Skip to main content

Scrutinizing pharmacological efficiency for Acacia auriculiformis by experimental and computational approach

Abstract

Background

The study sought to investigate the biological efficacy of methanol leave extract of Acacia auriculiformis (MEAA) via in vitro, in vivo, in silico approaches. The in vitro cytotoxicity was evaluated through brine shrimp lethality assay, and anti-inflammatory activity was determined by membrane stabilisation and protein denaturation methods (BSA and egg albumin). The in vivo antipyretic activity was examined via Brewer’s yeast induced pyrexia model.

Results

A. auriculiformis extract unveiled moderate cytotoxicity with significant anti-inflammatory efficacy (p < 0.001) compared to standard drug. This extract also exhibited dose-dependent time of paralysis and death for the worm (p < 0.001) in the anthelmintic test which was directly proportional to employed concentrations. A notable percentage of clot lysis effect (36.42 ± 1.95%, p < 0.001) was also observed for MEAA in human blood compared to control. However, this extract significantly (p < 0.05) reduced fever in a dose-dependent manner during the antipyretic experiment. Besides, in computer-aided investigations, two compounds (2,4-ditert-butylphenol and 3-hydroxy-β-damascone) revealed the best binding interaction with six proteins for cytotoxicity, inflammation, helminthic, thrombolytic and pyretic effect. Moreover, these two compounds satisfy Lipinski’s ‘Rule of Five’ and revealed drug-likeness profiles in the toxicological study.

Conclusions

These findings disclosed that methanol leaves extract of A. auriculiformis might be a potent source for anti-inflammatory, anti-helminthic, thrombolytic and antipyretic agents.

Background

Inflammation is a biological defensive response initiated by several stimuli, including irritants, chemicals, heat, infection or any immunological reactions [1]. In general condition, inflammation shows hyperthermia and pain in the living body and aims to repair damaged tissue by inactivating deleterious stimulus or pathogen. Hence, uncontrolled acute inflammation for a prolonged period can cause drastic pain and fever in the body. These can be pathophysiology of severe diseases like cancer, rheumatoid joint pain, asthma, diabetes, multiple sclerosis, autoimmune diseases, ischemic heart disease, arthritis, atherosclerosis and Alzheimer disease [2]. However, several inflammatory mediators like prostaglandins, bioactive lipid derivative are responsible for mediation of clotting, pain vasodilation, and pyrexia [3].

Pyrexia is a complex immune biochemical reaction that initiates by producing various endogenous pyrogens as a response to inflammatory or infectious stimulation [4]. The presence of pathogens or tissue damage in the body activates the immune system which produces and releases pyrogenic cytokines. In the hypothalamus, these synthesise cytokines react with prostaglandin receptors and alter the thermoregulation system. This alteration results in increased body temperature beyond the set point of 37.2 °C under the arm or axillar, 37.5–38.3 °C in the anus or rectum, 37.7 °C in the mouth and affects normal body functions [5]. Several antipyretic and anti-inflammatory agents like paracetamol, ibuprofen and NSAIDs are widely prescribed to reduce excessive body temperature [6]. These agents mainly exert their activity by inhibiting selective and non-selective prostaglandin and cyclooxygenase enzymes. Hence, long time inhibition of these enzymes can lead to serious consequences, including iatrogenic reactions, neutropenia, GIT disorder, kidney dysfunction, liver failure and other bleeding disorders [7]. A recent alternative strategy is the inhibition of microsomal mPGES-1 (prostaglandin E synthase-1), but till now, only two inhibitors were found [8]. For these reasons, a new therapeutic approach having multifaceted pharmacological targets and less side effect has been becoming an ultimate focus for researchers all around the world.

Since the hunter-gatherer age, humans have been using natural herbs to treat various diseases [9]. Natural products have always been a vital health concern due to their phytochemicals and nutritional values as they are a decent source of antioxidants, phenolics, vitamins, fibres, minerals, bioactive metabolites [10]. In many instances of modern synthetic drug development, phytoconstituents derived from natural plants screening program assists meticulously [11]. As plant metabolites contain elevated amounts of bio-chemical compounds with effective complementary pharmacological activities, their practice in alleviating chronic diseases is becoming a great area of interest [12]. Hence, appropriate strategic research on medicinal plants to discover novel bioactive agents is inevitable. Regarding this indigenous point of view, we aimed to investigate an ethnomedicinal plant Acacia auriculiformis, in our current study.

Acacia auriculiformis is locally known as Akashmoni in Bangladesh and babul in India. A. auriculiformis belongs to Plantae kingdom, Viridiplantae subkingdom, Streptophyta infrakingdom, spermatophyte phylum, Acacia mill genus and fabaceae/leguminosae family. Traditionally, this plant is used to treat rheumatism, aches, anthelmintic, pain, sore eyes, candidiasis and conjunctivitis [13]. Australian aborigines use root decoction of this plant in the treatment of rheumatism [14]. Seeds of this plant are traditionally used in the treatment of skin ailment, rashes, itching and allergy. In Nigeria, the Ibibio community uses this plant against malaria. Several phytochemicals, including flavonoids, carbohydrates, tannins, saponins and anthocyanidins, have been reported from A. auriculiformis [13]. Isolated compounds, namely acaciaside A and B, exhibited moderate anti-microbial and anti-fungal activity via in vitro assay [15]. However, ethanolic leave extract has shown dose-dependent antiplasmodial, anti-microbial, larvicidal activity along with memory improvement and acetylcholinesterase (AChE) enzyme inhibition in rats [16, 17]. Besides, Sathya et al. 2013 reported that petroleum ether extracts of empty pod and bark from this plant are potent candidates for diabetes and liver injury [18].

The plant A. auriculiformis possess several suitable medicinal properties, but until now, no study has been done reporting anti-inflammatory, anthelmintic, thrombolytic and antipyretic activities of this plant. Therefore, our present study aims to systematically explore these activities for methanol leaves extract of A. auriculiformis (MEAA). Most importantly, the current investigation aims to find out the therapeutic intervention of MEAA in mitigating inflammation by fever is the ultimate aim of our experiment. Additionally, a computational approach (molecular docking, ADME/T) was performed to predict possible insight targets of bioactive constituents of MEAA for a conceivable perspective.

Methods

Crude extract preparation

A. auriculiformis leaves were collected and identified from Rangamati region during September 19 and authenticated by a taxonomist under the accession number Anwar-0870. The leaves were washed and dried in a shaded place under 55–60 °C for 6 days. Coarse powder (490 g) was obtained from dried leaves by using an electric grinder (Sahara mixer grinder, Canbara industries PVT. Ltd., Mumbai-400072). The coarse powder was soaked in an appropriate amount (1000 mL) of methanol (90%) for 7 days with occasional shaking and stirring at room temperature. The filtration was done by Whatman No. 1 filter paper in a laboratory condition 27 ± 2 °C. The finale filtrate was allowed to dry and evaporate in a water bath at 45–50 °C. Four gram sticky, greenish, semisolid extract was found and preserved in 4 °C until further use as a methanol extract of A. auriculiformis (MEAA) [7].

Chemicals

Streptokinase vial (1,500,000 IU), loperamide and vincristine sulfate (1 mg/vial) WERE obtained from Beacon Pharmaceuticals Ltd. Bangladesh. Methanol (Merck, Germany), Tween 80 and DMSO were purchased from BDH Chemicals. Bovine serum albumin and diclofenac sodium were purchased from Square Pharmaceuticals Ltd. Normal saline solution (0.9% NaCl) was obtained from Orion Infusion Ltd. Other chemicals were procured from local sources. All the chemicals were in analytical grade.

