f, 7a, and 7f) was from 91.357% to 96.711%. The skin permeability value is log Kp = −2.735. The blood–brain permeation barrier (BBB permeability) ranged from −1.818 to −1.318. The central nervous system permeability (CNS permeability) ranged from −2.133 to −1.705. The results of the enzyme inhibition of the six Schiff bases (5a, 5d, 5e, 5f, 7a, and 7f) show that they can be inhibitors or non-inhibitors. The two toxicity endpoints are AMES toxicity and skin sensitization.

3. Discussion

All the pyrazole-based Schiff bases (5af, 6af, and 7af) have been characterized utilizing spectral analyses (1H NMR and 13C NMR spectra) as mentioned in our previous works [35,36]. The 1H NMR spectra of the pyrazole-based Schiff bases 5af, 6af, and 7af were characterized by a single signal in the range of δ from 8.64 to 8.93 ppm. This signal corresponds to an azomethine proton (–N=CH– proton) (See the Supplementary Materials).
The antioxidant activities of the pyrazole-based Schiff bases 5af, 6af, and 7af were assessed at equal concentrations by quantifying their TACs and IRP. The compounds 5a, 5d, 5e, 5f, 7a, and 7f showed higher TACs and IRP, and this finding is consistent with Ali et al. [37], who demonstrated that the antioxidant activity increased in the cyclized heterocyclic compounds due to the presence of a pyrazoline moiety, which has potent antioxidant activity, and the low number of rotatable bonds makes them more favorable compared to the other compounds. In addition, the electron-donating groups (OCH3 and CH3) are more beneficial than unsubstituted or mono chloro-substituted phenyl rings, which might be attributed to the mesomeric effects [38]. The compounds 5b and 7e showed the lowest TACs and IRP, and this might be related to the replacement of the N atom by the O atom, which gives a lower antioxidant activity [39]. During the current study, the scavenging activity was assessed by calculating the IC50 values of DPPH and the inhibition percentages of ABTS radicals. The compounds with a higher antioxidant activity were found to have lower IC50 values of DPPH and a higher inhibition percentage against ABTS radicals [40]. It was found that the ABTS assay is more sensitive than the DPPH assay, because the DPPH radical is only involved in hydrogen (H+) transfer (DPPH to DPPH-H), while the ABTS radical is involved in the electron transfer pathway (ABTS to ABTS+) [41].
The compounds 5a, 5d, 5e, 5f, 7a, and 7f were found to have lower IC50 values for DPPH and higher inhibition percentages against the ABTS radical compared to the other pyrazole-based Schiff bases. This might be attributed to the affinity of these compounds to donate hydrogen free radicals as proposed by Matta et al. (in 2023) [42].
The highest IC50 values for DPPH were noticed with the compounds 5b and 7e, and they showed the lowest inhibition percentages against the ABTS radical. This indicates a lower scavenging activity, which may be due to the presence of only an aromatic group attached to the compound. This lack of significant inhibitory activity towards in vitro antioxidant and scavenging activities suggests that the presence of substituents is important for these activities [43].
Type 2 diabetes mellitus (T2DM) is the most common form of diabetes, leading to the impairment of various physiological processes in the body due to hyperglycemia or abnormal blood glucose levels induced by insulin resistance [44]. The development of new anti-diabetic drugs is related to the inhibition of hydrolase enzymes, showing that the incorporation of pyrazole is required in the design of new anti-hyperglycemic agents with higher activities than acarbose [45]. Therefore, the anti-diabetic activities of the pyrazole-based Schiff bases 5af, 6af, and 7af were assessed in the present study by determining their inhibitory effects against α-amylase and α-glucosidase enzymes and comparing them to the efficiency of acarbose, which is used as a standard drug. In the present study, it was noticed that the pyrazole-based Schiff bases 5a, 5d, 5e, 5f, 7a, and 7f possessed a higher inhibitory effect on α-amylase and α-glucosidase compared to the efficiency of acarbose against α-amylase and α-glucosidase activities at equal concentrations. This may be attributed to the hydrogen bond interaction between the unsubstituted nitrogen atom of the pyrazole ring and the functional groups in the side chains of the amino acids at the active sites, which can also play a role [46]. Pogaku et al. added that pyrazole-based Schiff bases substituted with electron-withdrawing groups on the phenyl ring showed higher inhibitory activities against these hydrolase enzymes than compounds with electron-donating groups [47]. Moreover, these pyrazole-based Schiff bases showed their anti-diabetic activity through the formation of hydrophobic, van der Waals, and hydrogen bond interactions between the nitrogen atom of their pyrazole moiety and the oxygen atom of various amino acids in the active site of these enzymes [48]. Regarding the compounds 5b and 7e, it was found that they have the lowest inhibition percentages against α-amylase and α-glucosidase, and this might be related to their lower antioxidant and scavenging activities [49].
Alzheimer’s disease (AD) is a degenerative disease of the central nervous system characterized by mental deterioration, especially in the elderly [50]. According to “the cholinergic hypothesis”, which means inadequate cholinergic transmission in the synapse, AChE is the main enzyme responsible for acetylcholine (ACh) hydrolysis in the cholinergic synapses, constituting the basis of AD treatment [51]. Many studies have suggested that various therapeutic agents are capable of inhibiting the AChE enzyme and providing additional benefits for the treatment of AD, replacing the commercially available drugs, which only have symptomatic effects. Therefore, recent studies have focused on searching for more effective synthetic compounds to halt the progression of the disease [52].
The current study showed that the two pyrazole-based Schiff bases 7f and 5d possessed a higher inhibitory effect on AChE compared to the efficiency of donepezil at the same concentration, and this might be related to the presence of active substituents, which elicited a higher neuroprotective activity [53]. Coupling the phenyl group to the pyrazole scaffold through an imine linker resulted in the inhibitory activity, which varied with the substituent on the imine nitrogen due to its high affinity to bind to the active site of the AChE enzyme involved in the substrate inhibition characteristics of AChE. Furthermore, the phenyl group placed at this nitrogen was highly favorable for AChE inhibitory activity. Additionally, the addition of a hydroxyl group enhanced the activity [54]. The compounds 5b and 7e exhibited the lowest inhibition percentages against AChE, and this might be related to increasing the length of the alkyl substituent [55].
Arthritis is a chronic and progressive autoimmune disease in which bone and cartilage destruction occur due to chronic proliferative synovitis and synovial inflammation, resulting in significant joint damage and reduced functionality [56]. Protein denaturation is considered one of the predominant reasons for arthritic and inflammatory diseases [57]. Therefore, synthetic compounds such as pyrazole-based Schiff bases that can inhibit both protein denaturation and proteinase enzyme are a potential strategy for arthritic therapy [58,59]. The present study showed that the pyrazole-based Schiff bases 5a, 5d, 5e, 5f, 7a, and 7f possessed a higher inhibitory effect on both protein denaturation and the activity of the proteinase enzyme compared to the efficiency of diclofenac sodium at equal concentrations. This might refer to altering the tertiary as well as secondary structures of proteins, which consequently leads to impairments in the biological functions of most biological proteins. Moreover, the presence of a chemical entity possessing a substituent exerts the highest activity compared with all other compounds due to it binding effectively toward the active site of the proteins [60,61]. Joy et al. proposed that the anti-denaturation activity can be rationalized by the presence of electron-donating groups (–OH and –NH2) and nitrogen-rich fragments, which exhibit a superior anti-inflammatory potential compared to the other pyrazole-based Schiff bases molecules [62]. The lowest inhibitory activities were observed with compounds 5b and 7e, which inhibited both protein denaturation and the activity of the proteinase enzyme. This could possibly be due to the presence of substituents that affected the activities and were found to be the least active in this series [61].
Finally, based on the evaluations of the pyrazole-based Schiff bases 5af, 6af, and 7af, including their antioxidant and scavenging activities, anti-diabetes, anti-Alzheimer’s, and anti-inflammatory properties, it can be deduced that the six pyrazole-based Schiff bases 5a, 5d, 5e, 5f, 7a, and 7f exhibit highly active properties in these evaluations. Therefore, further studies on the cytotoxic and enzymatic activities of these six pyrazole-based Schiff bases are warranted.
Carcinoma is the abnormal growth of normal cells that typically grow beyond their original boundaries, invade surrounding areas, spread to other organs, and result in metastasis, which is one of the main causes of cancer-related deaths [63]. The present study showed that the three pyrazole-based Schiff bases 5d, 5e, and 7a were very active and exhibited the highest cytotoxic activities against the growth of the A549 cell line, resulting in the most significant decreases in cell viability. This finding is consistent with Czylkowska et al., who postulated that pyrazole derivatives are capable of effectively inhibiting the growth of the A549 cell line, possibly due to the introduction of electron-withdrawing substitutions that increase the lipophilicity of the molecule [64]. This enhancement in lipophilicity may improve the cell permeability and overall potency of these compounds [65]. Furthermore, these substituted pyrazole derivatives might be able to arrest the cell cycle at the G1/S phase in treated cells, displaying an accumulation of cells in the G0 phase and an increase in the percentage of cells in both the early and late apoptotic stages [66]. Therefore, these synthetic compounds may hold promise for the treatment of lung cancer. In regard to the Caco-2 cell line, the two pyrazole-based Schiff bases 5e and 7a exhibited the highest cytotoxic activities with the lowest IC50 values, and this may be attributed to the ability of the pyrazoline derivatives, which exhibit cell cycle-arrest properties, to induce cell cycle arrest in the G2/M phase and apoptosis [67]. Additionally, the pyrazole-based Schiff bases may have the ability to bind to DNA through intercalation, similar to doxorubicin [68].
Regarding the WI-38 cell line, the compounds 5d and 7a showed higher IC50 values followed by 5e, indicating the safety of these compounds for normal cells compared to the other pyrazole-based Schiff bases and doxorubicin.
Cysteine proteases, specifically caspases, play a vital role in programmed cell death by coordinating the cascade for degrading cellular components [69]. Caspase-3, a member of the cysteine–aspartate-specific protease family, is a ubiquitous protein in mammalian cells that contributes to apoptosis [70]. It activates the apoptosis pathway in response to a variety of stimuli, including chemotherapeutic compounds [71]. The B-cell lymphoma 2 (Bcl-2) belongs to the Bcl-2 protein family, which exhibits pro- and anti-apoptotic activities and is held in a delicate balance in healthy cells. It can cause cells to irreversibly head toward cell death or, conversely, allow cells to permanently escape apoptosis and become a malignant clone [72].
The present study showed that the two pyrazole-based Schiff bases 5d and 7a increased the activity of caspase-3 while decreasing the Bcl-2 level in the treated A549 cells compared to the untreated A549 cells. The pyrazole-based Schiff base 5a showed the lowest anti-proliferative effect, as indicated by the lower caspase-3 level and the higher Bcl-2 level. Furthermore, the two pyrazole-based Schiff bases 5e and 7a elevated the activity of caspase-3 while decreasing the Bcl-2 level in the treated Caco-2 cells compared to the untreated ones. The pyrazole-based Schiff base 5a showed the lowest anti-proliferative effect, as indicated by the lower caspase-3 and the higher Bcl-2 levels. The overall enzymatic assay showed that the pyrazole-based Schiff base 7a exhibited the highest anti-proliferative effect against both A549 and Caco-2 cell lines, while the pyrazole-based Schiff base 5a showed the lowest effect. The pyrazole-based Schiff bases, especially 7a, showed an anti-proliferative effect on both studied cell lines by stimulating the apoptotic pathway through the upregulation of caspase-3 and downregulation of Bcl-2 [73].
The pyrazole-based Schiff bases (5a, 5d, 5e, 5f, 7a, and 7f) were analyzed using the pkCSM website [74,75], and the data presented in Table 5 demonstrate the following:
-
The absorption results of the pyrazole-based Schiff bases (5a, 5d, 5e, 5f, 7a, and 7f) indicate high values (well-absorbed molecules) ranging from 91.357% to 96.711% for intestinal absorption. Skin permeability (Kp) refers to skin absorption and the rate of drug candidates penetrating the skin. The skin permeability of the Schiff bases indicates low values less than −2.5, therefore, showing good skin permeability (log Kp = −2.735) and the ability to penetrate through the outermost layer of the epidermal skin.
-
The distribution results conclude that (i) the blood–brain permeation barrier (BBB permeability) predicted results indicate that these Schiff bases exhibit a poor distribution, with values lower than −1. The blood–brain barrier (BBB) regulates the permeability of drugs to the brain. A poor distribution refers to impaired drug delivery into the brain. Therefore, the medicinal efficacy of the drugs decreases. (ii) The central nervous system permeability (CNS permeability) indicates that the four Schiff bases (5a, 5d, 5e, and 5f) show high penetration, but two Schiff bases (7a and 7f) show moderate penetration.
-
Drug metabolism is one of the essential factors in drug disposition. The five enzymes play a crucial role in the metabolic processes of drugs in the liver. The results indicated that the six Schiff bases (5a, 5d, 5e, 5f, 7a, and 7f) are non-inhibitors of the CYP2D6 enzyme. Also, three Schiff bases (5e, 5f, and 7f) are non-inhibitors of the CYP1A2 enzyme. Therefore, the Schiff bases are well-metabolized molecules in the liver, can be eliminated from the body, and have no potential adverse effects.
-
The prediction of the toxicity of pyrazole-based Schiff bases (5a, 5d, 5e, 5f, 7a, and 7f) suggests that all Schiff bases are non-mutagenic except 7f. Additionally, none of the Schiff bases (5a, 5d, 5e, 5f, 7a, and 7f) induce skin sensitization. Therefore, these compounds are considered safe.
Almost all Schiff bases possess promising ADMT properties (well absorbed, possess good skin permeability, and well metabolized in the liver), which may be due to the presence of the pyrazole moiety in their structures. The pyrazole motif has promising biological applications [18,19,20,21] and is effective in various disease treatments.
After the in silico ADMT properties study, in the future, we will extend the study to include molecular docking and simulation for caspase-3 and Bcl-2 enzymes.

