This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
1. Introduction
Diabetes mellitus (DM) is a category of metabolic disorders that affect glucose, lipid, and protein metabolism and consequently affect the overall health status [1, 2]. The prevalence of diabetes is very high, affecting millions of people globally [3]. It is characterized by the development of insulin resistance, abnormal insulin signaling, oxidative stress [4], inflammation [5], and organ dysfunction [6], leading to decreased life quality and high mortality [7]. In the past few decades, DM, particularly type 2 DM (T2DM), has become a global health problem that threatens millions of people in both developed and developing countries [2]. According to the Diabetes Atlas (10th edition) of the International Diabetes Federation (IDF) [8], there are 537 million people living with diabetes in 2021, and about 783 million cases are expected by 2045 [8], compared to 151 million sufferers in 2000 [9–11].
Although the pathogenesis and pathophysiology of T2DM are extremely complex and not fully understood, accumulating evidence has revealed that free radical generation, oxidative stress, and inflammation play critical implicative roles in the development of T2DM [12, 13]. Inflammation, a complex physiological response to injury and infection, plays a pivotal role in the development of chronic disorders, including arthritis, asthma, atherosclerosis, and cancers [14]. Experimental and clinical studies have provided evidence of the important roles of inflammation in DM [15, 16]. Reactive nitrogen species (RNS) and reactive oxygen species (ROS) were clearly implicated in the pathology of various diseases including aging, cancers, cardiovascular diseases (CVD), neuronal impairment, and diabetic complications [14, 17]. During hyperglycemia, the generation of free radicals (RNS and ROS), autooxidation of glucose, and depletion of endogenous antioxidants lead to oxidative stress and inflammatory conditions, apoptosis of pancreatic islet β cells, and impaired insulin secretion [18]. In addition, inflammation and oxidative stress also contribute to the development of diabetic complications such as hypertension, retinopathies, nephropathies, and neuropathies [19, 20].
Over the years, synthetic and chemical antioxidants and antidiabetic medications have been developed for treating diabetes and its associated complications [21]. However, their clinical applications have been limited by the loss of efficacy and more importantly by associated side effects including diarrhea, lactic acidosis, flatulence, and acute hepatitis [22, 23]. Hence, there is a need to focus on seeking new alternatives. Moreover, evidence from traditional health practice and experimental studies suggested that natural products, particularly medicinal plants, are rich sources of therapeutic agents that can offer better efficacy with minimal side effects compared to conventional therapies [24–26]. Thus, exploring bioactive metabolites from medicinal plants may offer multieffect glycemic control while modulating oxidoinflammatory aberrations and improving prognoses of diabetes and its complications.
Thespesia garckeana F. Hoffm., known as ‘snot apple’ or ‘kola of Tula,’ is a reputable medicinal plant that is widely distributed in Africa and some other tropical countries [27, 28]. As a traditional medicine, the plant is widely used for treating rheumatism, infections, diabetes, liver diseases, reproductive impairment, and other diseases [28, 29]. Several biological activities of the plant were also reported in the literature [28, 30–32]. Herein, we evaluated the hypoglycemic, antioxidant, and anti-inflammatory effects of the diethyl ether fraction of T. garckeana using various in vitro and in vivo models. Further characterization of the extract revealed the presence of various compounds which were subjected to a pharmacoinformatics analysis, thus unveiling their drug-likeness, good pharmacokinetics (PKs), and potential hypoglycemic properties. Altogether, our study established the preclinical efficacy of T. garckeana extract against inflammation, glycemic impairment, and oxidative stress, suggesting its future use for developing alternative therapies against diabetes complications.
2. Materials and Methods
2.1. Plant Collection, Extraction, and Fractionation
Thespesia garckeana plant was obtained from the Gombe State, Tula Village, Kaltungo Local Government Area (Latitude 9°48'51″N and Longitude11°18'32″E) Nigeria. Plant authentication was conducted at the Ebonyi State University, Nigeria, and a specimen voucher identification number was reported. The pulp was air-dried to a constant dry weight at 25°C for 2 weeks before been subjected to pulverization with the aid of a mechanical blending machine. Three hundred grams (300 g) of the pulverized sample was subjected to cold maceration in 1.5 L of methanol for 72 h. The resulting crude extract after concentration was further subjected to fractionation with diethyl ether, ethyl acetate, and n-butanol successively and the resulting fractions; diethyl ether (7.9% yield), ethyl acetate (3.10% yield), and n-butanol (2.45% yield) fractions were concentrated in a rotary vacuum evaporator under reduced pressure. The resulting diethyl ether fraction (dietl-eth_T. garckeana) which demonstrated the highest yield and phytochemical compositions in preliminary screening was used for subsequent analysis.
2.2. Analysis of Total Phenol and Flavonoid Contents
The total flavonoid content of the dietl-eth fraction of T. garckeana was determined according to the method of Chang et al. [33], while the total phenol content was evaluated using the Folin-Ciolcalteau’s reagent as described by Singleton et al. [34]. Gallic acid (total phenols) and quercetin (total flavonoids) were used to prepare the calibration curves.
2.3. In Vitro Antioxidant Assays
The in vitro antioxidant effects of dietl-eth_T. garckeana were evaluated using the DPPH, FRAP, ABTS, and LPO assays. The DPPH assay was conducted based on the scavenge ability of the extract on DPPH radicals [35]. FRAP activity was assayed according to the method of Oyaizu [35]. The extract was incubated in K3[Fe(CN)6] supplemented phosphate buffer at 50°C for 20 min, after which 10% TCA was added. The resulting solution was centrifuged and color development in the presence of 0.1% ferric chloride was monitored at 700 nm. The thiobarbituric acid-reactive substance (TBARS) protocol described by Panjamurthy et al. [36] was employ for the LPO analysis, while the ABTS assay was done as reported by Re et al. [37].
2.4. In Vitro Hypoglycemic Assays
Standard analysis protocol for alpha-amylase inhibition described by Worthington [38] was employed for the in vitro hypoglycemic analysis of the extract. Briefly, the extract/acarbose (12.5 ~ 100 μg/mL) mixed in an enzymatic porcine pancreatic solution was incubation with a starch solution for 30 min at 37°C, followed by the addition of 10 μL of HCl (1 M) and iodine reagent for color development. In the analysis of glucose uptake by yeast cells, glucose solution (5 ~ 25 mM) was incubated (37°C for 10 min) with the extract. A yeast suspension was added to the reaction mixture and incubated for another 45-60 min, after which glucose concentration was estimated.
2.5. In Vitro Anti-Inflammatory Assays
The in vitro anti-inflammatory activities of the dietl-eth_T. garckeana were evaluated by using the human red blood cell (RBC and HRBC) membrane stabilization, inhibition of protein denaturation, and proteinase inhibitory assays. The protein denaturation inhibition was assayed as described of Mizushima and Kobayashi [39], while the method of Oyedepo and Femurewa [40] was used for the proteinase inhibition assay. In the HRBC assay, a 10% human RBC suspension was incubated with the extract solution at 50-55°C for 30 min [41]. The resulting solution was centrifuged at 2500 rpm for 5 min, and the reaction changes was monitored at 560 nm.
2.6. Analysis of Toxicity and Extract Tolerated Dose
The dietl-eth_T. garckeana was subjected to an acute toxicity study to evaluate its safety and maximum tolerated dose upon oral administration according to the protocol described by Lorke [42]. Briefly, a total of eighteen (18) rats were grouped into six (6) and administered with 10, 100, 1000, 1600, 2800, and 5000 mg/kg bw of dietl-eth_T. garckeana to groups I-VI, respectively. The extract was administered once orally, after which the rats were observed for any sign of adverse toxicity and mortality within 14 days’ period.
2.7. Oral Glucose Tolerance Test (OGTT)
The OGTT was conducted according to the method described by Sisay et al. [43]. A total of twelve (12) overnight starved rats were grouped into 5 (3 rats per group) and treated with 2.0 mL/kg normal saline, 5 mg/kg glibenclimide, 150 mg/kg, and 300 mg/kg dietl-eth_T. garckeana for groups I-IV, respectively. After 30 minutes of treatment, 2 g/kg bw of glucose was orally administered to rats in each group. The blood glucose level was determined at 0 minutes (basal blood glucose level) and at intervals of 30 for a period of 2 hrs.