Experimental animal

Swiss albino male mice (weight: 25–30 g) were procured from an appropriate source. All mice were housed in a laboratory for 14 days to familiarise with the standard environment. Standard guidelines such as room temperate 25 ± 2 °C, relative humidity 55–60%, 12 h light/dark cycle, whereas the food pellets and water supply were maintained strictly until the experiment period. Using diethyl ether anaesthesia, the mice were euthanised after the experiment. All the mice have been slaughtered using diethyl ether anaesthesia at the end of the observation period. The ARRIVE guidelines are in compliance with for the mice model study.

In vitro study

Cytotoxicity activity by brine shrimp lethality bioassay

In vitro brine shrimp lethality bioassay for MEAA was performed by the method delineated by Meyer et al. 1982 [19]. Artificial seawater was obtained by dissolving NaCl (38 g) in distilled water (1000 mL). The pH at 8.0 was attuned by adding NaOH. MEAA (5 mg/mL) concentrations (31.25, 62.5, 125, 250 and 500 μg/mL) were obtained via serial dilution. Similarly, as a standard control, serially diluted vincristine sulfate (3.13, 6.25, 12.5, 25 50 and 100 μg/mL) was used. After obtaining the concentrations, ten alive brine shrimp were placed in each tube at 25 ± 1 °C. The number of alive nauplii was counted and recorded after 24 h by using the magnifying glass. The following equation used to determine the % of mortality,

$$ \mathrm{Mortality}\%=\frac{\mathrm{Nt}-\mathrm{Na}}{\mathrm{Nt}}\times 100 $$

Here, Nt = number of nauplii taken, Na = number of nauplii alive

Anti-inflammatory test on protein denaturation inhibition by egg albumin

The anti-inflammatory activity of MEAA was studied by the egg albumin inhibiting protein denaturation method used by Guha et al. 2020 with slight modification [20]. The reaction solution (5 mL) contains egg albumin (0.2 mL), phosphate-buffered saline at pH 6.4 (2.8 mL) and MEAA extract (2 mL) varying concentration of 62.5, 125, 250 and 500 μg/mL. The exact volume of distilled water was used as a control group. In a BOD incubator (Lab-line technology, Japan), all the solutions were incubated for 15 min at 37 ± 2 °C and heated for 5 min at 70 °C. After cooling, the absorbance was measured at 660 nm (SHIMADZU, UV 1800) where water (vehicle) was used as a control and diclofenac sodium concentrations (31.25, 62.5, 125, 250 and 500 μg/mL) used as a reference solution. The test was carried out triplicate times, and the following equation was used to determine the percentage of protein denaturation inhibition,

$$ \mathrm{Inhibition}\ \left(\%\right)=\frac{\mathrm{Abs}\ \mathrm{control}-\mathrm{Abs}\ \mathrm{test}}{\mathrm{Abs}\ \mathrm{control}}\times 100 $$

Abs control = absorbance of the control, Abs test = Absorbance of test

Anti-inflammatory test on protein denaturation inhibition by BSA

The anti-inflammatory activity of MEAA was studied by BSA inhibiting protein denaturation method formerly used by Tareq et al. 2020 with minor modifications [9]. The test mixture (0.5 mL) composed with 0.45 mL aqueous albumin (5% w/v) and 0.05 mL of MEAA extract while control mixture (0.5 mL) contain 0.45 mL aqueous albumin (5% w/v) and 0.05 mL of distilled water. The product mixture was composed of 0.05 mL of the test mixture and 0.45 mL of distilled water. The standard mixture (0.5 mL) comprises 0.05 mL of diclofenac sodium instead of distilled water. Different concentration for both test and standard mixture was obtained (125, 250, 500 and 1000 μg/mL) and pH was attuned to 6.3 by adding 1 N HCl in all concentrations. After that, all mixture was incubated for 20 min at 37 °C and then kept for 30 min at 57 °C. Finally, after cooling the mixture, phosphate buffer (2.5 mL) was added to each mixture and absorbance was recorded at 416 nm by using a UV-visible spectrophotometer. The test was carried out triplicate times and the following equation determined the percentage of protein denaturation inhibition,

$$ \mathrm{Inhibition}\ \left(\%\right)=100-\left(\frac{\ A-B}{C}\right)\times 100\Big) $$

A = Absorbance of test mixture; B = Absorbance of product mixture; C = absorbance of control mixture.

Anti-inflammatory test on membrane stabilisation by hypotonic induced haemolysis

In vitro membrane stabilisation activity for MEAA was assessed by the described method Sabiu et al. 2016 [21]. The text mixture was serially diluted at 31.25, 62.5, 125, 250, 500 and 1000 μg/mL concentrations. Each concentration contains MEAA (0.5 mL), phosphate buffer (1 mL), hyposaline (2 mL) and human red blood cell (HRBC) suspension (0.5 mL). The standard mixture (diclofenac sodium) contains the same chemicals except for the extract, whereas the control mixture contains all chemicals except the standard drug and extract. All the mixtures were incubated for 30 min at 37 °C. After cooling, centrifugation was for 20 min at 3000 rpm. Therefore, absorbance was taken for obtained supernatant solutions by using UV-visible spectrophotometer at 540 nm. The inhibitory haemolysis effect of MEAA was determined via the following equation

$$ \mathrm{Membrane}\ \mathrm{stability}=\left(\frac{\ A-B}{A}\right)\times 100 $$

Here, A = absorbance of control mixture, B = absorbance of test mixture.

In vitro anthelmintic activity on the worm Tubifex tubifex

The anthelmintic activity of MEAA was evaluated by the method previously used by Adnan et al. 2018, with contemptible modifications [22]. The worms (Tubifex tubifex) were collected from aquarium shop Chittagong with 2–2.5 cm average length. The test groups contained concentrations of MEAA (5, 8 and 10 mg/mL), whereas the control group consists of distilled water and the standard group contains levamisole (1 mg/mL). Five groups were divided into five petri dish each contains randomly selected ten worms. For each aimed groups, 3 mL of the mixture was added. After that, ‘time of paralysis’ and ‘time of death’ of worms were observed and recorded carefully. When the worms stopped movement but keep vigorous shaking was considered as ‘time of paralysis’, and the period when the worms completely stopped movement was regarded as ‘time of death’. The experiment was conducted triplet time.

In vitro thrombolytic activity via clot lysis

In vitro clot lysis activity was assessed by the described method of Prasad et al. 2006 [23]. As a test sample, MEAA crude extract (100 mg) was mixed with 10 mL distilled water and kept overnight. Streptokinase (1,500,000 IU) was used as a stock solution. A total of 5 mL venous blood was drawn from 10 healthy and young volunteers (no history of any substance/drug used in the past 7 days). Blood retrieved from each volunteer was dispersed in pre-weighed Eppendorf tubes. Each Eppendorf containing 0.5 mL blood was incubated for 45 min at 37 °C to form a clot. After incubation, layers of clot and serum were formed in each Eppendorf tubes. Serum was carefully aspirated out from each tube without disturbing the clot and the weight of tubes containing clots was measured. Therefore, 100 μL of MEAA (10 mg/mL) solution was added to each tube, whereas in the standard control tube streptokinase (SK) (100 μL), and in the control group distilled water (100 μL) was used. Then all the tubes were incubated for 90 min at 37 °C. The released fluid from each tube again removed after incubation without distressing the clot. Hereafter, all the tubes weighted again to observe clot disruption. The equation described by Banu et al. 2020 [24] was followed to determine the percentage of clot lysis.