4. Materials and Methods

4.1. Chemistry

The three sets of pyrazole-based Schiff bases, 5af, 6af, and 7af, were prepared according to the literature procedures mentioned in our previous works. All the pyrazole-based Schiff bases (5af, 6af, and 7af) have been characterized using spectral analyses. These spectral data were mentioned in our previous works [35,36] (see the Supplementary Materials).

4.2. Biological Evaluations

All in vitro biological activities were assessed in the tested compounds at equal concentrations (1000 µg/mL). All assays were carried out in triplicate.

4.2.1. The Antioxidant Activity

The antioxidant and scavenging activities of the pyrazole-based Schiff bases 5af, 6af, and 7af were assessed at equal concentrations according to the reported techniques in the literature [76,77,78,79]

4.2.2. The Anti-Diabetic Activity

The anti-diabetic activities of the pyrazole-based Schiff bases 5af, 6af, and 7af were assessed in the present study by determining their inhibitory effects against α-amylase and α-glucosidase according to the reported techniques in the literature [80,81]

4.2.3. The Anti-Alzheimer’s Activity

The anti-Alzheimer’s activities of the pyrazole-based Schiff bases 5af, 6af, and 7af were assessed by determining their inhibitory effects against acetylcholinesterase (AChE) according to the reported techniques in the literature [82].

4.2.4. The Anti-Inflammatory Activity

The anti-inflammatory activities of the pyrazole-based Schiff bases 5af, 6af, and 7af were evaluated by determining their inhibitory effects against both protein denaturation and proteinase enzyme according to the reported techniques in the literature [83,84,85].