2.8. Analysis of In Vivo Antidiabetic Activities in Rats
The dietl-eth_T. garckeana was evaluated for the in vivo antidiabetic effects in albino rats. Experimental rats were acquired from the experimental rodent facility of AE-FUNAI, Nigeria. Animal experiments were consented by the ethical committee of the AE-FUNAI, Nigeria. Streptozotocin (STZ; 40 mg/kg) was intraperitoneally administered to overnight starved rats, and rats with
2.9. In Vivo Anti-Inflammatory Analysis (Xylene-Induced Ear Swelling Assay)
The dietl-eth_T. garckeana was evaluated for its in vivo anti-inflammatory effects in mice using a xylene-induced ear edema test [45, 46]. Healthy mice were divided into five (
2.10. Collection and Processing of Samples
Experimental rats were euthanized under diethyl ether vapor for few minutes, and blood were obtained through cardiac puncture. The coagulated blood was centrifuged for 15 min (3000 × g) and the separated serum were stored at 4°C [47, 48]. The rat’s pancreases and liver tissues were harvested, homogenized in phosphate buffer (0.1 M, pH 7.4, 1 : 10
2.11. Tissue Assays for Antioxidant Parameters
The in vivo antioxidant effects of the extract were evaluated in liver homogenate using the lipid peroxidation (LPO), superoxide dismutase (SOD), reduced glutathione (GSH), and catalase (CAT) assays. The LPO assay was conducted by measuring the tissue levels of TBARSs [49], while the modified colorimetric method of Kum-Tatt and Tan [50] was adopted for GSH assay. The activity of SOD was evaluated by using an established protocol [51]. The tissue homogenate was incubated with carbonate buffer (pH 10.2) in the presence of 0.3 mM adrenaline. The increase in absorbance at an interval of 30 s were monitored at 480 nm [51]. CAT activity was determined using a standard protocol [52]. The liver homogenate (0.1 mL) in phosphate buffer (0.01 M, pH 7.0) was incubated with 2 M hydrogen peroxide (H2O2) for 20 min. This was followed by the addition of a dichromate acetate reagent to terminate the reaction. The changes in absorbances were monitored at the wavelength of 620 nm and the activity of CAT was expressed as a unit of H2O2/mg protein [52].
2.12. Measurement of Insulin Levels
The Insulin ELISA kit (Calbiochem-Behring Corp, CA; catalog no. IN374S) was used for the analysis of pancreatic and serum insulin concentrations as per the manual instructions.
2.13. High-Performance Liquid Chromatography (HPLC)
The dietl-eth_T. garckeana was subjected to HPLC analysis according to the method described in our previous study [20]. A hundred milligram (100 mg) of dietl-eth_T. garckeana was dissolved in five millilitres (5 mL) of an HPLC grade methanol. The extract solution was filtered and run on the HPLC (Agilent Technologies 1200) with the following chromatographic conditions: stationary phase (Hypersil BDS C18), mobile phase (acetonitrile and 0.1% formic acid), column dimension of
2.14. Pharmacoinformatics Analysis
2.14.1. Analysis of Drug-Likeness and ADMET-PK Properties
The most abundant compounds identified from the dietl-eth_T. garckeana were subjected to analysis of physicochemical properties, drug-likeness, PKs (ADMET), and medicinal chemistry using the ADMET-Lab, ADMETSar, and Swiss-ADME servers [53]. The human-intestinal absorption and permeability by blood-brain barrier (BBB) were modeled through the BOILED-EGG and support vector machine (SVM) tools [53].
2.14.2. Analysis of Ligand Receptor Interactions Using Molecular Docking
The most abundant bioactive compound characterized from the extract was subjected to an analysis of ligand-receptor interactions using molecular docking. The crystal three-dimensional (3D) forms of the compounds were built by the Avogadro visualization tool (version 1.XX) [54], while the PDB files of the target receptors including alpha-amylase and alpha-glucosidase were downloaded from the Protein Data Bank. The mol2 files were transformed into PDB files using PyMOL software, while the PDB formats were transformed to PDBQT using AutoDock Vina [55]. Water molecules were detached, while hydrogen atoms in polar forms and Kollman charges were added to the compounds during predocking preparations [56, 57]. The version 8 of AutoDock Vina tool was used for the receptor-ligand docking as described in previous studies [56, 58, 59], while visualization was conducted using the PyMOL and Discovery studio tools [60].
2.15. Data Analysis
The GraphPad vers. 8.0 software was used for the statistical analysis of replicate data. The one-way type analysis of variance and Student’s
3. Results and Discussion
3.1. Thespesia garckeana Fraction Exhibited Dose-Related In Vitro Antioxidant Activities
Plants, particularly those that are rich source of flavonoid and polyphenolic compounds, have been effective against various diseases including cancers and diabetes [61]. These phytochemicals are known to exhibit several biological activities including antioxidants, antimicrobial, anticancer, anti-inflammatory, and antidiabetic, etc. [62]. Interestingly, our results demonstrated that dietl-eth_T. garckeana contains total phenol contents of
The formation of free radicals and oxidative stress play important roles implicated in the development of T2DM [12, 13]. Therefore, the role of antioxidants in preventing the formation of free radicals is crucial to the control of DM [61]. Our in vitro studies revealed significant and dose-related antioxidant activities of the dietl-eth_T. garckeana in four different in vitro models of antioxidant assays; DPPH, FRAP, LPO, and ABTS, with IC50 values of
[figure(s) omitted; refer to PDF]
3.2. Thespesia garckeana Fraction Exhibited Dose-Related In Vitro Anti-Inflammatory Activities
Inflammation, a complex physiological response to injury and infection, plays a pivotal role in the development of chronic disorders, including arthritis, asthma, atherosclerosis, cancer, and DM [14]. Interestingly, T. garckeana exhibited dose-related anti-inflammatory activities. Our in vitro anti-inflammatory studies using membrane stabilization, protein denaturation, and proteinase activities revealed the effectiveness of the extract with respective IC50 values of
[figure(s) omitted; refer to PDF]
Proteinase inhibitory activity was studied to further explain the anti-inflammatory mechanism of dietl-eth_T. garckeana. Neutrophils contains high levels of serine proteinase [65] that plays a vital tissue damaging role during inflammatory events [68] and inhibitors of this proteinase protect against the damage [69]. Interestingly, the dietl-eth_T. garckeana exhibited dose related antiproteinase activity with IC50 of
3.3. Thespesia garckeana Fraction Demonstrated Safety Profile in Acute Oral Toxicity Study
According to the data obtained from this study, dietl-eth_T. garckeana demonstrated a good safety profile with a
Table 1
Acute oral toxicity profiles of diethyl-ether fraction of Thespesia garckeana in rats.
dietl-eth_T. garckeana (mg/kg) | Mortality | Adverse effect | Two weeks’ posttreatment observation (loss of weight, sign of toxicity) |
10 | 0/3 | Nil | Nil |
100 | 0/3 | Nil | Nil |
1000 | 0/3 | Nil (MTD) | Nil |
1600 | 0/3 | Restlessness (10-15 mins) | Nil |
2900 | 0/3 | Rubbing of the mouth on the wall of the cage (about 20 mins); restlessness (about 20 mins) | Nil |
5000 | 0/3 | Rubing of cages (20-30 mins), redness of the eye (30 mins), restlessness (30 mins), fur erection (30 mins), profuse breathing (24 h), weakness (only in day 2) | Nil |
MTD: maximum tolerated dose.
3.4. Thespesia garckeana Fraction Exhibited Dose-Related In Vitro Hypoglycemic Activities
The inhibition of carbohydrate-metabolizing enzymes and regulation of blood glucose levels are very critical to the management of DM [72]. The dietl-eth_T. garckeana also demonstrated a hypoglycemic effect via inhibition of α-amylase (IC50:
[figure(s) omitted; refer to PDF]
Inhibitors of α-amylase, a starch-catabolizing enzyme, are widely used as oral hypoglycemic agents for the regulation of sugar levels of T2DM patient [73]. However, most of these inhibitors are synthetic with of off-limit activity and associated side effects [74]. Consequently, the current results verify that the dietl-eth_T. garckeana exerts its hypoglycemic via the inhibition of carbohydrate-catabolizing enzymes, including the α-amylase. The inhibitory activities of the dietl-eth_T. garckeana on α-amylase activities suggests the attenuation of postprandial blood glucose increase by decreasing the carbohydrates flow into the bloodstream after carbohydrate intake [75].
Glucose is preferred by yeast as a primary source of fuel, and glucose absorption by yeast cells is simulate that occurring in the mammalian intestinal lumen; thus, yeast has become a commonly used model to study glucose absorption [76]. According to the present study, dietl-eth_T. garckeana significantly enhanced glucose uptake by yeast cells in a dose-related manner. This is important for the efficient utilization and control of glucose levels. Interestingly, a linear relationship between glucose concentration and rates of glucose uptake by the yeast cells was observed. This is in line with the finding of Keshala et al. [61] who reported a concentration-dependent increase in glucose uptake by yeast cells in the presence of plant extract. Collectively, the dietl-eth_T. garckeana exhibited dose-related in vitro hypoglycemic activities and, thus, could be regarded as a natural product with potential for the management of DM.
3.5. The dietl-eth_T. garckeana demonstrated hypoglycemic effect in Oral glucose tolerance test
In the present study, the evaluation of dietl-eth_T. garckeana for possible antidiabetic effect was also conducted using the oral glucose tolerance test. There were initial increases in glucose levels at 30 minutes of glucose dosing after which progressive decreases were observed from 30 minutes to 2 hours in all the treatment as well as the control rats. However, significant (
[figure(s) omitted; refer to PDF]
3.6. The dietl-eth_T. garckeana Demonstrated Hypoglycemic Effect and Improved Insulin Secretion in STZ-Induced Diabetic Rats
Previous experimental studies have proved the therapeutic efficacy of medicinal plants in animal models of diabetes, inflammation, and oxidative stress-associated diseases [79–81]. The in vivo antidiabetic effects of the dietl-eth_T. garckeana was evaluated in STZ-induced diabetic rats. Interestingly, our in vitro findings corroborated with the data generated in vivo. We found that treatment of the STZ-induced diabetic rats with dietl-eth_T. garckeana caused significant and progressive decreases in the fasting blood sugar (FBS) levels in a dose-related manner (Figure 5(a), Table 2) and prevented the body weight loss (Figure 5(b), Table 3), while diabetic untreated rats exhibited progressive increases in FBS levels and body weight loss. At the end of the treatment duration, the extract at 150 and 300 mg/kg demonstrated 62.67% and 78.88% hypoglycemic effects, respectively, while metformin demonstrated higher hypoglycemic activity of 84.25%. Thus, the gradual decreases in FBS and improvement of body weight recorded in the T. garckeana-receiving animal when compared with the untreated diabetic rats presaged the ameliorative effects of the fraction on experimentally induced diabetes.