In vivo study

Antipyretic test using Brewer’s yeast induced pyrexia mice model

The antipyretic activity of MEAA was ascertained by the method previously used by Ahmed et al. 2019 [25]. Sixteen Swiss albino rodents were taken randomly for our current experiment and identified numerically. Then, by using a digital thermometer, the initial body temperature for all of the rodents was recorded. In the initial period, the temperature 37.25 °C is considered as usual and above 37.50 °C was considered as fever. Twenty percent of an aqueous suspension of Brewer’s yeast (10 mL/kg, s.c.) was injected into all rodents to induce pyrexia. After pyrexia induction, all rodents were fasted overnight with only access to drink water. After 18 h, the temperature was again observed and recorded for each rodent via a digital thermometer. An increment of the temperature of more than 0.5 °C confirms the induction of pyrexia [26]. Therefore, rodents that did not show temperature increment were excluded from the current experiment. Hereafter, twelve male pyrexia-induced Swiss albino mice were divided into four groups, each containing three rodents. Group I was considered negative control and received 10 mL/kg saline, whereas group II was considered a control group and received paracetamol (150 mg/kg). The remaining two groups (III and IV) were considered test groups where group III received an MEAA dose of 200 mg/kg and group IV received an MEAA dose of 400 mg/kg. After drug administration, rectal temperature was again recorded and observed for 1 h, 2 h, 3 h and 4 h periodically. Using diethyl ether anaesthesia, the mice were euthanised after the experiment. All the mice have been slaughtered using diethyl ether anaesthesia at the end of the observation period. The percentage of pyrexia reduction was determined by the following equation,

$$ \%\mathrm{reduction}=\frac{PiT- Cn}{PiT- NbT}\times 100 $$

Here, PiT = Pyrexia induced body temperature, Cn = temperature after 1 h, 2 h, 3 h, 4 h, NbT = normal body temperature.

Molecular docking

Formerly isolated compounds from leaves extract of A. auriculiformis via GC-MS analysis were exploited for molecular docking by following previously elucidated protocol Rahman et al. 2020 [12, 27]. Three-dimensional structures of the following proteins were retrieved from Protein Data Bank [28]: cyclooxygenase (COX)-1 (PDB ID: 2OYE) [29], cyclooxygenase (COX)-2 (PDB ID: 6COX) [30], microsomal prostaglandin E synthase (mPGES)-1 (PDB ID: 3DWW) [31], human estrogen receptor (PDB ID: 1ERR) [32], tubulin-colchicine (PDB ID: 1SA0) [33], tissue plasminogen activator (PDB ID: 1A5H) [34]. Molecular docking study was analysed by Schrödinger Maestro (v11.1).

ADME/T and toxicological properties analysis

The drug-like attributes (ADME/T) of the compounds were analysed based on Lipinski’s rules of five by using the tool QikProp (Schrödinger v11.1) [35]. Toxicological properties were evaluated via admetSAR (Online tool) [36].

Statistical analysis

Data were uttered as mean ± SEM (standard error of the mean). The one-way statistical analysis of variance (ANOVA) was done by following post hoc Dunnett’s test for multiple comparisons. In the outcomes, p < 0.05, p < 0.01 and p < 0.001 are considered as statistically significant for all cases. The analysis was carried out by using either the statistical software package for social science (SPSS, version 25.0, IBM Corporation), Prism version 8.4.2 (GraphPad Software Inc., La Jolla, CA, USA), or Microsoft Excel '13.

Results

Effect of MEAA on brine shrimp lethality bioassay

To determine the cytotoxicity of MEAA, brine shrimp lethality bioassay was performed in the current study. Results demonstrates that the MEAA exhibited moderate cytotoxicity (LC50 = 426.38 μg/mL) against positive control vincristine sulfate (LC50 = 39.25 μg/mL). The lethal median dose (LC50) was calculated from the regression analysis curve and the best fit line is visualised in Fig. 1.

Fig. 1
figure 1

Cytotoxicity of Methanol leaves extract of A. auriculiformis (MEAA) and Vincristine sulfate (VS) in brine shrimp lethality assay

Effect of MEAA on protein denaturation inhibition by egg albumin

The results demonstrates that for 62.5, 125, 250 and 500 μg/mL concentrations, MEAA showed 47.05%, 42.89%, 41.42%, 40.68% inhibition of egg albumin whereas, diclofenac sodium showed 83.5%, 77.96%, 67.77% and 65.4% inhibition. Statistically, both MEAA and diclofenac sodium concentrations revealed significant (p < 0.001) egg albumin inhibition. The results are demonstrated in Fig. 2.

Fig. 2
figure 2

Percentages of egg albumin protein denaturation by methanol extract of methanol leaves extract of A. auriculiformis (MEAA) and standard drug diclofenac sodium. Values are mean ± SEM (n = 3). cp < 0.001, significantly different from control followed by Dunnett’s test

Effect of MEAA on protein denaturation inhibition by BSA

In albumin bovine serum denaturation method, different concentrations of MEAA was found significant (p < 0.001) compared to a control mixture. The results reveals that at different concentrations (125, 250, 500 and 1000 μg/mL), MEAA exhibited 73.81%, 76.67%, 79.05%, 80.95% inhibition of denaturation of bovine serum, whereas at same concentrations, diclofenac sodium showed 90%, 95.71%, 97.14%, 98.57% inhibitions (p < 0.001). The results are represented in Fig. 3.

Fig. 3
figure 3

Percentages of bovine serum albumin protein denaturation by methanol extract of methanol leaves extract of A. auriculiformis (MEAA) and standard drug diclofenac sodium. Values are mean ± SEM (n = 3). cp < 0.001, significantly different from control followed by Dunnett’s test

Effect of MEAA on membrane stabilisation by hypotonic induced haemolysis

The membrane stabilisation method is widely used to determine anti-inflammatory activity due to similarities in lysosomal membranes and human red blood cell membranes. Therefore, we also performed membrane stabilisation by hypotonic induced haemolysis method to further ascertain the anti-inflammatory activity of MEAA. In this experiment, different concentrations of MEAA were found to be dose-dependent and the percentage of haemolysis inhibition was significantly increased compared to the control mixture (p < 0.001). At different concentrations (31.25, 62.5, 125, 250, 500 and 1000 μg/mL), MEAA unveiled 63.39%, 73.63%, 77.95%, 81.98%, 86.78%, 89.88% of inhibition, whereas at same concentrations diclofenac sodium displayed 79.47%, 83.45%, 87.02%, 90.05%, 94.09%, 97.89% of haemolysis inhibition (p < 0.001). The result is summarised in Table 1.

Table 1 Membrane stabilising activity of methanol leaves extract of A. auriculiformis

Effect of MEAA on the worm Tubifex tubifex

Due to anatomical and physiological relevancy to intestinal worms (Annelida), Tubifex tubifex was used in our current experiment. Results have shown that the anthelmintic activity of MEAA was directly proportional to employed concentrations. From lowest to highest concentrations (5, 8 and 10 mg/mL), MEAA exhibited (p < 0.001) paralysis times of (26.03 ± 0.34), (20.67 ± 0.19) and (15.50 ± 0.04) minutes, while time of death were (59.65 ± 0.18), (50.18 ± 0.06), (45.14 ± 0.36) minutes respectively (p < 0.001). The positive control (levamisole, 1 mg/mL) exhibited paralysis time (3.22 ± 0.08) minutes and time to death of (6.19 ± 0.61) minutes (p < 0.001). Figure 4 outlines all the results.

Fig. 4
figure 4

Anthelmintic activity of methanol leaves extract of A. auriculiformis (MEAA). Values are represented in Mean ± SEM (n = 3). cp < 0.001 statistically significant compared to levamisole followed by one-way ANOVA (t test)

Effect of MEAA on clot lysis

In this module, MEAA revealed significant clot lysis activity (36.42 ± 1.95%, p < 0.001), whereas standard control 100 μL of streptokinase unveiled (51.31 ± 5.14%, p < 0.001) compared to negative control water (6.62 ± 2.83%). All the data are displayed in Fig. 5.