4.2.5. Cytotoxic Activity

The cytotoxicity of six pyrazole-based Schiff bases, 5a, 5d, 5e, 5f, 7a, and 7f, against human lung (A549) and colon (Caco-2) cancer types, as well as normal lung (WI-38) cell lines, was evaluated according to the reported techniques in the literature [86]. The therapeutic indexes (TIs) of the compounds were calculated according to the following equation:
Therapeutic index (TI) = IC50 on the normal cells/IC50 on the cancer cells

4.2.6. The Enzymatic Activity

The enzymatic activities of six pyrazole-based Schiff bases, 5a, 5d, 5e, 5f, 7a, and 7f, against caspase-3 and Bcl-2 were evaluated according to the reported techniques in the literature [87,88].

4.2.7. Statistical Analysis

The data were calculated from three replicates and presented as the mean ± SE.
The detailed methods for the in vitro biological activities have been added to the Supplementary Materials.

5. Conclusions

In summary, we re-synthesized three sets of pyrazole-based Schiff bases, 5af, 6af, and 7af, to evaluate their biological applications. The results of the biological evaluations of the pyrazole-based Schiff bases 5af, 6af, and 7af demonstrated that the six pyrazole-based Schiff bases, 5a, 5d, 5e, 5f, 7a, and 7f, possessed the highest biological properties among the compounds evaluated and acted as multi-target agents against diabetes, Alzheimer’s, and inflammatory diseases. As a result, the biological evaluation was extended, namely the cytotoxic and enzymatic activities of the six pyrazole-based Schiff bases 5a, 5d, 5e, 5f, 7a, and 7f. The cytotoxicity study showed that the three Schiff bases 5d, 5e, and 7a were very active against lung (A549) cells, with IC50 values of 48.61 ± 0.14, 47.74 ± 0.20, and 49.40 ± 0.18 μM, respectively. Concerning the colon (Caco-2) cells, the two Schiff bases 5e and 7a exhibited the highest cytotoxicity, with IC50 values of 40.99 ± 0.20 and 42.42 ± 0.18 μM, respectively. The safety and efficacy study showed that all pyrazole-based Schiff bases (5a, 5d, 5e, 5f, 7a, and 7f) possessed a therapeutic index higher than doxorubicin. The enzymatic activity study indicated that some Schiff bases affected the levels of caspase-3 and Bcl-2 in both A549 and Caco-2 cells. Additionally, we assessed the in silico ADMT properties of the pyrazole-based Schiff bases 5a, 5d, 5e, 5f, 7a, and 7f. The results demonstrate that Schiff bases are well absorbed, have good skin permeability, poor distribution through the BBB permeability, high or moderate penetration through CNS permeability, are non-inhibitors of the CYP2D6 enzyme, are non-mutagenic except for 7f, and do not induce skin sensitization. Currently, in our laboratories, with teams of colleagues, we are conducting further biochemistry, medicinal chemistry, molecular docking, and simulation studies on the six pyrazole-based Schiff bases 5a, 5d, 5e, 5f, 7a, and 7f after modifying their structures. In the future, we will report the results.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ph17050655/s1, Table S1: The anti-diabetic activity of pyrazole-based Schiff bases 5af, 6af, and 7af; Table S2: The anti-Alzheimer activity of pyrazole-based Schiff bases 5af, 6af, and 7af; Figure S1: The anti-inflammatory activity of pyrazole-based Schiff bases 5af, 6af, 7af, and diclofenac sodium as a standard drug; Table S3: Cytotoxic activity of the six pyrazole-based Schiff bases 5a, 5d, 5e, 5f, 7a, and 7f against human lung cancer (A549) cell line compared to Doxorubicin as a standard drug; Table S4: Cytotoxic activity of the six pyrazole-based Schiff bases 5a, 5d, 5e, 5f, 7a, and 7f against human colon cancer (Caco-2) cell line compared to Doxorubicin as a standard drug; Table S5: Cytotoxic activity of the six pyrazole-based Schiff bases 5a, 5d, 5e, 5f, 7a, and 7f against normal lung (WI-38) cell line compared to Doxorubicin as a standard drug.

Author Contributions

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

Funding

This research was funded by the Deanship of Scientific Research at King Saud University through Vice Deanship of Scientific Research Chairs; (Drug Exploration and Development Chair), and the Deanship of Scientific Research at Princess Nourah Bint Abdulrahman University through Researchers Supporting Project number: (PNURSP2024R35).

Institutional Review Board Statement

The experimental design involving human cancer cell lines was conducted in accordance with the protocol approved by the Medical Research Ethics Committee of the National Research Centre, located in Dokki, Cairo, Egypt (Approval No: EX-09461223).

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article.