[figure(s) omitted; refer to PDF]
Table 2
Effect of diethyl-ether fraction of Thespesia garckeana on blood glucose levels in STZ-induced diabetic rats.
Groups | 0 | 3 | 7 | 14 | 21 | Glucose reduction (%) |
Normal |
|
|
|
|
|
— |
Untreated |
|
|
|
|
|
— |
150 mg/kg Dietl_T.G |
|
|
|
|
|
62.67 |
300 mg/kg Dietl_T.G |
|
|
|
|
|
78.88 |
Metformin |
|
|
|
|
|
84.25 |
Dietl_T.G: diethyl-ether fraction of Thespesia garckeana.
Table 3
Effect of diethyl-ether fraction of Thespesia garckeana on body weight gain in STZ-induced diabetic rats.
Groups | 0 | 3 | 7 | 14 | 21 | Weight gain (%) |
Normal |
|
|
|
|
|
|
Untreated |
|
|
|
|
|
|
150 mg/kg Dietl_T.G |
|
|
|
|
|
|
300 mg/kg Dietl_T.G |
|
|
|
|
|
|
Metformin |
|
|
|
|
|
|
Dietl_T.G: diethyl-ether fraction of Thespesia garckeana.
Under a physiological condition, the pancreatic cells regulate blood glucose levels by regulating β cells of Islets of Langerhans’s activity through insulin secretions. Therefore, the hyperglycemia induced by STZ may be attributed to the impairment of insulin release as a consequence of the destroyed β cells of Islets of Langerhans in the pancreas [82]. Indeed, we found that the STZ-induced diabetic nontreated rats exhibited significant (
3.7. The dietl-eth_T. garckeana Exhibited Antioxidant Activities in Rats with STZ-Induced Diabetes
Inflammation and oxidative stress contribute to the development of diabetic complications such as hypertension, retinopathies, nephropathies, and neuropathies [19, 20]. The generation levels of ROS are controlled by the levels endogenous antioxidant including CAT, GSH, and SOD [84, 85]. In the present study, analysis of liver biochemical parameters of oxidative stress in rats with STZ-induced diabetes revealed significant decreases in the liver activities of antioxidant enzymes including SOD (
[figure(s) omitted; refer to PDF]
The liver is a central detoxification organ of the body and plays a vital role in regulating glucose homeostasis [86]. As a group of insulin-sensitive tissues, the liver is among the primary organs highly susceptible to the effects of hyperglycemia-provoked oxidative stress, which may impair liver integrity [87]. Hyperglycemia, mainly caused by insulin resistance, induces the generation of free radicals by the activated Kupffer cells (phagocytic hepatic macrophages) that help in maintaining the integrity of liver cells [88]. However, these cells are highly susceptible to the effects of the free radicals generated by their own immune reactions and the surrounding cells [89]. The excessive ROS production results in irreversible oxidative alterations of macromolecules including the carbohydrates, lipids, and proteins [90], thereby leading to increased oxidative stress and triggering the cascade of inflammatory events that activates the transcription of proapoptotic genes and damages hepatocytes [91, 92].
Therefore, the reduced SOD and CAT activities in the liver of STZ-induced diabetic rats may be associated with the free radical generations which inturm decreases the activities of these enzymes. GSH is a cellular defense antioxidant molecule that protects against the progressive destruction of the ß cell [93]. The increases in free radicals’ productions in the diabetic rat result in oxidative damage to membrane lipids and proteins and eventually causes a decrease in the levels of GSH, CAT, and SOD as recorded in rats with STZ-induced diabetes. However, our results revealed that treatment with the dietl-eth_T. garckeana attenuated depleted levels of GSH, CAT, and SOD and decreased the LPO. These observations validate the potential of dietl-eth_T. garckeana in preventing free radical generation and maintaining the antioxidant status of diabetic rats [93]. This finding corroborated with literature on oxidative stress-alleviating properties of plant extracts in hyperglycemic rodent [94]. Our findings, therefore, revealed the T. garckeana’s ability to ameliorate oxidative impairment and restore antioxidant status of rats in hyperglycemic condition.
3.8. The dietl-eth_T. garckeana Demonstrated In Vivo Anti-Inflammatory Activities in Xylene-Induced Ear Swelling of Mice
An in vivo analysis of the anti-inflammatory properties using a xylene-induced ear swelling model in mice revealed that the dietl-eth_T. garckeana significantly (
Table 4
In vitro anti-inflammatory effects of the diethyl-ether fraction of Thespesia (dietl-eth_T.) garckeana.
Treatment | Dose (mg/kg BW) | Swelling rate | WBCs (×109) |
dietl-eth_T. garckeana | 150 |
|
|
dietl-eth_T. garckeana | 300 |
|
|
Aspirin | 200 |
|
|
Untreated control | 2.5 mL/normal saline |
|
|
Normal control | Normal control | — |
|
Values are the
3.9. HPLC Characterization of the dietl-eth_T. garckeana
To characterized the dietl-eth_T. garckeana we conducted an HPLC analysis (Figure 7) and identified the presence of catechin (6.81e-1 ppm), rutin (8.46 e-1 ppm), myricetin, apigenin (4.019 e-1 ppm), and luteolin (15.09 ppm) with respective retention times (RTs) of 13.64, 24.269, 27.781, 29.58, and 32.23 min. HPLC chromatograms and chemical structures of the compounds are displayed in Figure 7. Luteolin appeared to be the most abundant compound in the dietl-eth_T. garckeana, and thus, we evaluated its prospective for targeting glucose metabolizing enzymes and an inflammatory mediator.
[figure(s) omitted; refer to PDF]
3.10. Drug-ADMET and Likeness Modeling of Compounds Identified from the dietl-eth_T. garckeana
The goal of modern drug discovery and development is to identify a drug candidate with desirable properties within the shortest possible period of time and to avoid time- and cost-consuming approaches which in most cases produces disappointing outcome in the clinics [95, 96]. Hence, drug-likeness and PK analysis is considered an important aspect of the modern drug discovery and development. Our modeling analysis of the drug-PK, and drug-likeness revealed that apigenin, myricetin, luteolin, and catechin identified from the dietl-eth_T. garckeana (Figures 8(a) and 8(b)) were potential drug-like molecules. These compounds passed the drug absorbability test and have desirable bioavailability attribute. P-glycoproteins (P-gp) are responsible for propelling compounds and drugs out of the cells [97]. The identified compounds from the dietl-eth_T. garckeana are nonsubstrate or inhibitors of Pgp, thus suggesting their stability and optimal drug delivery [98]. This was also evident by their absorption and permeability record. The high volume of distribution and low binding of plasma protein by the drug further ascertain the good bioavailability and hinted at the potential good therapeutic index of the compounds but were non-BBB permeant (except for catechin) (Figure 8(b), Table 5). Cytochrome P450 (CYP450) is a heme containing enzyme family that play central metabolic role on exogenous and endogenous substances [99]. Hence, impeding the activities of these enzyme isoforms may lead to deficient drug metabolism and toxic drug accumulation. Providentially, our data indicated that among the 5 isoforms analyzed, the compounds were nonsubstrate and had inhibitor tendencies for cytochrome P450 1A2 (CYP1A2) and CYP3A4. However, they were nonsubstrate nor inhibitors of CYP2C9, CYP2D6, and CYP2C19. Notwithstanding, luteolin demonstrated its ability to be a substrate for P450 CYP3A4 and CYPC19, while catechin demonstrated its ability to be a substrate for CYP2D6 (Table 5). The presence of these isoforms in the liver and intestines indicates that these organs are sites of clearance of the compounds. Among the five compounds, luteolin and catechin demonstrated the best and most similar half-lives of 0.745 and 0.720 h, high clearance rates of 1.919 and 1.914 mL/min/kg, and a high safety profile with LD50 values of 737.444 and 860.605 mg/kg, respectively. Interestingly, luteolin and catechin demonstrated nontoxic attributes; nonhuman ether go-go-related gene (hERG) blockers, nonhepatotoxic, and were nonirritants in Skinsen assays. Collectively, our analysis revealed that among the five compounds identified, luteolin and catechin exhibited the best drug-PK and drug-likeness characteristic and, thus, were used for receptor-ligand simulation analysis.
[figure(s) omitted; refer to PDF]
Table 5
Pharmacokinetic_drug-likeness characteristic of the compounds from the diethyl-ether fraction of Thespesia garckeana.