Fig. 5
figure 5

Methanol leaves extract of A. auriculiformis (MEAA) and standard drug streptokinase (SK) effects on the clot lysis of blood. Values are represented in Mean ± SEM (n = 6). cp < 0.001 were statistically significant in comparison with the negative control (water) followed by one-way ANOVA

Effect of MEAA on Brewer’s yeast induced pyrexia mice model

Yeast-induced pyrexia model was used in our present study to identify fever relieving activity of MEAA. Statistical analysis showed that at both doses 200 and 400 mg/kg, percent of pyrexia inhibition was significantly dose-dependent. After 3 h of 200 mg/kg MEAA dose administration, pyrexia level in mice reduced significantly 49.05% (p < 0.01). The maximum antipyretic activity 21.23% was observed for dose 200 mg/kg, whereas the 400 mg/kg dose exhibited 15.18% inhibition. Standard drug paracetamol (150 mg/kg) revealed 78.67% inhibition. The result of the antipyretic test is documented in Table 2.

Table 2 Antipyretic activity of methanol leaves extract of A. auriculiformis (MEAA) and standard drug

Molecular docking study for inflammation and pyretic

To perceive possible mechanism behind inflammatory and pyretic activity of A. auriculiformis, isolated selected compounds (2,4-ditert-butylphenol, 2-palmitoylglycerol, 3-hydroxy-β-damascone, α-monostearin) were docked with cyclooxygenase (COX)-1 (PDB ID: 2OYE), cyclooxygenase (COX)-2 (PDB ID: 6COX), mPGES-1 (PDB ID: 3DWW). For cyclooxygenase-1, 2-4-ditert-butylphenol revealed the best docking score (− 7.716 kcal/mol) bonded with the SER 530 (H-bonding), LEU 352, LEU 359, LEU 531, ALA 527 (Pi-alkyl-bonding) and with VAL 16, VAL 349 (Alkyl-bonding) pockets (Fig. 6). Whereas 2-palmitoylglycerol exhibited the lowest docking score (− 5.39 kcal/mol) and showed pocket bonding interactions with MET 522, SER 530 (H-bond); GLY 526 (van der waals); LEU 115, LEU 93, ALA 527 (Pi-alkyl-bonding); VAL 116, TYR 355, ILE 523, VAL 349, ILE 89 (Alkyl-bonding). The rank of binding scores were 2-4-ditert-butylphenol (− 7.716 kcal/mol) > 3-hydroxy-β-damascone (− 7.021 kcal/mol) > α-monostearin (− 5.6 kcal/mol) > 2-palmitoylglycerol (− 5.39 kcal/mol). Hence, 2,4-ditert-butylphenol also exhibited the best docking score for COX-2 receptor (− 6.954 kcal/mol) and α-monostearin showed the lowest binding score (− 4.072 kcal/mol). In this case, 2,4-ditert-butylphenol showed bonding interactions with HIS 90 (van der waals), LEU 531 (C-H bond); Leu 352 (Pi-alkyl-bonding), ALA 527 (Pi-alkyl-bonding), TYE 355 (Pi-alkyl-bonding), VLA 349 (alkyl-bonding), VAL 523 (alkyl-bonding) pockets and α-Monostearin showed LEU 352 (Pi-alkyl-bonding), ALA 516 (Pi-alkyl-bonding), PHE 518 (alkyl-bonding), HIS 90 (alkyl-bonding) pocket bonding interactions (Fig. 6). The ranking order of binding score for compounds with COX-2 receptor were 2,4-ditert-butylphenol (− 6.954 kcal/mol) > 3-hydroxy-β-damascone (− 6.882 kcal/mol) > 2-palmitoylglycerol (− 5.22 kcal/mol) > α-monostearin (− 4.072 kcal/mol). For mPGES-1 receptor, the best score was unveiled by 3-hydroxy-β-damascone (− 4.245). The pocket binding interactions are ASN 74 (H-bond and C-H bond), ARG 126 (H bond); ARG 73 (Pi-alkyl), ALA 31(Pi-alkyl); HIS 72 (alkyl), HIS 113 (alkyl), TYR 28 (alkyl). Minimum score exposed by 2-4-ditert-butylphenol (− 3.848) which bind at ALA 31 (H bond); HIS 72 (C-H bond); GLU 77 (Pi-Cation); ARG 126 (Pi-Anion); HIS 72 (Pi-Pi T-shaped). Conversely, 2-palmitoylglycerol and α-monostearin did not reveal any interaction with mPGES-1 receptor. All the results are shown in Table 3 and binding interactions displayed in Table 4.

Fig. 6
figure 6

3D representation of best docking score compounds. a 2,4-Ditert-butylphenol with COX1 receptor (PDB: 2OYE). b 2,4-Ditert-butylphenol with COX2 receptor (PDB: 6COX). c 3-Hydroxy-β-damascone with microsomal prostaglandin E synthase (mPGES)-1 (PDB: 3DWW). d 2-4-Ditert-butylphenol with human estrogen receptor (PDB: 1ERR). e 3-Hydroxy-β-damascone with tubulin-colchicine (PDB: 1SA0). f 2-4-Ditert-butylphenol with tissue plasminogen activator (PDB: 1A5H)

Table 3 Molecular docking score of isolated compounds from A. auriculiformis
Table 4 Binding interactions of isolated compounds with the respective proteins for different biological activities

Molecular docking for brine shrimp lethality bioassay

Human estrogen receptor (PDB ID: 1ERR) were used to perform molecular docking for brine shrimp lethality bioassay. Results show that 2-4-ditert-butylphenol gave the highest binding score (− 7.69 kcal/mol) (Fig. 6). Besides, 2-palmitoylglycerol gave the lowest binding score (− 4.241 kcal/mol) through one hydrogen bond to ASP 351. The binding score ranks are 2-4-ditert-butylphenol (− 7.69 kcal/mol) > 3-hydroxy-β-damascone (− 6.866 kcal/mol) > α-monostearin (− 4.948 kcal/mol) > 2-palmitoylglycerol (− 4.241 kcal/mol). All the results are shown in Table 3 and binding interactions displayed in Table 4.

Molecular docking for anthelmintic activity

Selected compounds are docked with the tubulin-colchicine receptor (PDB ID: 1SA0) for anthelmintic activity. The best docking fitness score shown by 3-hydroxy-β-damascone (− 6.515 kcal/mol) bonded to THR 179 pocket via H-bonding and with LEU 248, LEU 255, ALA 316, LYS 352, MET 259, ALA 180, VAL 181 pockets via alkyl-bonding (Fig. 6). Binding fitness score order are 3-Hydroxy-β-damascone damascone (− 6.515 kcal/mol) > 2-4-ditert-butylphenol (− 6.401 kcal/mol) > α-monostearin (− 5.077 kcal/mol) > 2-palmitoylglycerol (− 5.067 kcal/mol) as reported in Table 3. The binding interactions are shown in Table 4.

Molecular docking for clot lysis activity

The docking result for thrombolytic activity is documented in Table 3. In this assay, selected compounds are docked with tissue plasminogen activator receptor (PDB ID: 1A5H). The strongest docking interaction is exhibited by 2-4-ditert-butylphenol (− 5.42 kcal/mol) interacted with SER 195 (H-bonding), GLY 193 (H-bonding), GLN 192 (van der waals-bonding), HIS 57 (Pi-Pi T-shaped-bonding), CYS 42 (Pi-alkyl-bonding) pockets (Fig. 6). The lowest binding affinity was demonstrated by α-monostearin (− 2.706 kcal/mol). The other two compounds showed − 4.269 kcal/mol (2-palmitoylglycerol), − 3.494 kcal/mol (3-hydroxy-β-damascone) docking score, respectively. The binding interactions are documented in Table 4.