Acknowledgments

The authors extend their appreciation to the Deanship of Scientific Research, King Saud University for funding through Vice Deanship of Scientific Research Chairs; (Drug Exploration and Development Chair). Also, the authors are grateful to Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia for funding this work through Researchers Supporting Project number: (PNURSP2024R35).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Artasensi, A.; Pedretti, A.; Vistoli, G.; Fumagalli, L. Type 2 Diabetes Mellitus: A Review of Multi-Target Drugs. Molecules 2020, 25, 1987. [Google Scholar] [CrossRef] [PubMed]
  2. Ramsay, R.R.; Popovic-Nikolic, M.R.; Nikolic, K.; Uliassi, E.; Bolognesi, M.L. A perspective on multi-target drug discovery and design for complex diseases. Clin. Transl. Med. 2018, 7, 3. [Google Scholar] [CrossRef] [PubMed]
  3. Rohm, T.V.; Meier, D.T.; Olefsky, J.M.; Donath, M.Y. Inflammation in obesity, diabetes, and related disorders. Immunity 2022, 55, 31–55. [Google Scholar] [CrossRef] [PubMed]
  4. Leszek, J.; Mikhaylenko, E.V.; Belousov, D.M.; Koutsouraki, E.; Szczechowiak, K.; Kobusiak-Prokopowicz, M.; Mysiak, A.; Diniz, B.S.; Somasundaram, S.G.; Kirkland, C.E.; et al. The links between cardiovascular diseases and Alzheimer’s disease. Curr. Neuropharmacol. 2021, 19, 152–169. [Google Scholar] [CrossRef] [PubMed]
  5. Teles, F.; Collman, R.G.; Mominkhan, D.; Wang, Y. Viruses, periodontitis, and comorbidities. Periodontology 2000, 89, 190–206. [Google Scholar] [CrossRef] [PubMed]
  6. Al-Wahaibi, L.H.; Mohammed, A.F.; Abdelrahman, M.H.; Trembleau, L.; Youssif, B.G. Design, Synthesis, and Biological Evaluation of Indole-2-carboxamides as Potential Multi-Target Antiproliferative Agents. Pharmaceuticals 2023, 16, 1039. [Google Scholar] [CrossRef] [PubMed]
  7. Loganathan, V.; Ahamed, A.; Radhakrishnan, S.; Gaafar, A.R.Z.; Gurusamy, R.; Akbar, I. Synthesis of anthraquinone-connected coumarin derivatives via grindstone method and their evaluation of antibacterial, antioxidant, tyrosinase inhibitory activities with molecular docking, and DFT calculation studies. Heliyon 2024, 10, e25168. [Google Scholar] [CrossRef]
  8. Almehizia, A.A.; Aboulthana, W.M.; Naglah, A.M.; Hassan, A.S. In vitro biological studies and computational prediction-based analyses of pyrazolo[1,5-a]pyrimidine derivatives. RSC Adv. 2024, 14, 8397–8408. [Google Scholar] [CrossRef] [PubMed]
  9. Thakur, S.; Bhalla, A. Sustainable synthetic endeavors of pharmaceutically active Schiff bases and their metal complexes: A review on recent reports. Tetrahedron 2024, 153, 133836. [Google Scholar] [CrossRef]
  10. Khan, E.; Hanif, M.; Akhtar, M.S. Schiff bases and their metal complexes with biologically compatible metal ions; biological importance, recent trends and future hopes. Rev. Inorg. Chem. 2022, 42, 307–325. [Google Scholar] [CrossRef]
  11. Tsacheva, I.; Todorova, Z.; Momekova, D.; Momekov, G.; Koseva, N. Pharmacological activities of Schiff bases and their derivatives with low and high molecular phosphonates. Pharmaceuticals 2023, 16, 938. [Google Scholar] [CrossRef] [PubMed]
  12. Mushtaq, I.; Ahmad, M.; Saleem, M.; Ahmed, A. Pharmaceutical significance of Schiff bases: An overview. Future J. Pharm. Sci. 2024, 10, 16. [Google Scholar] [CrossRef]
  13. Mukhtar, S.S.; Hassan, A.S.; Morsy, N.M.; Hafez, T.S.; Hassaneen, H.M.; Saleh, F.M. Overview on synthesis, reactions, applications, and biological activities of Schiff bases. Egypt. J. Chem. 2021, 64, 6541–6554. [Google Scholar] [CrossRef]
  14. Çavuş, M.S.; Yakan, H.; Başkan, C.; Muğlu, H.; Babacan, A.A. Schiff bases based on thio/carbohydrazide: Synthesis, spectroscopic characterization, DFT, antimicrobial, DNA interactions and cytotoxicity studies. J. Mol. Struct. 2024, 1304, 137655. [Google Scholar] [CrossRef]
  15. Abdel-Baky, Y.M.; Omer, A.M.; El-Fakharany, E.M.; Ammar, Y.A.; Abusaif, M.S.; Ragab, A. Developing a new multi-featured chitosan-quinoline Schiff base with potent antibacterial, antioxidant, and antidiabetic activities: Design and molecular modeling simulation. Sci. Rep. 2023, 13, 22792. [Google Scholar] [CrossRef] [PubMed]
  16. Koçyiğit, Ü.M.; Gezegen, H.; Taslimi, P. Synthesis, characterization, and biological studies of chalcone derivatives containing Schiff bases: Synthetic derivatives for the treatment of epilepsy and Alzheimer’s disease. Arch. Pharm. 2020, 353, 2000202. [Google Scholar] [CrossRef] [PubMed]
  17. Hanif, M.; Hassan, M.; Rafiq, M.; Abbas, Q.; Ishaq, A.; Shahzadi, S.; Seo, S.Y.; Saleem, M. Microwave-assisted synthesis, in vivo anti-inflammatory and in vitro anti-oxidant activities, and molecular docking study of new substituted Schiff base derivatives. Pharm. Chem. J. 2018, 52, 424–437. [Google Scholar] [CrossRef]
  18. Aziz, H.; Zahoor, A.F.; Ahmad, S. Pyrazole bearing molecules as bioactive scaffolds: A review. J. Chil. Chem. Soc. 2020, 65, 4746–4753. [Google Scholar] [CrossRef]
  19. Brullo, C.; Rapetti, F.; Bruno, O. Pyrazolyl-ureas as interesting scaffold in medicinal chemistry. Molecules 2020, 25, 3457. [Google Scholar] [CrossRef]
  20. Alam, M.A. Pyrazole: An emerging privileged scaffold in drug discovery. Future Med. Chem. 2023, 15, 2011–2023. [Google Scholar] [CrossRef]
  21. Bastos, I.M.; Rebelo, S.; Silva, V.L. A review of poly (ADP-ribose) polymerase-1 (PARP1) role and its inhibitors bearing pyrazole or indazole core for cancer therapy. Biochem. Pharmacol. 2024, 221, 116045. [Google Scholar] [CrossRef] [PubMed]
  22. Mortada, S.; Karrouchi, K.; Hamza, E.H.; Oulmidi, A.; Bhat, M.A.; Mamad, H.; Aalilou, Y.; Radi, S.; Ansar, M.H.; Masrar, A.; et al. Synthesis, structural characterizations, in vitro biological evaluation and computational investigations of pyrazole derivatives as potential antidiabetic and antioxidant agents. Sci. Rep. 2024, 14, 1312. [Google Scholar] [CrossRef] [PubMed]
  23. Alkahtani, H.M.; Almehizia, A.A.; Al-Omar, M.A.; Obaidullah, A.J.; Zen, A.A.; Hassan, A.S.; Aboulthana, W.M. In Vitro Evaluation and Bioinformatics Analysis of Schiff Bases Bearing Pyrazole Scaffold as Bioactive Agents: Antioxidant, Anti-Diabetic, Anti-Alzheimer, and Anti-Arthritic. Molecules 2023, 28, 7125. [Google Scholar] [CrossRef]