Property | Rutin | Myricetin | Apigenin | Luteolin | Catechin | |
Physicochemical | Molecular weight | 610.521 | 318.237 | 270.24 | 286.239 | 290.271 |
Log |
-1.687 | 1.694 | 2.57 | 2.282 | 1.546 | |
HB acceptor | 16 | 8 | 5 | 6 | 6 | |
HB donor | 10 | 6 | 3 | 4 | 5 | |
TPSA | 269.43 | 151.59 | 90.9 | 111.13 | 110.38 | |
LogS (solubility) | 315.283 μg/mL | 191.757 μg/mL | 68.667 μg/mL | 90.101 μg/mL | 273.403 μg/mL | |
LogD7.4 | 0.992 | -0.068 | 0.487 | 0.302 | 0.115 | |
Log |
-1.687 | 1.694 | 2.577 | 2.282 | 1.546 | |
Absorption | Papp (colorectal adenocarcinoma permeability) | -6.606 cm/s | -6.63 cm/s | -4.985 cm/s | -5.123 cm/s | -4.637 cm/s |
P-glycoprotein-inhibitor | -(0.365) | ---(0.208) | -(0.328) | -(0.366) | ---(0.223) | |
P-glycoprotein-substrate | ---(0.168) | ---(0.064) | ---(0.023) | ---(0.038) | ---(0.045) | |
Intestinal absorption (HIA) | ---(0.21) | +(0.438) | +(0.531) | -(0.438) | -(0.401) | |
Bioavailability | +(0.572) | +(0.557) | +(0.542) | +(0.557) | -(0.487) | |
Distribution | Plasma protein binding (PPB) | 76.65% | 76.595% | 90.031% | 91.796% | 93.86% |
VD (L/kg) | -1.052 | -1.391 | -0.578 | -0.934 | -0.67 | |
Blood brain barrier | ---(0.018) | -(0.369) | -(0.464) | -(0.464) | +++(0.708) | |
Metabolism | CYP-1A2 inhibitor | ---(0.197) | +++(0.968) | +++(0.987) | +++(0.968) | ---(0.034) |
CYP-1A2 subst. | ---(0.258) | -(0.376)) | -(0.408) | -(0.412) | -(0.368) | |
CYP-3A4 inhibitor | +(0.518) | -(0.459) | +++(0.931) | ++(0.867) | ---(0.221) | |
CYP-3A4 subst. | -(0.418) | -(0.33) | -(0.37) | -(0.328) | -(0.398) | |
CYP-2C9 inhibitor | ---(0.199) | +(0.656) | -(0.12) | ---(0.071) | ---(0.028) | |
CYP-2C9 subst. | ---(0.298) | +(0.557) | +(0.631) | -(0.496) | -(0.049) | |
CYP-2C19 inhibitor | ---(0.072) | ---(0.068) | ---(0.124) | ---(0.124) | -(0.431) | |
CYP-2C19 subst. | -(0.49) | -(0.345) | -(0.412) | +(0.542) | -(0.476) | |
CYP-2D6 inhibitor | ---(0.29) | -(0.318) | +(0.611) | -(0.463) | -(0.367) | |
CYP-2D6 subst. | ---(0.218) | -(0.18) | -(0.488) | -(0.401) | +(0.523) | |
Elimination | T 1/2 (half-life time) | 2.138 h | 0.915 h | 1.331 h | 0.745 h | 0.720 h |
Clearance rate (mL/min/kg) | 0.641 | 1.709 | 1.885 | 1.919 | 1.914 | |
Toxicity/alert | hERG blocker | +(0.626) | -(0.353) | +(0.598) | -(0.436) | -(0.371) |
(hepatotoxicity (H-HT) | ---(0.154) | -(0.332) | +(0.6) | +(0.592) | -(0.466) | |
SkinSen | ---(0.235) | ---(0.278) | ---(0.264) | ---(0.278) | -(0.329) | |
50% acute toxicity (LD50 mg/kg) | 419.47 | 648.262 | 858.518 | 737.444 | 860.605 |
TPSA: topological polar surface area; CYP: cytochrome P450; hERG: human ether-a-go-go-related gene.
3.11. Receptor-Ligand Simulation Analysis Revealed Luteolin’s Properties for Targeting Glucose-Metabolizing Enzymes and an Inflammatory Mediator
Molecular docking is an innovative and widely approved strategies for mimicking a small-molecule interaction with a target receptor/protein [57, 100, 101]. It provides qualitative and quantitative estimation of the affinity between a compound and the corresponding protein/receptor [102]. It also gives a preamble insight into mechanistic aspect of the compound and its behavior when in contact with the corresponding target [103–107]. Docking analysis in the present study revealed that luteolin docked efficiently to the substrate interaction domain of α-amylase with a binding affinity of –8.6 kcal/mol. The complex was bound by four hydrogen bonds to Gln63 (2.62 Å), Asp300 (2.81 Å), Arg195 (2.16 Å), and Glu233 (2.32 Å); pi-pi stacked (Trp59), pi-anion (Asp300), and several van der Waals forces including His305, Leu162, Asp197, His299, Tyr62, Trp58, Ala198, Thr163, and Leu165 (Figure 9). In addition, the complex was bound by four hydrophobic contacts with Thr62A and TRP59A with interaction distances of 3.80, 3.52, 3.75, and 3.64 Å.
[figure(s) omitted; refer to PDF]
Luteolin interacted with glucosidase by –8.5 kcal/mol binding efficacy. Luteolin was interposed to the cavity of glucosidase mainly by hydrogen bonds with Ser150 and TRP143 residues of glucosidase domain in respective proximity of 3.28 and 2.52 Å. The luteolin-glucosidase complex stabilization was also achieved by a pi-anion interaction with Asp136, two alkyl bonding with Pro139A and Pro139B, and several van der Waals forces, including Lys134A, Asp215, Lys134B, Asn141, Asn142A, Thr138, Asn142B, Ala151, Leu144, Ser150, and Trp152, molded at the luteolin backbone. In addition, there were three hydrophobic contacts with Thr138A (3.75 Å), Asn142A (3.75 Å), and Lys134B (3.80 Å), and the carboxylate group of luteolin formed a salt bridge interaction with the binding domain of alpha-glucosidase (Figure 10).
[figure(s) omitted; refer to PDF]
The luteolin-cyclooxygenase complex was stabilized by several pi interactions, including pi-alkyl (Val295), pi-sigma (Leu391), amide-pi-stacked (Ala202), and pi-pi T shape (His388), yielding a high ligand-binding affinity of –8.8 kcal/mol. In total, 13 van der Waals forces with Val444, Phe404, Phe395, Phe407, Phe200, Gln203, His207, His386, Phe210, Tyr385, Leu390, Trp387, and Thr206 residues of the cyclooxygenases were found around the luteolin backbone. In addition, hydrophobic contact of Gln203A with proximity of 3.80 Å was found in the complex (Figure 11). Overall, data presented in this study provides some scientific affirmation based on preclinical model of the anti-inflammatory, hypoglycemic, and antioxidant properties of the T. garckeana extract. This hinted at its potentiality for exploration in the development of alternative therapies for the management and possibly treatment of diabetes complication
[figure(s) omitted; refer to PDF]
4. Conclusions
The present study provides experimental evidence of the therapeutic efficacy of the diethyl-ether fraction of Thespesia garckeana for treating diabetes. The extract not only enhanced the activities of antioxidant enzymes but also inhibited inflammatory responses, and demonstrated in vivo antidiabetic effects in experimental models. The compound most abundant (luteolin) in the fraction demonstrated good drug-PK and drug-likeness and prospective for the targeting of glucose-catabolizing enzymes. Thus, the present study provides preclinical insights into the bioactive constituents of T. garckeana, its anti-inflammatory and antioxidant effects, and its potential for treating diabetes.
Authors’ Contributions
All authors read and approved the final version of the manuscript. Uchenna Blessing Alozieuwa and Bashir Lawal contributed equally to this work.
Acknowledgments
ATHW is funded by the Ministry of Science and Technology, Taiwan (111-2314-B-038 -142 and 111-2314-B-038 -098). The authors would like to extend their sincere appreciation to the Taif University Researchers Supporting Project number (TURSP-2020/202) Taif University, Taif, Saudi Arabia.
[1] R. A. DeFronzo, "Pathogenesis of type 2 diabetes mellitus," Medical clinics, vol. 88 no. 4, pp. 787-835, ix, DOI: 10.1016/j.mcna.2004.04.013, 2004.
[2] L. M. Janssen, M. Hiligsmann, A. M. Elissen, M. A. Joore, N. C. Schaper, J. H. Bosma, C. D. Stehouwer, S. J. Sep, A. Koster, M. T. Schram, S. M. Evers, "Burden of disease of type 2 diabetes mellitus: cost of illness and quality of life estimated using the Maastricht Study," Diabetic Medicine, vol. 2020,DOI: 10.1111/dme.14285, 2020.
[3] R. A. DeFronzo, E. Ferrannini, L. Groop, R. R. Henry, W. H. Herman, J. J. Holst, F. B. Hu, C. R. Kahn, I. Raz, G. I. Shulman, "Type 2 diabetes mellitus," Nature reviews Disease primers, vol. 1, 2015.