ADME/T screening for drug-likeness

Compounds compatibility as a drug molecule was investigated in the current study. According to Lipinski’s ‘Rules of Five’, any orally administered compound must have molecular weight < 500 g/mol, hydrogen bond acceptor ≤ 10, hydrogen bond donor ≤ 5, lipophilicity value (LogP) ≤ 5. Two compounds (2-palmitoylglycerol, α-monostearin) in our study violated Lipinski’s ‘Rules of Five’. Further, the toxicological properties were predicted by using admetSAR online tool and confirm that none of the compounds possess carcinogenicity or AMES toxicity (Table 5). This elucidates that two derivatives (2,4-ditert-butylphenol and 3-hydroxy-β-damascone) can be considered as possible lead compounds and a good candidate for the advancement of new drugs.

Table 5 ADME/T and toxicological properties prediction of the bioactive compounds

Discussion

Brine shrimp lethality bioassay is known as a safe, economical and effective method to evaluate the bioactivity of plant products. Although an ideal biological response depends on bioactive constituents from plant parts but analysing plant crude extract is important. A good relationship has been found for pesticidal and cytotoxic activity between this method and solid human tumours which is beneficial for natural pesticides and pre-screening tests for antitumor research [19]. In this design, the higher the value of LD50, the lower the toxicity of the extract is and vice versa [12]. In the current study, MEAA showed an LC50 value of 426.38 μg/mL, which is moderately toxic, and the standard control vincristine sulfate exhibited an LC50 value of 39.25 μg/mL. These moderate toxicity might be due to the presence of triterpenoid and saponins in A. auriculiformis; therefore, it can be used as an active chemopreventive agent previously suggested by Kaur et al. 2002 [37].

Traditionally, A. auriculiformis is recommended for rheumatism, pain, itching and allergy [38]. As these causes initiate by inflammation, therefore current investigation aimed to assess the anti-inflammatory activity of MEAA. Implicit drawbacks are associated with using animal models in pharmacological research, including lack of justifications and ethical concerns [20]. Therefore, in vitro protein denaturation and membrane stabilisation methods were proposed in the ongoing study to evaluate the anti-inflammatory property. In the living body, HRBC lysis (human red blood cell) and protein denaturation are often caused by inflammation, which initiates by the excess synthesis of COX [24]. During these conditions, proteins secondary and tertiary structures gone misplaced due to heat, strong acid or base, injurious agent, stress or organic solvent [10, 39]. In the current study, at both of protein denaturation model (egg albumin, BSA) MEAA exhibited a significant percentage of inhibition (p < 0.001), which was parallel to diclofenac sodium (standard drug). Besides, in membrane stabilisation assay, dose-dependent haemolysis inhibition of MEAA (p < 0.001) was found a comparative preventative response as close to the standard diclofenac sodium (p < 0.001). An earlier finding indicated the presence of flavonoids, tannins in A. auriculiformis [13]. Studies revealed that a number of flavonoids have strong inhibitory ability against the synthesis of cyclooxygenase and prostaglandin and several other enzymes such as phospholipase A2, protein kinase C, phosphodiesterases and protein tyrosine kinases [40, 41]. Besides, many investigations proposed plant metabolites like alkaloids, flavonoids, phenols and tannins possess anti-inflammatory activity due to their membrane stabilisation capabilities [20, 42]. Thus it can be presumed that due to the presence of flavonoids, tannins and phenols, A. auriculiformis possess significant anti-inflammatory activity. However, in-depth investigations by isolation and identification of bioactive metabolites from this plant extracts and fractions are highly recommended to reveal the exact pathway responsible for the aforementioned attributes.

Around 2 billion people all around the world suffer from parasitic infections. Parasitic worm infections are known as one of the major causes for developing many complicated chronic maladies like schistosomiasis, elephantiasis and onchocerciasis [43]. Though many synthetic agents are available for anthelmintic treatment but these agents produce severe complications. Therefore, due to less or fewer side effects in plant-derived, a new agent from a natural source has become an intense interest. A. auriculiformis is widely used as an anthelmintic in various regions of the world [13]. To verify this ethnopharmacological use, we planned an experiment on the methanol leaf extract of A. auriculiformis on the worm. In our current investigation, all concentrations provoked the worm’s paralysis and caused death in a dose-dependent manner. Period of paralysis and death of worms were inversely proportional to extract concentrations (p < 0.001). Numerous studies advocated that phytochemicals like alkaloids, tannins, phenolic and flavonoids are responsible for anthelmintic activity [44]. Tannins might unveil activity by uncoupling oxidative phosphorylation and interacting with the worm’s energy production, leading to the death of the worm. In contrast, alkaloid acts on the CNS of worms [44]. Previously, the presence of tannins has been reported from this plant [13]. Therefore, we can assume that, due to the ubiquity of tannins in A. auriculiformis, MEAA might interact with the glycoprotein of the parasite in the gastrointestinal tract and inhibit the worm’s energy production which ultimately leads to the death of the worm.

Diverse assortments of thrombolytic agents are available, which clears the network of cross-linking fibrins via triggering the plasminogen enzyme. This process makes the clot soluble and renovates blood flow in blood vessels by clearing the clogged artery, thus avoid permanent tissue damage. This therapy is useful to treat several conditions like myocardial infarction, deep vein thrombosis, thromboembolic strokes and pulmonary embolism. Streptokinase is one of that kind agent and used in our current study as a positive control group. In our investigation, MEAA disclosed significant (p < 0.001) clot lysis against the control group. Besides, Jia et al. 2016 hypothesised that a formulation of flavans and free B-ring flavonoids from this plant could be useful to inhibit platelet-induced thrombosis diseases [45]. Therefore, MEAA could be denoted as a potential source to inhibit blood clots in the future, although this outcome is not as significant as streptokinase.

Brewer’s yeast-induced pyrexia model is known as a convenient test for screening antipyretic activity of synthetic drugs along with plant phytochemicals [26]. Yeast induces in the living body causes a biochemical cascade and increase the synthesis of cytokines such as tumour necrosis factor-α (TNF-α), IL-6, interferon-α and interleukin-1β especially prostaglandins. Increased synthesis of prostaglandins alters thermoregulation in the hypothalamus [46]. Therefore, any agent that can constrain this cascade of pyrexia-inducing mediators will be marked as an antipyretic agent (Paracetamol) [47]. In the current study, administration of MEAA (200 and 400 mg/kg) exhibited reduction of rectal temperature in rodents (p < 0.05). This reduction of temperature was observed might be due to the presence of some pharmacologically active constituents in A. auriculiformis, which interfere with the synthesis of prostaglandin. However, phytochemicals, specifically steroids and flavonoids, are known as a good antipyretic agent and in the previous study presence of these phytochemicals has been observed in A. auriculiformis [13, 48]. Nevertheless, it must be noted that diverse biochemical reactions take place during the synthesis of prostaglandins. Therefore, further in-depth investigation is worthwhile to regulate the exact biosynthetic pathway where the extract exerts fever relieving effects.

Computer-aided molecular docking helps to predict the molecular mechanism by ligand-target interactions and correlate the experimental data with a pharmacological response. This process plays a crucial role to bolster medical and pharmacological innovations by analysing the binding modes of active compounds against the targeted receptors. Therefore, in the present study, contemporary computer-aided molecular docking was done for previously isolated bioactive compounds via GC-MS analysis. Compounds were chosen based on Lipinski’s ‘Rules of Five’ and further ADME/T properties were investigated. Against six targeted protein in the current study, 2-4-ditert-butylphenol unveiled promising score for inflammatory and pyretic (PDB ID: 2OYE, 6COX, 3DWW), cytotoxic (PDB ID: 1ERR) and thrombolytic (PDB ID: 1A5H) compared to other compounds. This outcome confirms that 2-4-ditert-butylphenol can directly bind with targeted enzymes and decrease enzymatic activities and reveal potential activity. Hence, in earlier, this compound expressed interaction with COX2, interleukin IL-6, IL-1b and TNF-α in RAW 264.7 cells and MCF-7 cell line activation of P53 gene has observed [49]. However, 3-hydroxy-β-damascone exhibited the best interaction score against helminthic (PDB ID: 1SA0), inflammatory and pyretic (PDB ID: 3DWW). Based on our current finding, we can conclude that due to the presence of these compounds, MEAA disclosed significant activity, and it could be a potential bioactive compound for treating several diseases.