  24. Hassan, A.S.; Morsy, N.M.; Aboulthana, W.M.; Ragab, A. Exploring novel derivatives of isatin-based Schiff bases as multi-target agents: Design, synthesis, in vitro biological evaluation, and in silico ADMET analysis with molecular modeling simulations. RSC Adv. 2023, 13, 9281–9303. [Google Scholar] [CrossRef] [PubMed]
  25. Abdelazeem, N.M.; Aboulthana, W.M.; Hassan, A.S.; Almehizia, A.A.; Naglah, A.M.; Alkahtani, H.M. Synthesis, in silico ADMET prediction analysis, and pharmacological evaluation of sulfonamide derivatives tethered with pyrazole or pyridine as anti-diabetic and anti-Alzheimer’s agents. Saudi Pharm. J. 2024, 23, 102025. [Google Scholar] [CrossRef]
  26. Elgiushy, H.R.; Mohamed, S.H.; Taha, H.; Sawaf, H.; Hassan, Z.; Abou-Taleb, N.A.; El-Labbad, E.M.; Hassan, A.S.; Abouzid, K.A.; Hammad, S.F. Identification of a promising hit from a new series of pyrazolo[1,5-a]pyrimidine based compounds as a potential anticancer agent with potent CDK1 inhibitory and pro-apoptotic properties through a multistep in vitro assessment. Bioorg. Chem. 2022, 120, 105646. [Google Scholar] [CrossRef] [PubMed]
  27. Morsy, N.M.; Hassan, A.S.; Hafez, T.S.; Mahran, M.R.; Sadawe, I.A.; Gbaj, A.M. Synthesis, antitumor activity, enzyme assay, DNA binding and molecular docking of Bis-Schiff bases of pyrazoles. J. Iran. Chem. Soc. 2021, 18, 47–59. [Google Scholar] [CrossRef]
  28. Hassan, A.S.; Osman, S.A.; Hafez, T.S. 5-Phenyl-2-furaldehyde: Synthesis, reactions and biological activities. Egypt. J. Chem. 2015, 58, 113–139. [Google Scholar]
  29. Hassan, A.S. Mixed isatin with 3-(2-(aryl)hydrazono)acetylacetone Mn(II), Co(II) and Ni(II) complexes: Antibacterial evaluation and molecular properties prediction. Bull. Chem. Soc. Ethiop. 2020, 34, 533–541. [Google Scholar] [CrossRef]
  30. Abdelghany, A.M.; Khatab, T.K.; Hassan, A.S. Copper-based glass-ceramic as an efficient catalyst in the synthesis of pyrazolo[1,5-a]pyrimidine under solvent-free condition with docking validation as COVID-19 main protease (Mpro) inhibitor. Bull. Chem. Soc. Ethiop. 2021, 35, 185–196. [Google Scholar] [CrossRef]
  31. Khatab, T.K.; Hassan, A.S.; Hafez, T.S. V2O5/SiO2 as an efficient catalyst in the synthesis of 5-aminopyrazole derivatives under solvent free condition. Bull. Chem. Soc. Ethiop. 2019, 33, 135–142. [Google Scholar] [CrossRef]
  32. Hassan, A.S.; Hafez, T.S.; Osman, S.A. Synthesis, characterization, and cytotoxicity of some new 5-aminopyrazole and pyrazolo[1,5-a]pyrimidine derivatives. Sci. Pharm. 2015, 83, 27–39. [Google Scholar] [CrossRef] [PubMed]
  33. Soliman, D.H.; Eldehna, W.M.; Ghabbour, H.A.; Kabil, M.M.; Abdel-Aziz, M.M.; Abdel-Aziz, H.A. Novel 6-phenylnicotinohydrazide derivatives: Design, synthesis and biological evaluation as a novel class of antitubercular and antimicrobial agents. Biol. Pharm. Bull. 2017, 40, 1883–1893. [Google Scholar] [CrossRef] [PubMed]
  34. Xu, C.-J.; Shi, Y.-Q. Synthesis and crystal structure of 5-chloro-3-methyl-1-phenyl-1H-pyrazole-4-carbaldehyde. J. Chem. Crystallogr. 2011, 41, 1816–1819. [Google Scholar] [CrossRef]
  35. Mukhtar, S.S.; Hassan, A.S.; Morsy, N.M.; Hafez, T.S.; Saleh, F.M.; Hassaneen, H.M. Design, synthesis, molecular prediction and biological evaluation of pyrazole-azomethine conjugates as antimicrobial agents. Synth. Commun. 2021, 51, 1564–1580. [Google Scholar] [CrossRef]
  36. Hassan, A.S.; Askar, A.A.; Naglah, A.M.; Almehizia, A.A.; Ragab, A. Discovery of new Schiff bases tethered pyrazole moiety: Design, synthesis, biological evaluation, and molecular docking study as dual targeting DHFR/DNA gyrase inhibitors with immunomodulatory activity. Molecules 2020, 25, 2593. [Google Scholar] [CrossRef]
  37. Ali, S.A.; Awad, S.M.; Said, A.M.; Mahgoub, S.; Taha, H.; Ahmed, N.M. Design, synthesis, molecular modelling and biological evaluation of novel 3-(2-naphthyl)-1-phenyl-1H-pyrazole derivatives as potent antioxidants and 15-Lipoxygenase inhibitors. J. Enzyme Inhib. Med. Chem. 2020, 35, 847–863. [Google Scholar] [CrossRef] [PubMed]
  38. Lavanya, G.; Prakash, T.B.; Sravya, G.; Padmavathi, V.; Padmaja, A. Synthesis and antioxidant activity of bis unsaturated sulfones, bispyrroles, and bispyrazoles. Res. Chem. Interm. 2015, 41, 8815–8828. [Google Scholar] [CrossRef]
  39. Achutha, D.K.; Kameshwar, V.H.; Ningappa, M.B.; Kariyappa, A.K. Synthesis and in vitro biological evaluation for antioxidant, anti-inflammatory activities and molecular docking studies of novel pyrazole derivatives. Biointer. Res. Appl. Chem. 2017, 7, 2040–2047. [Google Scholar]
  40. Hassan, A.S.; Aboulthana, W.M. Synthesis, In Vitro Biological Investigation, and In Silico Analysis of Pyrazole-Based Derivatives as Multi-target Agents. Egypt. J. Chem. 2023, 66, 441–455. [Google Scholar] [CrossRef]
  41. Zhong, G.; Shen, J.; Chen, Z.; Lin, Z.; Long, L.; Wu, J.; Long, C.; Huang, S.; Lian, P.; Luo, G. Antioxidant and Antitumor Activities of Newly Synthesized Hesperetin Derivatives. Molecules 2022, 27, 879. [Google Scholar] [CrossRef] [PubMed]
  42. Matta, R.; Pochampally, J.; Dhoddi, B.N.; Bhookya, S.; Bitla, S.; Akkiraju, A.G. Synthesis, antimicrobial and antioxidant activity of triazole, pyrazole containing thiazole derivatives and molecular docking studies on COVID-19. BMC Chem. 2023, 17, 61. [Google Scholar] [CrossRef]
  43. Shankar, B.; Jalapathi, P.; Anil, V.; Kumar, K.; Saikrishna, B.; Karunakarrao, K. Synthesis and biological evaluation of new 2-(6-alkyl-pyrazin-2-yl)-1H-benz[d]imidazoles as potent anti-inflammatory and anti-oxidant agents. Med. Chem. Res. 2017, 26, 1835–1846. [Google Scholar] [CrossRef]
  44. Nikookar, H.; Khanaposhtani, M.M.; Imanparast, S.; Faramarzi, M.A.; Ranjbar, P.R.; Mahdavi, M.; Larijani, B. Design, synthesis and in vitro α-glucosidase inhibition of novel dihydropyrano[3,2-c]quinoline derivatives as potential anti-diabetic agents. Bioorg. Chem. 2018, 77, 280–286. [Google Scholar] [CrossRef] [PubMed]
  45. Ozil, M.; Emirik, M.; Belduz, A.; Ulker, S. Molecular docking studies and synthesis of novel bis benzimidazole derivatives as inhibitors of α-glucosidase. Bioorg. Med. Chem. 2016, 24, 5103–5114. [Google Scholar] [CrossRef] [PubMed]