[4] A. Maritim, R. A. Sanders, J. B. Watkins, "Diabetes, oxidative stress, and antioxidants: a review," Journal of Biochemical and Molecular Toxicology, vol. 17 no. 1, pp. 24-38, DOI: 10.1002/jbt.10058, 2003.
[5] S. Tsalamandris, A. S. Antonopoulos, E. Oikonomou, G.-A. Papamikroulis, G. Vogiatzi, S. Papaioannou, S. Deftereos, D. Tousoulis, "The role of inflammation in diabetes: current concepts and future perspectives," European cardiology review, vol. 14 no. 1, pp. 50-59, DOI: 10.15420/ecr.2018.33.1, 2019.
[6] Y. Wang, J. Zhai, D. Yang, N. Han, Z. Liu, Z. Liu, S. Li, J. Yin, "Antioxidant, anti-inflammatory, and antidiabetic activities of bioactive compounds from the fruits of Livistona chinensis based on network pharmacology prediction," Oxidative Medicine and Cellular Longevity, vol. 2021,DOI: 10.1155/2021/7807046, 2021.
[7] M. Piero, G. Nzaro, J. Njagi, "Diabetes mellitus-a devastating metabolic disorder," Asian journal of biomedical and pharmaceutical sciences, vol. 4 no. 40,DOI: 10.15272/ajbps.v4i40.645, 2015.
[8] H. Sun, P. Saeedi, S. Karuranga, M. Pinkepank, K. Ogurtsova, B. B. Duncan, C. Stein, A. Basit, J. C. N. Chan, J. C. Mbanya, M. E. Pavkov, A. Ramachandaran, S. H. Wild, S. James, W. H. Herman, P. Zhang, C. Bommer, S. Kuo, E. J. Boyko, D. J. Magliano, "IDF Diabetes Atlas: global, regional and country-level diabetes prevalence estimates for 2021 and projections for 2045," Diabetes Research and Clinical Practice, vol. 183,DOI: 10.1016/j.diabres.2021.109119, 2022.
[9] N. Cho, J. Shaw, S. Karuranga, Y. Huang, J. da Rocha Fernandes, A. Ohlrogge, B. Malanda, "IDF Diabetes Atlas: global estimates of diabetes prevalence for 2017 and projections for 2045," Diabetes Research and Clinical Practice, vol. 138, pp. 271-281, DOI: 10.1016/j.diabres.2018.02.023, 2018.
[10] O. A. Ojo, M. A. Okesola, L. I. Ekakitie, B. O. Ajiboye, B. E. Oyinloye, P. E. Agboinghale, A. S. Onikanni, "Gongronema latifoliumBenth. leaf extract attenuates diabetes‐induced neuropathy via inhibition of cognitive, oxidative stress and inflammatory response," Journal of the Science of Food and Agriculture, vol. 100 no. 12, pp. 4504-4511, DOI: 10.1002/jsfa.10491, 2020.
[11] D. R. Whiting, L. Guariguata, C. Weil, J. Shaw, "IDF Diabetes Atlas: global estimates of the prevalence of diabetes for 2011 and 2030," Diabetes Research and Clinical Practice, vol. 94 no. 3, pp. 311-321, DOI: 10.1016/j.diabres.2011.10.029, 2011.
[12] Z. Li, Y.-N. Geng, J.-D. Jiang, W.-J. Kong, "Antioxidant and anti-inflammatory activities of berberine in the treatment of diabetes mellitus," Evidence-based Complementary and Alternative Medicine, vol. 2014,DOI: 10.1155/2014/289264, 2014.
[13] M. Al-Zharani, A. F. Nasr, M. O. Noman, R. Conte, E. H. E. Y. Amal, "Antioxidant, anti-inflammatory and antidiabetic proprieties of LC-MS/MS identified polyphenols from coriander seeds," Molecules, vol. 26 no. 2,DOI: 10.3390/molecules26020487, 2021.
[14] S.-S. Huang, S.-Y. Su, J.-S. Chang, H.-J. Lin, W.-T. Wu, J.-S. Deng, G.-J. Huang, "Antioxidants, anti-inflammatory, and antidiabetic effects of the aqueous extracts from Glycine species and its bioactive compounds," Botanical Studies, vol. 57 no. 1,DOI: 10.1186/s40529-016-0153-7, 2016.
[15] M. Y. Donath, S. E. Shoelson, "Type 2 diabetes as an inflammatory disease," Nature Reviews Immunology, vol. 11 no. 2, pp. 98-107, DOI: 10.1038/nri2925, 2011.
[16] W. Xie, L. Du, "Diabetes is an inflammatory disease: evidence from traditional Chinese medicines," Diabetes, Obesity and Metabolism, vol. 13 no. 4, pp. 289-301, DOI: 10.1111/j.1463-1326.2010.01336.x, 2011.
[17] A. S. Onikanni, B. Lawal, A. O. Olusola, J. O. Olugbodi, S. Sani, B. O. Ajiboye, O. B. Ilesanmi, M. Alqarni, G. Mostafa-Hedeab, A. J. Obaidullah, G. E. Batiha, A. T. H. Wu, "Sterculia tragacantha Lindl leaf extract ameliorates STZ-induced diabetes, oxidative stress, inflammation and neuronal impairment," Journal of Inflammation Research, vol. 14, pp. 6749-6764, DOI: 10.2147/jir.s319673, 2021.
[18] J. L. Evans, I. D. Goldfine, B. A. Maddux, G. M. Grodsky, "Are oxidative stress− activated signaling pathways mediators of insulin resistance and β -cell dysfunction?," Diabetes, vol. 52 no. 1,DOI: 10.2337/diabetes.52.1.1, 2003.
[19] H. Yang, X. Jin, C. W. K. Lam, S.-K. Yan, "Oxidative stress and diabetes mellitus," Clinical Chemistry and Laboratory Medicine, vol. 49 no. 11, pp. 1773-1782, DOI: 10.1515/CCLM.2011.250, 2011.
[20] A. S. Onikanni, B. Lawal, B. E. Oyinloye, G. Mostafa-Hedeab, M. Alorabi, S. Cavalu, A. O. Olusola, C.-H. Wang, G. E.-S. Batiha, "Therapeutic efficacy of Clompanus pubescens leaves fractions via downregulation of neuronal cholinesterases/Na + -K + ATPase/IL-1 β , and improving the neurocognitive and antioxidants status of streptozotocin- induced diabetic rats," Biomedicine & Pharmacotherapy, vol. 148,DOI: 10.1016/j.biopha.2022.112730, 2022.
[21] A. Chaudhury, C. Duvoor, V. S. Reddy Dendi, S. Kraleti, A. Chada, R. Ravilla, A. Marco, N. S. Shekhawat, M. T. Montales, K. Kuriakose, A. Sasapu, A. Beebe, N. Patil, C. K. Musham, G. P. Lohani, W. Mirza, "Clinical review of antidiabetic drugs: implications for type 2 diabetes mellitus management," Frontiers in endocrinology, vol. 8,DOI: 10.3389/fendo.2017.00006, 2017.
[22] R. F. Calson, "Miglitol and hepatotoxicity in type 2 diabetes mellitus," American Family Physician, vol. 62, 2000.
[23] R. Amoretti, C. Cicconetti, M. De Nichilo, C. Teodonio, R. Fasani, M. Carosi, "Acarbose in the treatment of type 2 diabetes," Minerva Dietologica e Gastroenterologica, vol. 32 no. 1, pp. 53-59, 1986.
[24] M. Jung, M. Park, H. C. Lee, Y.-H. Kang, E. S. Kang, S. K. Kim, "Antidiabetic agents from medicinal plants," Current Medicinal Chemistry, vol. 13 no. 10, pp. 1203-1218, DOI: 10.2174/092986706776360860, 2006.
[25] N. H. Salleh, I. N. Zulkipli, H. Mohd Yasin, F. Ja'afar, N. Ahmad, W. A. N. Wan Ahmad, S. R. Ahmad, "Systematic review of medicinal plants used for treatment of diabetes in human clinical trials: an ASEAN perspective," Evidence-based Complementary and Alternative Medicine, vol. 2021,DOI: 10.1155/2021/5570939, 2021.
[26] M. I. Yatoo, A. Saxena, A. Gopalakris, M. Alagawany, K. Dhama, "Promising antidiabetic drugs, medicinal plants and herbs: an update," International Journal of Pharmacology, vol. 13 no. 7, pp. 732-745, DOI: 10.3923/ijp.2017.732.745, 2017.
[27] W. Mojeremane, S. Tshwenyane, "Azanza garckeana: a valuable edible indigenous fruit tree of Botswana," Pakistan Journal of Nutrition, vol. 3 no. 5, pp. 264-267, DOI: 10.3923/pjn.2004.264.267, 2004.
[28] A. A. Yusuf, B. Lawal, S. Sani, R. Garba, B. A. Mohammed, D. B. Oshevire, D. A. Adesina, "Pharmacological activities of Azanza garckeana (Goron Tula) grown in Nigeria," Clinical Phytoscience, vol. 6, 2020.
[29] Y. Dikko, M. Khan, T. Tor-Anyiin, J. Anyam, U. Linus, "In vitro antimicrobial activity of fruit pulp extracts of Azanza garckeana (F. Hoffm.) Exell & Hillc. and isolation of one of its active principles, betulinic acid," Journal of Pharmaceutical Research International, vol. 14 no. 1,DOI: 10.9734/BJPR/2016/30152, 2016.