Pharmacokinetic (ADME/T) and toxicological properties are imperative before the introduction of drugs in the commercial market. This step helps to understand the pharmacokinetic view and reduce cost during drug development. Therefore, selected compounds from methanol leaves extract of A. auriculiformis subjected to ADME/T analysis. Results exhibited that selected isolated compounds 2,4-ditert-butylphenol and 3-hydroxy-β-damascone satisfy Lipinski’s ‘Rule of Five’ and indicate as a promising drug candidate for anti-inflammatory, antipyretic, anthelmintic and thrombolytic. In the toxicological study, selected compounds displayed non-Ames toxicity, non-carcinogenicity and none possess rat acute toxicity. Though two compounds α-monostearin and 2-palmitoylglycerol showed LD50 values more than 5000 mg/kg in acute oral toxicity, which means that these compounds are not orally safe. Thus, in silico results in this study referred all selected isolated compounds were accountable and similarities between biological and computer-aided models results were witnessed. Therefore, this investigation gives a better understanding via correlating with pharmacokinetics and pharmacodynamics properties with the experimental outcomes for next step investigation before the clinical trial.

Conclusion

This experiment confirms that methanol leave extract of A. auriculiformis has an interesting paradox having the potentiality to contribute to the development of new anti-inflammatory, antipyretic, anthelmintic and thrombolytic agents due to the presence of several secondary metabolites. In the computer-aided investigation, among all the previously isolated selected compounds, two compounds satisfied Lipinski’s ‘Rule of Five’. 2-4-Ditert-butylphenol unveiled significant binding interaction for anti-inflammatory, antipyretic and thrombolytic activities. Moreover, 3-hydroxy-β-damascone disclosed most promising score for anthelmintic activity. In pharmacokinetic (ADME/T) and toxicological analysis, two compounds (2,4-ditert-butylphenol and 3-Hydroxy-β-damascone) revealed a safety profile with oral bioavailability from the druggable point of view. Therefore, comprehensive, in-depth analysis of this plant with isolated compounds is highly recommended to confirm the mechanisms of action for aforesaid pharmacological activities by the animal model and subsequently in humans for clinical efficacy.

Availability of data and materials

The datasets used and/or analysed during the current study are available from the corresponding author on reasonable request.

Abbreviations

MEAA:

Methanol extracts of A. auriculiformis

BSA:

Bovine serum albumin

SK:

Streptokinase

VS:

Vincristine sulfate

s.c:

Subcutaneous

BW:

Bodyweight

ANOVA:

Analysis of variance

SEM:

Standard error of the mean

ADME/T:

Absorption, Distribution, Metabolism, Excretion, and Toxicity

References

  1. Sharma VC, Kaushik A, Dey YN, Srivastava B, Wanjari M, Jaiswal B (2020) Analgesic, anti-inflammatory and antipyretic activities of ethanolic extract of stem bark of Anogeissus latifolia Roxb. Clin Phytosci 6(1):22. https://doi.org/10.1186/s40816-020-00171-2

    Article  CAS  Google Scholar 

  2. Osman WA, Labib DA, Abdelhalim MO, Elrokh EM (2017) Synergistic analgesic, antipyretic and anti-inflammatory effects of extra virgin olive oil and ibuprofen in different experimental models of albino mice. Int J Rheum Dis 20(10):1326–1336. https://doi.org/10.1111/1756-185X.13105

    Article  CAS  PubMed  Google Scholar 

  3. Chirisa E, Mukanganyama S (2016) Evaluation of in vitro anti-inflammatory and antioxidant activity of selected zimbabwean plant extracts. Int J Geogr Inf Syst 22(2):157–172. https://doi.org/10.1080/10496475.2015.1134745

    Article  Google Scholar 

  4. Choi J, Chang S, Ahn JG (2020) Comparison of fever-reducing effects in self-reported data from the mobile app: antipyretic drugs in pediatric patients. Sci Rep 10(1):3879. https://doi.org/10.1038/s41598-020-60193-1

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Asante D-B, Henneh IT, Acheampong DO, Kyei F, Adokoh CK, Ofori EG, Domey NK, Adakudugu E, Tangella LP, Ameyaw EO (2019) Anti-inflammatory, anti-nociceptive and antipyretic activity of young and old leaves of Vernonia amygdalina. Biomed Pharmacother 111:1187–1203. https://doi.org/10.1016/j.biopha.2018.12.147

    Article  CAS  PubMed  Google Scholar 

  6. Ludwig J, McWhinnie H (2019) Antipyretic drugs in patients with fever and infection: literature review. Br J Nurs 28(10):610–618. https://doi.org/10.1016/j.biopha.2018.12.147

    Article  CAS  PubMed  Google Scholar 

  7. Adnan M, Chy MNU, Kamal ATMM, Chowdhury MR, Islam MS, Hossain MA, Tareq AM, Bhuiyan MIH, Uddin MN, Tahamina A, Azad MOK, Lim YS, Cho DH (2020) Unveiling pharmacological responses and potential targets insights of identified bioactive constituents of Cuscuta reflexa roxb. Leaves through in vivo and in silico approaches. Pharmaceuticals 13(3):50. https://doi.org/10.3390/ph13030050

    Article  CAS  PubMed Central  Google Scholar 

  8. Psarra A, Nikolaou A, Kokotou MG, Limnios D, Kokotos G (2017) Microsomal prostaglandin E2 synthase-1 inhibitors: a patent review. Expert Opin Ther Pat 27(9):1047–1059. https://doi.org/10.1080/13543776.2017.1344218

    Article  CAS  PubMed  Google Scholar 

  9. Tareq AM, Farhad S, Neshar Uddin ABM, Hoque M, Nasrin MS, Uddin MMR, Hasan M, Sultana A, Munira MS, Lyzu C, Moazzem Hossen SM, Ali Reza ASM, Emran TB (2020) Chemical profiles, pharmacological properties, and in silico studies provide new insights on Cycas pectinata. Heliyon 6(6):e04061. https://doi.org/10.1016/j.heliyon.2020.e04061

    Article  PubMed  PubMed Central  Google Scholar 

  10. Khan MF, Kader FB, Arman M, Ahmed S, Lyzu C, Sakib SA, Tanzil SM, Zim AFMIU, Imran MAS, Venneri T, Romano B, Haque MA, Capasso R (2020) Pharmacological insights and prediction of lead bioactive isolates of Dita bark through experimental and computer-aided mechanism. Biomed Pharmacother 131:110774. https://doi.org/10.1016/j.biopha.2020.110774

    Article  CAS  PubMed  Google Scholar 

  11. Adnan M, Nazim Uddin Chy M, Mostafa Kamal ATM, Azad MOK, Paul A, Uddin SB, Barlow JW, Faruque MO, Park CH, Cho DH (2019) Investigation of the biological activities and characterisation of bioactive constituents of Ophiorrhiza rugosa var. prostrata (D.Don) & Mondal leaves through in vivo, in vitro, and in silico approaches. Molecules 24(7):1367. https://doi.org/10.3390/molecules24071367

    Article  CAS  PubMed Central  Google Scholar 

  12. Rahman J, Tareq AM, Hossain MM, Sakib SA, Islam MN, Ali MH, Uddin ABMN, Hoque M, Samima Nasrin M, Emran TB, Capasso R, Reza ASMA, Simal-Gandara J (2020) Biological evaluation, DFT calculations and molecular docking studies on the antidepressant and cytotoxicity activities of Cycas pectinata Buch.-ham. Compounds. Pharmaceuticals 13(9):232–232. https://doi.org/10.3390/ph13090232