  46. Chaudhry, F.; Naureen, S.; Ashraf, M.; Al-Rashida, M.; Jahan, B.; Munawar, M.A.; Khan, M.A. Imidazole-pyrazole hybrids: Synthesis, characterization and in-vitro bio evaluation against α-glucosidase enzyme with molecular docking studies. Bioorg. Chem. 2019, 82, 267–273. [Google Scholar] [CrossRef]
  47. Pogaku, V.; Gangarapu, K.; Basavoju, S.; Tatapudic, K.K.; Katragadda, S.B. Design, synthesis, molecular modelling, ADME prediction and antihyperglycemic evaluation of new pyrazole-triazolo pyrimidine hybrids as potent α-glucosidase inhibitors. Bioorg. Chem. 2019, 93, 103307. [Google Scholar] [CrossRef]
  48. Duhan, M.; Singh, R.; Devi, M.; Sindhu, J.; Bhatia, R.; Kumar, A.; Kumar, P. Synthesis, molecular docking and QSAR study of thiazole clubbed pyrazole hybrid as α-amylase inhibitor. J. Biomol. Struct. Dyn. 2019, 39, 91–107. [Google Scholar] [CrossRef]
  49. Pillai, R.R.; Karrouchi, K.; Fettach, S.; Armaković, S.; Armaković, S.J.; Brik, Y.; Taoufik, J.; Radi, S.; Faouzi, M.E.; Ansar, M. Synthesis, spectroscopic characterization, reactive properties by DFT calculations, molecular dynamics simulations and biological evaluation of Schiff bases tethered 1,2,4-triazole and pyrazole rings. J. Mol. Struct. 2019, 1177, 47–54. [Google Scholar] [CrossRef]
  50. Tok, F.; Koçyiğit-Kaymakçıoğlu, B.; Sağlık, B.N.; Levent, S.; Özkay, Y.; Kaplancıklı, Z.A. Synthesis and biological evaluation of new pyrazolone Schiff bases as monoamine oxidase and cholinesterase inhibitors. Bioorg. Chem. 2019, 84, 41–50. [Google Scholar] [CrossRef]
  51. Alam, M.S.; Ahmed, J.U. Synthesis, crystal structure, biological evaluation, in silico ADME properties, enzymatic target prediction and molecular docking studies of pyrazolone-azomethine analogs. J. Mol. Struct. 2023, 1294, 136504. [Google Scholar] [CrossRef]
  52. Abou-Zied, H.A.; Beshr, E.A.M.; Hayallah, A.M.; Abdel-Aziz, M. Emerging insights into pyrazoline motifs: A comprehensive exploration of biological mechanisms and prospects for future advancements. J. Mol. Struct. 2024, 1296, 136807. [Google Scholar] [CrossRef]
  53. Gedawy, E.M.; Kassab, A.E.; El Kerdawy, A.M. Design, Synthesis and Biological Evaluation of Novel Pyrazole Sulfonamide Derivatives as Dual COX-2/5-LOX Inhibitors. Eur. J. Med. Chem. 2020, 189, 112066. [Google Scholar] [CrossRef]
  54. Pohanka, M. Inhibitors of acetylcholinesterase and butyrylcholinesterase meet immunity. Int. J. Mol. Sci. 2014, 15, 9809–9825. [Google Scholar] [CrossRef]
  55. Kumar, A.; Jain, S.; Parle, M.; Jain, N.; Kumar, P. 3-Aryl-1-phenyl-1H-pyrazole derivatives as new multi target directed ligands for the treatment of Alzheimer’s disease, with acetylcholinesterase and monoamine oxidase inhibitory properties. EXCLI J. 2013, 12, 1030–1042. [Google Scholar] [PubMed]
  56. Jeon, H.; Yoon, W.J.; Ham, Y.M.; Yoon, S.A.; Kang, S.C. Anti-arthritis effect through the anti-inflammatory effect of Sargassummuticum extract in collagen-induced arthritic (CIA) mice. Molecules 2019, 24, 276. [Google Scholar] [CrossRef]
  57. Hossain, M.M.; Kabir, M.S.H.; Hasanat, A.; Kabir, M.I.; Chowdhury, T.A.; Kibria, A.S.M.G. Investigation of in vitro anti-arthritic and membrane stabilizing activity of ethanol extracts of three Bangladeshi plants. Pharm. Innov. J. 2015, 4, 76–80. [Google Scholar]
  58. Hassan, A.S.; Morsy, N.M.; Aboulthana, W.M.; Ragab, A. In vitro enzymatic evaluation of some pyrazolo[1,5-a]pyrimidine derivatives: Design, synthesis, antioxidant, anti-diabetic, anti-Alzheimer, and anti-arthritic activities with molecular modeling simulation. Drug Dev. Res. 2023, 84, 3–24. [Google Scholar] [CrossRef] [PubMed]
  59. Ayman, R.; Radwan, A.M.; Elmetwally, A.M.; Ammar, Y.A.; Ragab, A. Discovery of novel pyrazole and pyrazolo[1,5-a]pyrimidine derivatives as cyclooxygenase inhibitors (COX-1 and COX-2) using molecular modeling simulation. Arch. Pharm. 2023, 356, e2200395. [Google Scholar] [CrossRef]
  60. Sivaramakarthikeyan, R.; Iniyaval, S.; Saravanan, V.; Lim, W.M.; Mai, C.W.; Ramalingan, C. Molecular Hybrids Integrated with Benzimidazole and Pyrazole Structural Motifs: Design, Synthesis, Biological Evaluation, and Molecular Docking Studies. ACS Omega 2020, 5, 10089–10098. [Google Scholar] [CrossRef]
  61. Arya, C.G.; Gondru, R.; Li, Y.; Banothu, J. Coumarin–benzimidazole hybrids: A review of developments in medicinal chemistry. Eur. J. Med. Chem. 2022, 227, 113921. [Google Scholar]
  62. Joy, M.N.; Guda, M.R.; Zyryanov, G.V. Evaluation of Anti-Inflammatory and Anti-Tubercular Activity of 4-Methyl-7-Substituted Coumarin Hybrids and Their Structure Activity Relationships. Pharmaceuticals 2023, 16, 1326. [Google Scholar] [CrossRef] [PubMed]
  63. Angre, T.; Kumar, A.; Singh, A.K.; Thareja, S.; Kumar, P. Role of collagen regulators in cancer treatment: A comprehensive review. Anti-Cancer Agents Med. Chem. 2022, 22, 2956–2984. [Google Scholar] [CrossRef] [PubMed]
  64. Czylkowska, A.; Szczesio, M.; Raducka, A.; Rogalewicz, B.; Kręcisz, P.; Czarnecka, K.; Szymański, P.; Pitucha, M.; Pawlak, T. Cytotoxic Activity against A549 Human Lung Cancer Cells and ADMET Analysis of New Pyrazole Derivatives. Int. J. Mol. Sci. 2021, 22, 6692. [Google Scholar] [CrossRef] [PubMed]
  65. Nagender, P.; Kumar, R.N.; Reddy, G.M.; Swaroop, D.K.; Poornachandra, Y.; Kumar, C.G.; Narsaiah, B. Synthesis of novel hydrazone and azole functionalized pyrazolo [3, 4-b]pyridine derivatives as promising anticancer agents. Bioorg. Med. Chem. Lett. 2016, 26, 4427–4432. [Google Scholar] [CrossRef] [PubMed]
  66. Halim, P.A.; Sharkawi, S.M.Z.; Labib, M.B. Novel pyrazole-based COX-2 inhibitors as potential anticancer agents: Design, synthesis, cytotoxic effect against resistant cancer cells, cell cycle arrest, apoptosis induction and dual EGFR/Topo-1 inhibition. Bioorg. Chem. 2023, 131, 106273. [Google Scholar] [CrossRef] [PubMed]
  67. Fayed, E.A.; Sabour, R.; Harras, M.F.; Mehany, A. Design, synthesis, biological evaluation and molecular modeling of new coumarin derivatives as potent anticancer agents. Med. Chem. Res. 2019, 28, 1284–1297. [Google Scholar] [CrossRef]