[30] B. B. Bukar, F. Ezeh, S. Y. Sabo, "Methanol Extract of Azanza garckeana Fruit Pulps Protects against Formalin-Induced Reproductive Toxicity in Adult Albino Male Mice," Journal of Advances in Medical and Pharmaceutical Sciences, vol. 23 no. 8, pp. 38-49, DOI: 10.9734/jamps/2021/v23i830253, 2021.
[31] Y. Dikko, M. E. Khan, T. A. Tor-Anyiin, J. V. Anyam, U. Linus, "In vitro Antimicrobial Activity of Fruit Pulp Extracts of Azanza garckeana (F. Hoffm.) Exell & Hillc. and Isolation of One of its Active Principles, Betulinic Acid," British Journal of Pharmaceutical Research, vol. 14, 2016.
[32] B. B. Bukar, N. E. Tsokwa, O. D. Orshi, "Ameliorative and fecundity potentials of aqueous extract of Azanza garckeana (T. Hoffm) fruit pulp in formalin-induced toxicity on male albino mice," Journal of Pharmacy & Bioresources, vol. 17, pp. 164-173, 2020.
[33] C.-C. Chang, M.-H. Yang, H.-M. Wen, J.-C. Chern, "Estimation of total flavonoid content in propolis by two complementary colometric methods," Journal of Food and Drug Analysis, vol. 10 no. 3,DOI: 10.38212/2224-6614.2748, 2002.
[34] V. L. Singleton, R. Orthofer, R. M. Lamuela-Raventós, "Analysis of total phenols and other oxidation substrates and antioxidants by means of folin-ciocalteu reagent," Methods in enzymology 1999 Jan 1 (Vol. 299, pp. 152-178), vol. 299, pp. 152-178, 1999.
[35] M. Oyaizu, "Studies on products of browning reaction antioxidative activities of products of browning reaction prepared from glucosamine," The Japanese journal of nutrition and dietetics, vol. 44 no. 6, pp. 307-315, DOI: 10.5264/eiyogakuzashi.44.307, 1986.
[36] K. Panjamurthy, S. Manoharan, C. R. Ramachandran, "Lipid peroxidation and antioxidant status in patients with periodontitis," Cellular & Molecular Biology Letters, vol. 10 no. 2, pp. 255-264, 2005.
[37] R. Re, N. Pellegrini, A. Proteggente, A. Pannala, M. Yang, C. Rice-Evans, "Antioxidant activity applying an improved ABTS radical cation decolorization assay," Free Radical Biology and Medicine, vol. 26 no. 9-10, pp. 1231-1237, DOI: 10.1016/S0891-5849(98)00315-3, 1999.
[38] K. Worthington, Alpha Amylase Worthington Enzyme Manual, 1993.
[39] Y. Mizushima, M. Kobayashi, "Interaction of anti-inflammatory drugs with serum proteins, especially with some biologically active proteins," Journal of Pharmacy and Pharmacology, vol. 20 no. 3, pp. 169-173, DOI: 10.1111/j.2042-7158.1968.tb09718.x, 1968.
[40] O. Oyedapo, A. J. Famurewa, "Antiprotease and membrane stabilizing activities of extracts of Fagara zanthoxyloides, Olax subscorpioides and Tetrapleura tetraptera," International Journal of Pharmacognosy, vol. 33 no. 1, pp. 65-69, DOI: 10.3109/13880209509088150, 1995.
[41] V. Thenmozhi, V. Elango, J. Sadique, "Anti-inflamatory activity of some Indian medicinal plants," Ancient Science of Life, vol. 8 no. 3-4, pp. 258-261, 1989.
[42] D. Lorke, "A new approach to practical acute toxicity testing," Archives of Toxicology, vol. 54 no. 4, pp. 275-287, DOI: 10.1007/BF01234480, 1983.
[43] W. Sisay, Y. Andargie, M. Molla, "Antidiabetic activity of hydromethanolic extract of crude Dorstenia barnimiana root: validation of in vitro and in vivo antidiabetic and antidyslipidemic activity," Journal of Experimental Pharmacology, vol. Volume 14, pp. 59-72, DOI: 10.2147/JEP.S343735, 2022.
[44] E. Etuk, "Animals models for studying diabetes mellitus," Agriculture and Biology Journal of North America, vol. 1, pp. 130-134, 2010.
[45] Q. Zhang, X. Li, J. Li, Y. Hu, J. Liu, F. Wang, W. Zhang, F. Chang, "Mechanism of anti-inflammatory and antibacterial effects of QingXiaoWuWei decoction based on network pharmacology, molecular docking and in vitro experiments," Frontiers in Pharmacology, vol. 12,DOI: 10.3389/fphar.2021.678685, 2021.
[46] B. Lawal, S. Sani, A. S. Onikanni, Y. O. Ibrahim, A. R. Agboola, H. Y. Lukman, F. Olawale, A. A. Jigam, G. E.-S. Batiha, S. B. Babalola, G. Mostafa-Hedeab, C. M. G. Lima, A. T. H. Wu, H. S. Huang, C. A. Conte-Junior, "Preclinical anti-inflammatory and antioxidant effects of Azanza garckeana in STZ-induced glycemic-impaired rats, and pharmacoinformatics of it major phytoconstituents," Biomedicine & Pharmacotherapy, vol. 152,DOI: 10.1016/j.biopha.2022.113196, 2022.
[47] O. Shittu, B. Lawal, O. Oluyomi, "Effects of methanol extract of Musca domestica larvae on antioxidants enzymes in T. Brucei infected rats," Nigerian Journal of Biochemistry and Molecular Biology, vol. 29, 2014.
[48] J. Ibrahim, A. Y. Kabiru, T. Abdulrasheed-Adeleke, B. Lawal, A. H. Adewuyi, "Antioxidant and hepatoprotective potentials of curcuminoid isolates from turmeric (Curcuma longa) rhizome on CCl₄-induced hepatic damage in Wistar rats," Journal of Taibah University for Science, vol. 14 no. 1, pp. 908-915, DOI: 10.1080/16583655.2020.1790928, 2020.
[49] K. Shagirtha, N. Bashir, S. MiltonPrabu, "Neuroprotective efficacy of hesperetin against cadmium induced oxidative stress in the brain of rats," Toxicology and Industrial Health, vol. 33 no. 5, pp. 454-468, DOI: 10.1177/0748233716665301, 2017.
[50] L. Kum-Tatt, I.-K. Tan, "A new colorimetric method for the determination of glutathione in erythrocytes," Clinica Chimica Acta, vol. 53 no. 2, pp. 153-161, DOI: 10.1016/0009-8981(74)90093-X, 1974.
[51] H. P. Misra, I. Fridovich, "The role of superoxide anion in the autoxidation of epinephrine and a simple assay for superoxide dismutase," Journal of Biological Chemistry, vol. 247 no. 10, pp. 3170-3175, DOI: 10.1016/S0021-9258(19)45228-9, 1972.
[52] A. K. Sinha, "Colorimetric assay of catalase," Analytical Biochemistry, vol. 47 no. 2, pp. 389-394, DOI: 10.1016/0003-2697(72)90132-7, 1972.
[53] H. Liu, L. Wang, M. Lv, R. Pei, P. Li, Z. Pei, Y. Wang, W. Su, X.-Q. Xie, "AlzPlatform: an Alzheimer’s disease domain-specific chemogenomics knowledgebase for polypharmacology and target identification research," Journal of Chemical Information and Modeling, vol. 54 no. 4, pp. 1050-1060, DOI: 10.1021/ci500004h, 2014.
[54] M. D. Hanwell, D. E. Curtis, D. C. Lonie, T. Vandermeersch, E. Zurek, G. R. Hutchison, "Avogadro: an advanced semantic chemical editor, visualization, and analysis platform," Journal of Cheminformatics, vol. 4 no. 1,DOI: 10.1186/1758-2946-4-17, 2012.
[55] O. Trott, A. J. Olson, "AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading," Journal of Computational Chemistry, vol. 31 no. 2, pp. 455-461, DOI: 10.1002/jcc.21334, 2010.
[56] B. Lawal, Y.-L. Liu, N. Mokgautsi, H. Khedkar, M. R. Sumitra, A. T. H. Wu, H.-S. Huang, "Pharmacoinformatics and preclinical studies of NSC765690 and NSC765599, potential STAT3/CDK2/4/6 inhibitors with antitumor activities against NCI60 human tumor cell lines," Biomedicine, vol. 9 no. 1,DOI: 10.3390/biomedicines9010092, 2021.
[57] S.-Y. Wu, K.-C. Lin, B. Lawal, A. T. H. Wu, C.-Z. Wu, "MXD3 as an onco-immunological biomarker encompassing the tumor microenvironment, disease staging, prognoses, and therapeutic responses in multiple cancer types," Computational and Structural Biotechnology Journal, vol. 19, pp. 4970-4983, DOI: 10.1016/j.csbj.2021.08.047, 2021.