    Article  CAS  Google Scholar 

  13. Rangra N, Samanta S, Pradhan K (2019) A comprehensive review on phytopharmacological investigations of Acacia auriculiformis a.Cunn. Ex Benth. Asian Pac J Trop Biomed 9(1):1–11. https://doi.org/10.4103/2221-1691.250263

    Article  CAS  Google Scholar 

  14. Girijashankar V (2011) Micropropagation of multipurpose medicinal tree Acacia auriculiformis. J Med Plant Res 5(3):462–466. https://doi.org/10.5897/JMPR.9000895

    Article  CAS  Google Scholar 

  15. Mandal P, Sinha Babu SP, Mandal NC (2005) Antimicrobial activity of saponins from Acacia auriculiformis. Fitoterapia 76(5):462–465. https://doi.org/10.1016/j.fitote.2005.03.004

    Article  CAS  PubMed  Google Scholar 

  16. Sharma A, Shetty M, Parida A, Adiga S, Kamath S, Sowjanya (2014) Effect of ethanolic extract of Acacia auriculiformis leaves on learning and memory in rats. Pharm Res 6(3):246–250. https://doi.org/10.4103/0974-8490.132605

    Article  Google Scholar 

  17. Chew YL, Ling Chan EW, Tan PL, Lim YY, Stanslas J, Goh JK (2011) Assessment of phytochemical content, polyphenolic composition, antioxidant and antibacterial activities of Leguminosae medicinal plants in peninsular Malaysia. BMC Complement Altern Med 11(1):12. https://doi.org/10.1186/1472-6882-11-12

    Article  PubMed  PubMed Central  Google Scholar 

  18. Sathya A, Siddhuraju P (2013) Protective effect of bark and empty pod extracts from Acacia auriculiformis against paracetamol intoxicated liver injury and alloxan induced type II diabetes. Food Chem Toxicol 56:162–170. https://doi.org/10.1016/j.fct.2013.02.031

    Article  CAS  PubMed  Google Scholar 

  19. Meyer BN, Ferrigni NR, Putnam JE, Jacobsen LB, Nichols DE, McLaughlin JL (1982) Brine shrimp: a convenient general bioassay for active plant constituents. Planta Med 45(05):31–34. https://doi.org/10.1055/s-2007-971236

    Article  CAS  PubMed  Google Scholar 

  20. Guha B, Arman M, Islam MN, Tareq SM, Rahman MM, Sakib SA, Mutsuddy R, Tareq AM, Emran TB, Alqahtani AM (2020) Unveiling pharmacological studies provide new insights on Mangifera longipes and Quercus gomeziana. Saudi J Biol Sci 28(1):183–190. https://doi.org/10.1016/j.sjbs.2020.09.037

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Sabiu S, Ashafa AOT (2016) Membrane stabilisation and kinetics of carbohydrate metabolising enzymes (α-amylase and α-glucosidase) inhibitory potentials of Eucalyptus obliqua L.her. (Myrtaceae) Blakely ethanolic leaf extract: an in vitro assessment. S Afr J Bot 105:264–269. https://doi.org/10.1016/j.sajb.2016.04.007

    Article  CAS  Google Scholar 

  22. Adnan M, Chy MNU, Rudra S, Tahamina A, Das R, Tanim MAH, Siddique TI, Hoque A, Tasnim SM, Paul A, Kamal AM (2019) Evaluation of Bonamia semidigyna (Roxb.) for antioxidant, antibacterial, anthelmintic and cytotoxic properties with the involvement of polyphenols. Orient Pharm Exp Med 19(2):187–199. https://doi.org/10.1007/s13596-018-0334-x

    Article  CAS  Google Scholar 

  23. Prasad S, Kashyap RS, Deopujari JY, Purohit HJ, Taori GM, Daginawala HF (2006) Development of an in vitro model to study clot lysis activity of thrombolytic drugs. Thromb J 4(1):14. https://doi.org/10.1186/1477-9560-4-14

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Banu N, Alam N, Nazmul Islam M, Islam S, Sakib SA, Hanif NB, Chowdhury MR, Tareq AM, Hasan Chowdhury K, Jahan S, Azad A, Emran TB, Simal-Gandara J (2020) Insightful valorisation of the biological activities of pani heloch leaves through experimental and computer-aided mechanisms. Molecules 25(21). https://doi.org/10.3390/molecules25215153

  25. Ahmed S, Rakib A, Islam MA, Khanam BH, Faiz FB, Paul A, Chy MNU, Bhuiya NMMA, Uddin MMN, Ullah SMA, Rahman MA (2019) In vivo and in vitro pharmacological activities of Tacca integrifolia rhizome and investigation of possible lead compounds against breast cancer through in silico approaches. Clin Phytosci 5(1):36. https://doi.org/10.1186/s40816-019-0127-x

    Article  CAS  Google Scholar 

  26. Rakib A, Ahmed S, Islam MA, Haye A, Uddin SMN, Uddin MMN, Hossain MK, Paul A, Emran TB (2020) Antipyretic and hepatoprotective potential of Tinospora crispa and investigation of possible lead compounds through in silico approaches. Food Sci Nutr 8(1):547–556. https://doi.org/10.1002/fsn3.1339

    Article  CAS  PubMed  Google Scholar 

  27. Rangra N, Samanta S, Pradhan K (2019) In vivo antiinflammatory potential of leaf extracts of Acacia auriculiformis Benth. Indian J Pharm Sci 81(4):709–719. https://doi.org/10.36468/pharmaceutical-sciences.562

    Article  CAS  Google Scholar 

  28. Berman HM, Battistuz T, Bhat TN, Bluhm WF, Bourne PE, Burkhardt K, Feng Z, Gilliland GL, Iype L, Jain S, Fagan P, Marvin J, Padilla D, Ravichandran V, Schneider B, Thanki N, Weissig H, Westbrook JD, Zardecki C (2002) The protein data Bank. Acta Crystallogr D Biol Crystallogr 58(6 Part 1):899–907. https://doi.org/10.1107/s0907444902003451

    Article  PubMed  Google Scholar 

  29. Harman CA, Turman MV, Kozak KR, Marnett LJ, Smith WL, Garavito RM (2007) Structural basis of enantioselective inhibition of cyclooxygenase-1 by S-alpha-substituted indomethacin ethanolamides. J Biol Chem 282(38):28096–28105. https://doi.org/10.1074/jbc.M701335200

    Article  CAS  PubMed  Google Scholar 

  30. Kurumbail RG, Stevens AM, Gierse JK, McDonald JJ, Stegeman RA, Pak JY, Gildehaus D, Penning TD, Seibert K, Isakson PCJN (1996) Structural basis for selective inhibition of cyclooxygenase-2 by anti-inflammatory agents. Nature 384(6610):644–648. https://doi.org/10.1038/384644a0

    Article  CAS  PubMed  Google Scholar 

  31. Jegerschöld C, Pawelzik S-C, Purhonen P, Bhakat P, Gheorghe KR, Gyobu N, Mitsuoka K, Morgenstern R, Jakobsson P-J, Hebert H (2008) Structural basis for induced formation of the inflammatory mediator prostaglandin E2. Proc Natl Acad Sci U S A 105(32):11110–11115. https://doi.org/10.1073/pnas.0802894105

    Article  PubMed  PubMed Central  Google Scholar 

  32. Brzozowski AM, Pike ACW, Dauter Z, Hubbard RE, Bonn T, Engström O, Öhman L, Greene GL, Gustafsson J-Å, Carlquist M (1997) Molecular basis of agonism and antagonism in the oestrogen receptor. Nature 389(6652):753–758. https://doi.org/10.1038/39645

    Article  CAS  PubMed  Google Scholar 

  33. Ravelli RBG, Gigant B, Curmi PA, Jourdain I, Lachkar S, Sobel A, Knossow M (2004) Insight into tubulin regulation from a complex with colchicine and a stathmin-like domain. Nature 428(6979):198–202. https://doi.org/10.1038/nature02393

    Article  CAS  PubMed  Google Scholar 

  34. Renatus M, Bode W, Huber R, Stürzebecher J, Prasa D, Fischer S, Kohnert U, Stubbs MT (1997) Structural mapping of the active site specificity determinants of human tissue-type plasminogen activator. Implications for the design of low molecular weight substrates and inhibitors. J Biol Chem 272(35):21713–21719. https://doi.org/10.1074/jbc.272.35.21713

    Article  CAS  PubMed  Google Scholar 

  35. Lipinski CA, Lombardo F, Dominy BW, Feeney PJ (1997) Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Deliv Rev 23(1):3–25. https://doi.org/10.1016/S0169-409X(96)00423-1

    Article  CAS  Google Scholar 

  36. Cheng F, Li W, Zhou Y, Shen J, Wu Z, Liu G, Lee PW, Tang Y, admetSAR (2012) A comprehensive source and free tool for assessment of chemical ADMET properties. J Chem Inf Model 59(11):4959. https://doi.org/10.1021/ci300367a

    Article  CAS  Google Scholar 

  37. Kaur K, Arora S, Hawthorne ME, Kaur S, Kumar S, Mehta RGA (2002) A correlative study on antimutagenic and chemopreventive activity of Acacia auriculiformis a. Cunn. And Acacia nilotica (L.) Willd. Ex Del. Drug Chem Toxicol 25(1):39–64. https://doi.org/10.1081/dct-100108471

    Article  CAS  PubMed  Google Scholar 

  38. Acacia auriculiformis herb uses, benefits, cures, side effects, nurients reperoty [Online] [https://herbpathy.com/Uses-and-Benefits-of-Acacia-Auriculiformis-Cid6574]

  39. Jyoti MA, Barua N, Hossain MS, Hoque M, Bristy TA, Mahmud S, Kamruzzaman AM, Chy MNU, Paul A, Hossain ME, Emran TB, Simal-Gandara J (2020) Unravelling the biological activities of the Byttneria pilosa leaves using experimental and computational approaches. Molecules 25(20):4737. https://doi.org/10.3390/molecules25204737

    Article  CAS  PubMed Central  Google Scholar 

  40. Middleton E (1998) Effect of plant flavonoids on immune and inflammatory cell function. In: Manthey JA, Buslig BS (eds) Flavonoids in the living system, vol 439. Springer US, Boston, pp 175–182. https://doi.org/10.1007/978-1-4615-5335-9_13

    Chapter  Google Scholar 

  41. Manthey JA, Guthrie N, Grohmann K (2001) Biological properties of citrus flavonoids pertaining to cancer and inflammation. Curr Med Chem 8(2):135–153. https://doi.org/10.2174/0929867013373723

    Article  CAS  PubMed  Google Scholar 

  42. Issa AY, Volate SR, Wargovich MJ (2006) The role of phytochemicals in inhibition of cancer and inflammation: new directions and perspectives. J Food Compos Anal 19(5):405–419. https://doi.org/10.1016/j.jfca.2006.02.009

    Article  CAS  Google Scholar 

  43. Chy MNU, Chakrabarty N, Roy A, Paul A, Emu KA, Dutta T, Dutta E, Ferdous I, Das R, Hasan MJ, Tasnim SM (2019) Antibacterial, anthelmintic, and analgesic activities of Piper sylvaticum (Roxb.) leaves and in silico molecular docking and PASS prediction studies of its isolated compounds. J Complement Integr Med 16(4). https://doi.org/10.1515/jcim-2018-0176

  44. Uddin MS, Millat MS, Islam MS, Hussain MS, Uddin MG, Siddiqui SA, Ferdous M (2020) Exploration of in vitro thrombolytic, anthelminthic, cytotoxic and in vivo anxiolytic potentials with phytochemical screening of flowers of Brassica nigra. Futur J Pharm Sci 6(1):73. https://doi.org/10.1186/s43094-020-00099-x

    Article  Google Scholar 

  45. Jia Q, Zhao Y (2006) Formulation of a mixture of free-B-ring flavonoids and flavans as a therapeutic agent. In: U.S. patent application 11/373,576

    Google Scholar 

  46. Mworia JK, Kibiti CM, Ngugi MP, Ngeranwa JN (2019) Antipyretic potential of dichloromethane leaf extract of Eucalyptus globulus (Labill) and Senna didymobotrya (Fresenius) in rats models. Heliyon 5(12):e02924. https://doi.org/10.1016/j.heliyon.2019.e02924

    Article  PubMed  PubMed Central  Google Scholar 

  47. Muhammad A, Khan B, Iqbal Z, Khan AZ, Khan I, Khan K, Alamzeb M, Ahmad N, Khan K, Lal Badshah S, Ullah A, Muhammad S, Tariq Jan M, Nadeem S, Kabir N (2019) Viscosine as a potent and safe antipyretic agent evaluated by yeast-induced pyrexia model and molecular docking studies. ACS Omega 4(10):14188–14192. https://doi.org/10.1021/acsomega.9b01041

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Dutta T, Paul A, Majumder M, Sultan RA, Emran TB (2020) Pharmacological evidence for the use of Cissus assamica as a medicinal plant in the management of pain and pyrexia. Biochem Biophys Rep 21:100715. https://doi.org/10.1016/j.bbrep.2019.100715

    Article  PubMed  Google Scholar 

  49. Zhao F, Wang P, Lucardi RD, Su Z, Li S (2020) Natural sources and bioactivities of 2,4-di-tert-butylphenol and its analogs. Toxins 12(1). https://doi.org/10.3390/toxins12010035

Download references

Acknowledgements

Authors are very much thankful to the Department of Pharmacy, International Islamic University Chittagong, Bangladesh for providing research facilities and other logistic supports.

Plant authentication

Acacia auriculiformis leaves were collected and identified from Rangamati region and authenticated by a taxonomist under the accession number Anwar-0870.

Funding

This work is conducted with the individual funding of all authors.

Author information

Authors and Affiliations

Authors

Contributions

SN and MMH planned and designed the research. RK arranged the whole facilities for the research and supervised the whole research. SN, MMH and NI conducted the entire laboratory works with NBH and AMT. SN, NI, AMT and NBH imparted in study design and interpreted the results putting efforts on statistical analysis. The computational study carried out by MMH, AMT and SN. MMH, SN, AMT, NI and MAS participated in the manuscript draft and has thoroughly checked and revised the manuscript for necessary changes in format, grammar and English standard. MAS critically reviewed the manuscript. All authors read and agreed on the final version of the manuscript.

Corresponding author

Correspondence to Mohammed Abu Sayeed.

Ethics declarations

Ethics approval and consent to participate

The Swiss albino male mice of were procured from the Jahangirnagar University, Dhaka-1342, Bangladesh. The study approved by the Institutional Animal Ethical Committee, Department of Pharmacy, International Islamic University Chittagong, Bangladesh according to governmental guidelines under the reference of Pharm/P&D/AEC-147/13-19. For ethical reasons, each animal was used only once and all animals were sacrificed at the end of the study.

Consent for publication

Not applicable.

Competing interests

The authors declared that they have no conflict of interest.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Nur, S., Hossain, M.M., Islam, N. et al. Scrutinizing pharmacological efficiency for Acacia auriculiformis by experimental and computational approach. Futur J Pharm Sci 7, 71 (2021). https://doi.org/10.1186/s43094-021-00221-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s43094-021-00221-7

Keywords