  68. El-Gohary, N.; Gabr, M.; Shaaban, M. Synthesis, molecular modeling and biological evaluation of new pyrazolo[3,4-b]pyridine analogs as potential antimicrobial, antiquorum-sensing and anticancer agents. Bioorg. Chem. 2019, 89, 102976. [Google Scholar] [CrossRef]
  69. Cao, A.L.; Tang, Q.F.; Zhou, W.C.; Qiu, Y.Y.; Hu, S.J.; Yin, P.H. Ras/ERK signaling pathway is involved in curcumin-induced cell cycle arrest and apoptosis in human gastric carcinoma AGS cells. J. Asian Nat. Prod. Res. 2015, 17, 56–63. [Google Scholar] [CrossRef]
  70. Harras, M.F.; Sabour, R. Design, synthesis and biological evaluation of novel 1,3,4-trisubstituted pyrazole derivatives as potential chemotherapeutic agents for hepatocellular carcinoma. Bioorg. Chem. 2018, 78, 149–157. [Google Scholar] [CrossRef]
  71. Lepiarczyk, M.; Kałuża, Z.; Bielawska, A.; Czarnomysy, R.; Gornowicz, A.; Bielawski, K. Cytotoxic activity of octahydro pyrazin[2,1-a:5,4-a’]diisoquinoline derivatives in human breast cancer cells. Arch. Pharm. Res. 2015, 38, 628–641. [Google Scholar] [CrossRef]
  72. Carneiro, B.A.; El-Deiry, W.S. Targeting apoptosis in cancer therapy. Nat. Rev. Clin. Oncol. 2020, 17, 395–417. [Google Scholar] [CrossRef]
  73. Hassan, A.S.; Moustafa, G.O.; Awad, H.M.; Nossier, E.S.; Mady, M.F. Design, Synthesis, Anticancer Evaluation, Enzymatic Assays, and a Molecular Modeling Study of Novel Pyrazole−Indole Hybrids. ACS Omega 2021, 6, 12361–12374. [Google Scholar] [CrossRef]
  74. Badraoui, R.; Rebai, T.; Elkahoui, S.; Alreshidi, M.; Veettil, N.V.; Noumi, E.; Al-Motair, A.K.; Aouadi, K.; Kadri, A.; De Feo, V.; et al. Allium subhirsutum L. as a Potential Source of Antioxidant and Anticancer Bioactive Molecules: HR-LCMS Phytochemical Profiling, In Vitro and In Vivo Pharmacological Study. Antioxidants 2020, 9, 1003. [Google Scholar] [CrossRef]
  75. Hassan, A.S. Antimicrobial evaluation, in silico ADMET prediction, molecular docking, and molecular electrostatic potential of pyrazole-isatin and pyrazole-indole hybrid molecules. J. Iran. Chem. Soc. 2022, 19, 3577–3589. [Google Scholar] [CrossRef]
  76. Prieto, P.; Pineda, M.; Aguilar, M. Spectrophotometric Quantitation of Antioxidant Capacity through the Formation of a Phosphomolybdenum Complex: Specific Application to the Determination of Vitamin E. Anal. Biochem. 1999, 269, 337–341. [Google Scholar] [CrossRef]
  77. Oyaizu, M. Studies on products of browning reaction antioxidative activities of products of browning reaction prepared from glucosamine. Jpn. J. Nutr. Diet. 1986, 44, 307–315. [Google Scholar] [CrossRef]
  78. Rahman, M.M.; Islam, M.B.; Biswas, M.; Khurshid Alam, A.H.M. In vitro antioxidant and free radical scavenging activity of different parts of Tabebuia pallida growing in Bangladesh. BMC Res. Notes 2015, 8, 621. [Google Scholar] [CrossRef]
  79. Arnao, M.B.; Cano, A.; Acosta, M. The hydrophilic and lipophilic contribution to total antioxidant activity. Food Chem. 2001, 73, 239–244. [Google Scholar] [CrossRef]
  80. Wickramaratne, M.N.; Punchihewa, J.; Wickramaratne, D. In-vitro alpha amylase inhibitory activity of the leaf extracts of Adenanthera pavonina. BMC Complement Altern. Med. 2016, 16, 466. [Google Scholar] [CrossRef]
  81. Pistia-Brueggeman, G.; Hollingsworth, R.I. A preparation and screening strategy for glycosidase inhibitors. Tetrahedron 2001, 57, 8773–8778. [Google Scholar] [CrossRef]
  82. Ellman, G.L.; Courtney, K.D.; Andres, V.J.; Featherstone, R.M. A new and rapid colorimetric determination of acetylcholinesterase activity. Biochem. Pharmacol. 1961, 7, 88–95. [Google Scholar] [CrossRef]
  83. Das, S.; Sureshkumar, P. Effect of methanolic root extract of Blepharispermum subsesssile DC in controlling arthritic activity. Res. J. Biotechnol. 2016, 11, 65–74. [Google Scholar]
  84. Oyedapo, O.O.; Famurewa, A.J. Antiprotease and Membrane Stabilizing Activities of Extracts of Fagara Zanthoxyloides, Olax Subscorpioides and Tetrapleura Tetraptera. Int. J. Pharmacogn. 1995, 33, 65–69. [Google Scholar] [CrossRef]
  85. Meera, S.; Ramaiah, N.; Kalidindi, N. Illustration of anti-rheumatic mechanism of rheumavedic capsule. Saudi Pharm. J. 2011, 19, 279–284. [Google Scholar] [CrossRef]
  86. Vichai, V.; Kirtikara, K. Sulforhodamine B colorimetric assay for cytotoxicity screening. Nat. Protoc. 2006, 1, 1112–1116. [Google Scholar] [CrossRef]
  87. Hassan, A.S.; Awad, H.M.; Magd-El-Din, A.A.; Hafez, T.S. Synthesis and in vitro antitumor evaluation of novel Schiff bases. Med. Chem. Res. 2018, 27, 915–927. [Google Scholar] [CrossRef]
  88. Pandey, P.; Khan, F.; Alzahrani, F.A.; Qari, H.A.; Oves, M. A Novel Approach to Unraveling the Apoptotic Potential of Rutin (Bioflavonoid) via Targeting Jab1 in Cervical Cancer Cells. Molecules 2021, 26, 5529. [Google Scholar] [CrossRef]
Figure 1. Bioactive derivatives of Schiff bases (AD) and pyrazole scaffold (EG).
Figure 1. Bioactive derivatives of Schiff bases (AD) and pyrazole scaffold (EG).
Pharmaceuticals 17 00655 g001
Scheme 1. Synthesis of pyrazole-based Schiff bases 5af, 6af, and 7af.
Scheme 1. Synthesis of pyrazole-based Schiff bases 5af, 6af, and 7af.
Pharmaceuticals 17 00655 sch001
Figure 2. The anti-diabetic activities of pyrazole-based Schiff bases 5af, 6af, 7af, and acarbose (anti-diabetic standard).
Figure 2. The anti-diabetic activities of pyrazole-based Schiff bases 5af, 6af, 7af, and acarbose (anti-diabetic standard).
Pharmaceuticals 17 00655 g002
Figure 3. The anti-Alzheimer’s activities of pyrazole-based Schiff bases 5af, 6af, 7af, and donepezil (anti-Alzheimer’s standard).
Figure 3. The anti-Alzheimer’s activities of pyrazole-based Schiff bases 5af, 6af, 7af, and donepezil (anti-Alzheimer’s standard).
Pharmaceuticals 17 00655 g003
Table 1. Antioxidant and scavenging activities of pyrazole-based Schiff bases 5af, 6af, and 7af.
Table 1. Antioxidant and scavenging activities of pyrazole-based Schiff bases 5af, 6af, and 7af.
Pyrazole-Based Schiff BasesAntioxidant ActivityScavenging Activity
Total Antioxidant Capacity
(TAC, mg Gallic Acid/g)
Iron-Reducing Power
(IRP, µg/mL)
DPPH
(IC50 µM)
ABTS
(%)
5a *52.25 ± 0.1934.27 ± 0.0726.43 ± 0.0175.46 ± 0.05