[58] B. Lawal, C.-Y. Lee, N. Mokgautsi, M. R. Sumitra, H. Khedkar, A. T. H. Wu, H.-S. Huang, "mTOR/EGFR/iNOS/MAP2K1/FGFR/TGFB1 are druggable candidates for N-(2,4-difluorophenyl)-2 ′ ,4 ′ -difluoro-4-hydroxybiphenyl-3-carboxamide (NSC765598), with consequent anticancer implications," Frontiers in Oncology, vol. 11,DOI: 10.3389/fonc.2021.656738, 2021.
[59] J. O. Olugbodi, K. Samaila, B. Lawal, O. O. Anunobi, R. S. Baty, O. B. Ilesanmi, G. E.-S. Batiha, "Computational and preclinical evidence of anti-ischemic properties of L-carnitine-rich supplement via stimulation of anti-inflammatory and antioxidant events in testicular torsed rats," Oxidative Medicine and Cellular Longevity, vol. 2021,DOI: 10.1155/2021/5543340, 2021.
[60] Visualizer, DS BIOVIA, Dassault Systèmes, BIOVIA Workbook, Release 2020; BIOVIA pipeline pilot, release 2020, 2020.
[61] K. K. Keshala, A. M. P. W. Bandara, C. Padumadasa, L. D. C. Peiris, "Bioactivities and GC-MS profiling of Malewana Madhumeha Choorna polyherbal hot infusion," South African Journal of Botany, vol. 140, pp. 194-203, DOI: 10.1016/j.sajb.2021.04.015, 2021.
[62] D. Tungmunnithum, A. Thongboonyou, A. Pholboon, A. Yangsabai, "Flavonoids and other phenolic compounds from medicinal plants for pharmaceutical and medical aspects: an overview," Medicine, vol. 5 no. 3,DOI: 10.3390/medicines5030093, 2018.
[63] D. Villaño, M. Fernández-Pachón, M. L. Moyá, A. Troncoso, M. García-Parrilla, "Radical scavenging ability of polyphenolic compounds towards DPPH free radical," Talanta, vol. 71 no. 1, pp. 230-235, DOI: 10.1016/j.talanta.2006.03.050, 2007.
[64] S. C. Tiwari, N. Husain, "Biological activities and role of flavonoids in human health–a," Indian Journal of Science Research, vol. 12, pp. 193-196, 2017.
[65] M. G. Chaudhari, B. B. Joshi, K. N. Mistry, "In vitro anti-diabetic and anti-inflammatory activity of stem bark of Bauhinia purpurea," Bulletin of Pharmaceutical and Medical Sciences (BOPAMS), vol. 1, pp. 139-150, 2013.
[66] M. Govindappa, T. Sadananda, R. Channabasava, V. B. Raghavendra, "In vitro anti-inflammatory, lipoxygenase, xanthine oxidase and acetycholinesterase inhibitory activity of Tecoma stans (l.) Juss. Ex kunth," International Journal of Pharma and Bio Sciences, vol. 2, pp. 275-285, 2011.
[67] S. Rajasekaran, M. Dinesh, C. Kansrajh, F. H. A. Baig, "Amaranthus spinosus leaf extracts and its anti-inflamatory effects on cancer," Indian Journal of Research in Pharmacy and Biotechnology, vol. 2, 2014.
[68] S. Doumas, A. Kolokotronis, P. Stefanopoulos, "Anti-inflammatory and antimicrobial roles of secretory leukocyte protease inhibitor," Infection and Immunity, vol. 73 no. 3, pp. 1271-1274, DOI: 10.1128/IAI.73.3.1271-1274.2005, 2005.
[69] K. Hibbetts, B. Hines, D. Williams, "An overview of proteinase inhibitors," Journal of Veterinary Internal Medicine, vol. 13 no. 4, pp. 302-308, DOI: 10.1111/j.1939-1676.1999.tb02185.x, 1999.
[70] H. C. Hodge, J. H. Sterner, "Tabulation of toxicity classes," American Industrial Hygiene Association Quarterly, vol. 10 no. 4, pp. 93-96, DOI: 10.1080/00968204909344159, 1949.
[71] I. J. Iyojo, R. P. Ibrahim, A. Tagang, L. Allam, A. J. Olusegun, "Effects of different extracts of Azanza garckeana fruit pulp on haematological and biochemical parameters of New Zealand White (NZW) rabbit bucks," Comparative Clinical Pathology, vol. 31 no. 3, pp. 453-463, DOI: 10.1007/s00580-022-03344-2, 2022.
[72] A. O. Owoade, A. Adetutu, O. S. Olorunnisola, K. S. Ayinde, "The in-vitro antioxidant properties and phytochemical constituents of Citrullus colocynthis methanolic extract," Elixir Appl. Botany, vol. 121, pp. 51556-51562, 2018.
[73] R. Tundis, M. Loizzo, F. Menichini, "Natural products as α -amylase and α -glucosidase inhibitors and their hypoglycaemic potential in the treatment of diabetes: an update," Mini Reviews in Medicinal Chemistry, vol. 10 no. 4, pp. 315-331, DOI: 10.2174/138955710791331007, 2010.
[74] W. A. S. Chamika, T. C. Ho, V. C. Roy, A. T. Kiddane, J.-S. Park, G.-D. Kim, B.-S. Chun, "In vitro characterization of bioactive compounds extracted from sea urchin ( Stomopneustes variolaris ) using green and conventional techniques," Food Chemistry, vol. 361,DOI: 10.1016/j.foodchem.2021.129866, 2021.
[75] N. Mahmood, "A review of α -amylase inhibitors on weight loss and glycemic control in pathological state such as obesity and diabetes," Comparative Clinical Pathology, vol. 25 no. 6, pp. 1253-1264, DOI: 10.1007/s00580-014-1967-x, 2016.
[76] F. Rolland, J. Winderickx, J. M. Thevelein, "Glucose-sensing and -signalling mechanisms in yeast," FEMS Yeast Research, vol. 2 no. 2, pp. 183-201, DOI: 10.1016/S1567-1356(02)00046-6, 2002.
[77] K. Ninomiya, Y. Yamaguchi, F. Shinmachi, H. Kumagai, H. Kumagai, "Suppression of postprandial blood glucose elevation by buckwheat ( Fagpopyrum esculentum ) albumin hydrolysate and identification of the peptide responsible to the function," Food Science and Human Wellness, vol. 11 no. 4, pp. 992-998, DOI: 10.1016/j.fshw.2022.03.026, 2022.
[78] S. Chen, B. Lin, J. Gu, T. Yong, X. Gao, Y. Xie, C. Xiao, J. Y. Zhan, Q. Wu, "Binding interaction of betulinic acid to α -glucosidase and its alleviation on postprandial hyperglycemia," Molecules, vol. 27 no. 8,DOI: 10.3390/molecules27082517, 2022.
[79] B.-E. Van Wyk, M. Wink, Medicinal Plants of the World, 2017.
[80] K. Neha, M. R. Haider, A. Pathak, M. S. Yar, "Medicinal prospects of antioxidants: a review," European Journal of Medicinal Chemistry, vol. 178, pp. 687-704, DOI: 10.1016/j.ejmech.2019.06.010, 2019.
[81] D. Rajendiran, S. Packirisamy, K. Gunasekaran, "A review on role of antioxidants in diabetes," Asian Journal of Pharmaceutical and Clinical Research, vol. 11 no. 2, pp. 48-53, DOI: 10.22159/ajpcr.2018.v11i2.23241, 2018.
[82] D. M. Adams, M. T. Yakubu, "Aqueous extract of Digitaria exilis grains ameliorate diabetes in streptozotocin-induced diabetic male Wistar rats," Journal of Ethnopharmacology, vol. 249,DOI: 10.1016/j.jep.2019.112383, 2020.
[83] M. C. Petersen, D. F. Vatner, G. I. Shulman, "Regulation of hepatic glucose metabolism in health and disease," Nature Reviews Endocrinology, vol. 13 no. 10, pp. 572-587, DOI: 10.1038/nrendo.2017.80, 2017.
[84] B. Lawal, O. K. Shittu, P. C. Ossai, A. N. Abubakar, A. M. Ibrahim, "Evaluation of antioxidant activity of giant African snail (Achachatina maginata) haemolymph in CCl4-induced hepatotoxixity in albino rats," Journal of Pharmaceutical Research International, vol. 6 no. 3, pp. 141-154, DOI: 10.9734/BJPR/2015/15887, 2015.
[85] O. I. Aruoma, "Free radicals, oxidative stress, and antioxidants in human health and disease," Journal of the American Oil Chemists' Society, vol. 75 no. 2, pp. 199-212, DOI: 10.1007/s11746-998-0032-9, 1998.
[86] L. Tappy, "Metabolism of sugars: a window to the regulation of glucose and lipid homeostasis by splanchnic organs," Clinical Nutrition, vol. 40 no. 4, pp. 1691-1698, DOI: 10.1016/j.clnu.2020.12.022, 2021.
[87] J. Mohamed, A. H. Nazratun Nafizah, A. H. Zariyantey, S. B. Budin, "Mechanisms of diabetes-induced liver damage: the role of oxidative stress and inflammation," Sultan Qaboos University medical journal, vol. 16, pp. e132-e141, DOI: 10.18295/squmj.2016.16.02.002, 2016.
[88] M. J. Hossen, M. A. Matin, M. H. Sikder, M. S. Ahmed, M. Rahman, "Kupffer cells and liver," Recent Advancements in Microbial Diversity, pp. 361-395, 2022.
[89] H. Maeda, Y. Ishima, J. Saruwatari, Y. Mizuta, Y. Minayoshi, S. Ichimizu, H. Yanagisawa, T. Nagasaki, K. Yasuda, S. Oshiro, M. Taura, M. J. McConnell, K. Oniki, K. Sonoda, T. Wakayama, M. Kinoshita, T. Shuto, H. Kai, M. Tanaka, Y. Sasaki, Y. Iwakiri, M. Otagiri, H. Watanabe, T. Maruyama, "Nitric oxide facilitates the targeting Kupffer cells of a nano-antioxidant for the treatment of NASH," Journal of Controlled Release, vol. 341, pp. 457-474, DOI: 10.1016/j.jconrel.2021.11.039, 2022.
[90] J. S. Bhatti, A. Sehrawat, J. Mishra, I. S. Sidhu, U. Navik, N. Khullar, S. Kumar, G. K. Bhatti, P. H. Reddy, "Oxidative stress in the pathophysiology of type 2 diabetes and related complications: current therapeutics strategies and future perspectives," Free Radical Biology and Medicine, vol. 184, pp. 114-134, DOI: 10.1016/j.freeradbiomed.2022.03.019, 2022.
[91] Y. M. Yang, Y. E. Cho, S. Hwang, "Crosstalk between oxidative stress and inflammatory liver injury in the pathogenesis of alcoholic liver disease," International Journal of Molecular Sciences, vol. 23 no. 2,DOI: 10.3390/ijms23020774, 2022.
[92] C. Matyas, G. Haskó, L. Liaudet, E. Trojnar, P. Pacher, "Interplay of cardiovascular mediators, oxidative stress and inflammation in liver disease and its complications," Nature Reviews Cardiology, vol. 18 no. 2, pp. 117-135, DOI: 10.1038/s41569-020-0433-5, 2021.
[93] M. Kaleem, M. Asif, Q. Ahmed, B. Bano, "Antidiabetic and antioxidant activity of Annona squamosa extract in streptozotocin-induced diabetic rats," Singapore Medical Journal, vol. 47 no. 8, pp. 670-675, 2006.
[94] K. O. Karigidi, C. O. Olaiya, "Antidiabetic activity of corn steep liquor extract of Curculigo pilosa and its solvent fractions in streptozotocin-induced diabetic rats," Journal of Traditional and Complementary Medicine, vol. 10 no. 6, pp. 555-564, DOI: 10.1016/j.jtcme.2019.06.005, 2020.
[95] D. Cao, J. Wang, R. Zhou, Y. Li, H. Yu, T. Hou, "ADMET evaluation in drug discovery. 11. PharmacoKinetics Knowledge Base (PKKB): a comprehensive database of pharmacokinetic and toxic properties for drugs," Journal of Chemical Information and Modeling, vol. 52 no. 5, pp. 1132-1137, DOI: 10.1021/ci300112j, 2012.
[96] I. Kola, J. Landis, "Can the pharmaceutical industry reduce attrition rates?," Nature Reviews. Drug Discovery, vol. 3 no. 8, pp. 711-716, DOI: 10.1038/nrd1470, 2004.
[97] M. L. Amin, "P-glycoprotein inhibition for optimal drug delivery," Drug target insights, vol. 7, pp. 27-34, DOI: 10.4137/DTI.S12519, 2013.
[98] J. Robert, C. Jarry, "Multidrug resistance reversal agents," Journal of Medicinal Chemistry, vol. 46 no. 23, pp. 4805-4817, DOI: 10.1021/jm030183a, 2003.
[99] K. Shankar, H. M. Mehendale, "Cytochrome P450," Encyclopedia of Toxicology, pp. 1125-1127, 2014.
[100] Y.-C. Yeh, B. Lawal, M. Hsiao, T.-H. Huang, C. Y. F. Huang, C.-Y. F. Huang, "Identification of NSP3 (SH2D3C) as a prognostic biomarker of tumor progression and immune evasion for lung cancer and evaluation of organosulfur compounds from Allium sativum L. as therapeutic candidates," Biomedicine, vol. 9 no. 11,DOI: 10.3390/biomedicines9111582, 2021.
[101] B. Lawal, Y.-C. Wang, A. T. H. Wu, H.-S. Huang, "Pro-oncogenic c-met/EGFR, biomarker signatures of the tumor microenvironment are clinical and therapy response prognosticators in colorectal cancer, and therapeutic targets of 3-phenyl-2H-benzo[e][1,3]-oxazine-2,4(3H)-dione derivatives," Frontiers in Pharmacology, vol. 12,DOI: 10.3389/fphar.2021.691234, 2021.
[102] D. M. I. H. Dissanayake, D. D. B. D. Perera, L. R. Keerthirathna, S. Heendeniya, R. J. Anderson, D. E. Williams, L. D. C. Peiris, "Antimicrobial activity of Plumbago indica and ligand screening of plumbagin against methicillin-resistant Staphylococcus aureus," Journal of Biomolecular Structure and Dynamics, vol. 40 no. 7, pp. 3273-3284, DOI: 10.1080/07391102.2020.1846622, 2022.
[103] X.-Y. Meng, H.-X. Zhang, M. Mezei, M. Cui, "Molecular docking: a powerful approach for structure-based drug discovery," Current Computer-Aided Drug Design, vol. 7 no. 2, pp. 146-157, DOI: 10.2174/157340911795677602, 2011.
[104] D. B. Kitchen, H. Decornez, J. R. Furr, J. Bajorath, "Docking and scoring in virtual screening for drug discovery: methods and applications," Nature Reviews Drug Discovery, vol. 3 no. 11, pp. 935-949, DOI: 10.1038/nrd1549, 2004.
[105] J.-H. Chen, A. T. H. Wu, B. Lawal, D. T. W. Tzeng, J.-C. Lee, C.-L. Ho, T.-Y. Chao, "Identification of cancer hub gene signatures associated with immune-suppressive tumor microenvironment and ovatodiolide as a potential cancer immunotherapeutic agent," Cancers, vol. 13 no. 15,DOI: 10.3390/cancers13153847, 2021.
[106] B. Lawal, Y.-C. Kuo, S.-L. Tang, F.-C. Liu, A. T. H. Wu, H.-Y. Lin, H.-S. Huang, "Transcriptomic-based identification of the immuno-oncogenic signature of cholangiocarcinoma for HLC-018 multi-target therapy exploration," Cell, vol. 10 no. 11,DOI: 10.3390/cells10112873, 2021.
[107] B. Lawal, Y.-C. Kuo, M. R. Sumitra, A. T. Wu, H.-S. Huang, "In vivo pharmacokinetic and anticancer studies of HH-N25, a selective inhibitor of topoisomerase I, and hormonal signaling for treating breast cancer," Journal of Inflammation Research, vol. 14, 2021.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
Copyright © 2022 Uchenna Blessing Alozieuwa et al. This is an open access article distributed under the Creative Commons Attribution License (the “License”), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License. https://creativecommons.org/licenses/by/4.0/
Abstract
The present study evaluated the polyphenolic contents and hypoglycemic, antioxidant, and anti-inflammatory effects of the diethyl ether fraction of Thespesia garckeana using various in vitro and in vivo models. Total phenol and flavonoid contents of the extract were
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
Details







1 Department of Biochemistry, Faculty of Natural and Applied Sciences, Veritas University Abuja, FCT-Abuja, Nigeria
2 PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei and Academia Sinica, Taipei 11031, Taiwan; Graduate Institute for Cancer Biology & Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
3 Department of Biochemistry and Molecular Biology, Faculty of Science, Federal University Ndufu-Alike Ikwo, P.M., B 1010 Abakaliki, Ebonyi State., Nigeria
4 Department of Chemical Sciences, Biochemistry Unit, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria; College of Medicine, Graduate Institute of Biomedical Science, China Medical University, Taiwan
5 Department of Chemistry, Faculty of Physical Sciences, Alex-Ekwueme Federal University Ndufu-Alike, Ebonyi State, Nigeria
6 Department of Biochemistry, Federal University of Technology, Minna, Nigeria
7 Department of Chemistry, Federal University of Technology, Minna, Nigeria
8 Pharmacology Department & Health Research Unit, Medical College Jouf University, Jouf, Saudi Arabia; Pharmacology Department Faculty of Medicine, Beni-Suef University, Egypt
9 Clinical Nutrition Department, Applied Medical Sciences College, Jazan University, Jazan 82817, Saudi Arabia
10 Department of Biotechnology, College of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
11 Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, AlBeheira, Egypt
12 TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan; The PhD Program of Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; Clinical Research Center, Taipei Medical University Hospital, Taipei Medical University, Taipei 11031, Taiwan; Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 11490, Taiwan
13 PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei and Academia Sinica, Taipei 11031, Taiwan; Graduate Institute for Cancer Biology & Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; School of Pharmacy, National Defense Medical Center, Taipei 11490, Taiwan; PhD Program in Biotechnology Research and Development College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan
14 Center for Food Analysis (NAL), Technological Development Support Laboratory (LADETEC), Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro 21941-598, Brazil