5b15.26 ± 0.0610.01 ± 0.0259.80 ± 0.0322.03 ± 0.01
5c30.01 ± 0.1119.68 ± 0.0442.86 ± 0.0243.34 ± 0.03
5d *54.13 ± 0.2035.51 ± 0.0723.98 ± 0.0178.18 ± 0.05
5e *53.23 ± 0.2034.91 ± 0.0723.71 ± 0.0176.87 ± 0.05
5f *53.14 ± 0.2034.85 ± 0.0722.83 ± 0.0176.74 ± 0.05
6a29.86 ± 0.1119.58 ± 0.0443.35 ± 0.0243.12 ± 0.03
6b34.93 ± 0.1322.91 ± 0.0436.02 ± 0.0250.45 ± 0.03
6c34.99 ± 0.1322.95 ± 0.0534.58 ± 0.0250.54 ± 0.03
6d30.42 ± 0.1119.95 ± 0.0440.11 ± 0.0243.93 ± 0.03
6e30.52 ± 0.1120.02 ± 0.0438.94 ± 0.0244.08 ± 0.03
6f30.47 ± 0.1119.98 ± 0.0437.60 ± 0.0244.00 ± 0.03
7a *52.34 ± 0.1934.33 ± 0.0724.94 ± 0.0175.59 ± 0.05
7b29.91 ± 0.1119.62 ± 0.0442.45 ± 0.0243.19 ± 0.03
7c29.96 ± 0.1119.65 ± 0.0440.72 ± 0.0243.27 ± 0.03
7d30.36 ± 0.1119.92 ± 0.0440.53 ± 0.0243.85 ± 0.03
7e15.68 ± 0.0610.29 ± 0.0251.98 ± 0.0322.65 ± 0.01
7f *54.04 ± 0.2035.45 ± 0.0721.37 ± 0.0178.05 ± 0.05
STDAscorbic AcidAscorbic Acid
79.10 ± 0.1171.85 ± 0.0529.30 ± 0.0141.25 ± 0.01
Data calculated from three replicates and presented as the mean ± SE. * Indicates the most potent pyrazole-based Schiff bases.
Table 2. The anti-inflammatory activities of pyrazole-based Schiff bases 5af, 6af, and 7af.
Table 2. The anti-inflammatory activities of pyrazole-based Schiff bases 5af, 6af, and 7af.
Pyrazole-Based Schiff BasesAnti-Inflammatory
Inhibition (%)
Protein DenaturationProteinase
5a *26.83 ± 0.0323.33 ± 0.03
5b7.83 ± 0.016.81 ± 0.01
5c15.41 ± 0.0213.40 ± 0.02
5d *27.79 ± 0.0324.17 ± 0.03
5e *27.33 ± 0.0323.76 ± 0.03
5f *27.28 ± 0.0323.72 ± 0.03
6a15.33 ± 0.0213.33 ± 0.02
6b17.94 ± 0.0215.60 ± 0.02
6c17.97 ± 0.0215.62 ± 0.02
6d15.62 ± 0.0213.58 ± 0.02
6e15.67 ± 0.0213.63 ± 0.02
6f15.64 ± 0.0213.60 ± 0.02
7a *26.87 ± 0.0323.37 ± 0.03
7b15.36 ± 0.0213.35 ± 0.02
7c15.38 ± 0.0213.38 ± 0.02
7d15.59 ± 0.0213.56 ± 0.02
7e8.05 ± 0.014.81 ± 0.01
7f *27.75 ± 0.0324.13 ± 0.03
STDDiclofenac Sodium
49.08 ± 0.0146.11 ± 0.02
Data calculated from three replicates and presented as the mean ± SE. * Indicates the most potent pyrazole-based Schiff bases.
Table 3. The cytotoxic activities (IC50 μM) and therapeutic indexes (TIs) of pyrazole-based Schiff bases 5a, 5d, 5e, 5f, 7a, 7f, and doxorubicin against lung (A549), colon (Caco-2), and normal lung (WI-38) cell lines.
Table 3. The cytotoxic activities (IC50 μM) and therapeutic indexes (TIs) of pyrazole-based Schiff bases 5a, 5d, 5e, 5f, 7a, 7f, and doxorubicin against lung (A549), colon (Caco-2), and normal lung (WI-38) cell lines.
Pyrazole-Based Schiff BasesLung (A549)Colon (Caco-2)Normal Lung (WI-38)Lung (A549)Colon (Caco-2)
(IC50 μM)Therapeutic Index (TI)
5a68.84 ± 0.1460.29 ± 0.14441.69 ± 8.806.417.32
5d48.61 ± 0.1462.33 ± 0.14731.72 ± 10.4614.8311.73
5e47.74 ± 0.2040.99 ± 0.20648.12 ± 7.5713.5715.81
5f60.45 ± 0.1261.98 ± 0.12493.07 ± 9.888.157.95
7a49.40 ± 0.1842.42 ± 0.18736.26 ± 7.9514.9017.35
7f55.74 ± 0.2449.01 ± 0.24542.51 ± 8.529.7311.06
Doxorubicin36.45 ± 0.1654.94 ± 0.16304.94 ± 4.728.365.55
Data calculated from three replicates and presented as the mean ± SE.
Table 4. The enzymatic assay values after the treatments of lung (A549) and colon (Caco-2) cells with pyrazole-based Schiff bases 5a, 5d, 5e, 5f, 7a, 7f, and doxorubicin.
Table 4. The enzymatic assay values after the treatments of lung (A549) and colon (Caco-2) cells with pyrazole-based Schiff bases 5a, 5d, 5e, 5f, 7a, 7f, and doxorubicin.
Pyrazole-Based Schiff BasesLung (A549)Colon (Caco-2)
Caspase-3
(pg/mL)
Bcl-2
(ng/mL)
Caspase-3
(pg/mL)
Bcl-2
(ng/mL)
DMSO85.92 ± 0.129.83 ± 0.0597.06 ± 0.147.99 ± 0.08
5a184.74 ± 0.266.55 ± 0.03208.68 ± 0.315.33 ± 0.05
5d300.73 ± 0.422.81 ± 0.01242.65 ± 0.363.20 ± 0.03
5e214.81 ± 0.303.93 ± 0.02315.45 ± 0.462.46 ± 0.02
5f236.29 ± 0.333.57 ± 0.02266.92 ± 0.392.91 ± 0.03
7a322.21 ± 0.452.62 ± 0.01363.98 ± 0.542.13 ± 0.02
7f244.88 ± 0.343.45 ± 0.02276.62 ± 0.412.80 ± 0.03
Doxorubicin330.80 ± 0.462.55 ± 0.01373.68 ± 0.552.08 ± 0.02
Data calculated from three replicates and presented as the mean ± SE.
Table 5. ADMT properties of pyrazole-based Schiff bases (5a, 5d, 5e, 5f, 7a, and 7f) using the pkCSM website.
Table 5. ADMT properties of pyrazole-based Schiff bases (5a, 5d, 5e, 5f, 7a, and 7f) using the pkCSM website.
Properties5a5d5e5f7a7fThe Ideal Values
Absorption
Intestinal absorption
(human)
91.35795.51096.00396.71193.72393.474Less than 30% is poorly absorbed
Skin permeability
(log Kp)
−2.735−2.735−2.735−2.735−2.735−2.735>−2.5 is low
Distribution
BBB permeability−1.318−1.530−1.550−1.704−1.433−1.818Poorly is <−1,
high is >0.3
CNS permeability−1.705−1.886−1.817−1.776−2.061−2.133Penetrate is >−2,
unable is <−3
Metabolism
CYP1A2 inhibitorYesYesNoNoYesNoNo
CYP2C19 inhibitorYesYesYesYesYesYesNo
CYP2C9 inhibitorYesYesYesYesYesYesNo
CYP2D6 inhibitorNoNoNoNoNoNoNo
CYP3A4 inhibitorYesYesYesYesYesYesNo
Toxicity
AMES toxicityNoNoNoNoNoYesNo
Skin sensitizationNoNoNoNoNoNoNo
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Naglah, A.M.; Almehizia, A.A.; Al-Wasidi, A.S.; Alharbi, A.S.; Alqarni, M.H.; Hassan, A.S.; Aboulthana, W.M. Exploring the Potential Biological Activities of Pyrazole-Based Schiff Bases as Anti-Diabetic, Anti-Alzheimer’s, Anti-Inflammatory, and Cytotoxic Agents: In Vitro Studies with Computational Predictions. Pharmaceuticals 2024, 17, 655. https://doi.org/10.3390/ph17050655

AMA Style

Naglah AM, Almehizia AA, Al-Wasidi AS, Alharbi AS, Alqarni MH, Hassan AS, Aboulthana WM. Exploring the Potential Biological Activities of Pyrazole-Based Schiff Bases as Anti-Diabetic, Anti-Alzheimer’s, Anti-Inflammatory, and Cytotoxic Agents: In Vitro Studies with Computational Predictions. Pharmaceuticals. 2024; 17(5):655. https://doi.org/10.3390/ph17050655

Chicago/Turabian Style

Naglah, Ahmed M., Abdulrahman A. Almehizia, Asma S. Al-Wasidi, Amirah Senaitan Alharbi, Mohammed H. Alqarni, Ashraf S. Hassan, and Wael M. Aboulthana. 2024. "Exploring the Potential Biological Activities of Pyrazole-Based Schiff Bases as Anti-Diabetic, Anti-Alzheimer’s, Anti-Inflammatory, and Cytotoxic Agents: In Vitro Studies with Computational Predictions" Pharmaceuticals 17, no. 5: 655. https://doi.org/10.3390/ph17050655

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop