Next Article in Journal
Regulation of Hydrogen Sulfide Metabolism by Nitric Oxide Inhibitors and the Quality of Peaches during Cold Storage
Previous Article in Journal
Induction of Redox-Active Gene Expression by CoCl2 Ameliorates Oxidative Stress-Mediated Injury of Murine Auditory Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Antioxidant and Anticholinesterase Activities of Macrosphyra Longistyla (DC) Hiern Relevant in the Management of Alzheimer’s Disease

by
Taiwo O. Elufioye
1,*,
Chidimma G. Chinaka
2 and
Adebola O. Oyedeji
1
1
Department of Chemistry, Walter Sisulu University, Mthatha 5117, South Africa
2
Department of Pharmacognosy, Faculty of Pharmacy, University of Ibadan, Ibadan 200284, Nigeria
*
Author to whom correspondence should be addressed.
Antioxidants 2019, 8(9), 400; https://doi.org/10.3390/antiox8090400
Submission received: 3 June 2019 / Revised: 10 July 2019 / Accepted: 20 July 2019 / Published: 16 September 2019

Abstract

:
Macrosphyra longistyla has been used in many traditional systems of medicine for its anti-hemorrhagic, antidiabetic, anti-ulcer, and anti-diarrhea properties. The acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) inhibitions of the crude methanol extracts and its various partitioned fractions were determined by a modified method of Ellman. An evaluation of the antioxidant activity was carried out using 1,1-diphenyl-2-picryl-hydrazyl (DPPH) radical scavenging, ferric reducing power, and nitric oxide scavenging assays. The total flavonoids were estimated based on the aluminum chloride method, while the total tannins and phenolics were estimated based on the vanillin–HCl and Folin–Ciocalteu method, respectively. The ethyl acetate fraction had the highest DPPH radical scavenging activity, and the highest ferric reducing power with a concentration providing 50% inhibition (IC50) of 0.079 mg/mL and 0.078 mg/mL, respectively, while the crude methanol extract had the highest nitric oxide scavenging activity with an IC50 of 0.008 mg/mL. The methanol extract had the highest phenolics and flavonoids contents, while the aqueous fraction had the highest tannin content. The crude methanol extract had the best AChE and BuChE inhibitory action, with an IC50 of 0.556 µg/mL and 5.541 µg/mL, respectively, suggesting that the plant had a better AChE inhibiting potential. A moderate correlation was observed between the phenolic content and DPPH radical scavenging, NO radical scavenging, and AChE inhibitory activities (r2 = 0.439, 0.430, and 0.439, respectively), while a high correlation was seen between the flavonoid content and these activities (r2 = 0.695, 0.724, and 0.730, respectively), and the ferric reducing antioxidant power correlated highly with the proautocyanidin content (r2 = 0.801). Gas chromatography mass spectrometry (GCMS) revealed decanoic acid methyl ester (24.303%), 11,14-eicosadienoic acid methyl ester (16.788%), linoelaidic acid (10.444%), pentadecanoic acid (9.300%), and 2-methyl-hexadecanal (9.285%). Therefore, we suggest that M. longistyla contain bioactive chemicals, and could be a good alternative for the management of Alzheimer’s and other neurodegenerative diseases.

1. Introduction

Alzheimer’s disease and other neurodegenerative conditions are usually characterized by the slow, but progressive, dysfunction and loss of neurons in the central nervous system [1]. About 55 million people are suffering from one form of neurodegenerative disease (ND) or another, with an expected rise in this figure the with increasing age of the population [2,3]. Despite the volume of research on the pathogenesis of neurodegenerative conditions, appropriate treatment is yet to be found [4]. However, several factors, including aging [5] and some pathological conditions, such as impaired mitochondrial function [6], aggregated proteins deposit [7], neuroinflammation [8], cholinergic deficit [9], and oxidative stress [10], have been associated with NDs. Thus, the management of NDs involves addressing one or more of the associated conditions. The currently available therapies for Alzheimer’s disease (AD) are cholinesterase inhibitors such as rivastigmine and donepezil, which only reduce disease progression and provide symptomatic relieve [11]. Thus, efforts are still being made to find alternative and better therapeutic options.
Macrosphyra longistyla is a shrub found in several tropical countries. It has long, arching stems that are about 4 m long [12]. M. longistyla has been used traditionally as an antihemorrhagic in Benin [13], as an antidiabetic in Nigeria and Côte d’Ivoire [14,15], as a contraceptive and for the restoration of fertility [16], for ulcers [17], and for diarrhea [18]. Fresh wildly-collected leaves are eaten as a vegetable by the Gourmantché, Aïzo, and Cotafon people in Benin [19]. It is also widely consumed in Togo [20]. The leaves have been suggested an indigenous food ingredient for complementary food formulations to combat infant malnutrition [21]. To the best of our knowledge, the chemical constituents and biological activity of this plant have not been reported in the literature. This study therefore investigates its anticholinesterase and anti-oxidant potentials, as well as phytochemical characterization.

2. Materials and Methods

2.1. Plant Material

The leaves of Macrosphyra longistyla were collected from Agbogi village in Osun State in December 2017. The plant was identified and authenticated by Mr. Odewo of the Forest Herbarium Ibadan (FHI), with voucher number FHI 112042. The voucher specimen were deposited at the herbarium of the Department of Pharmacognosy, University of Ibadan.

2.2. Plant Extraction and Partitioning

The leaves were air-dried and pulverized. About 2.25 kg of the powdered leaf was macerated using 100% methanol. The extract was filtrated using a Buchner funnel, and concentrated in vacuo so as to obtain a crude methanol extract. Then, 80 g of the crude methanol extract was partitioned into n-hexane, ethyl acetate, and water, to obtain the respective fractions, which were concentrated in vacuo and used for the subsequent experiments. The percentage yield of both extract and fractions were determined.

2.3. Phytochemical Screening

The preliminary phytochemical screening of the crude methanol extract was carried out using standard procedures. These include tests for alkaloids using Dragendorff and Wagner reagents, the Borntrager’s test for anthraquinones, and a ferric chloride test for phenolic compounds [22,23].

2.4. Determination of the Total Phenol Content (TPC)

The total phenol content in the methanolic extract and various fractions of M. longistyla were determined based on a previously described procedure [24]. Then, 2.5 mL of 10% Folin–Ciocalteau’s reagent was mixed with 2 mL of 2% sodium carbonate solution (Na2CO3), followed by the addition of 0.5 mL of methanolic extract and fractions of M. longistyla (1 mg/mL). The mixture was incubated at 45 °C for 15 min, and absorbance was taken at 765 nm. The quantification was done with respect to the standard of gallic acid at different concentrations (1, 0.5, 0.25, 0.125, 0.063, and 0.031 mg/mL). The content of the total phenolic compounds was calculated based on a standard curve prepared using gallic acid and expressed as milligrams of gallic acid equivalent (GAE) per gram of sample.

2.5. Determination of Total Flavonoid Contents (TFC)

The total flavonoid content was determined using the aluminum chloride colorimetric method [25]. In this method, 1 mL of crude extract or fractions of M. longistyla were mixed with 3 mL of methanol, followed by 0.2 mL of 10% aluminum chloride (AlCl3), 0.2 mL of potassium acetate (1 M), and 5.6 mL of distilled water, and left at room temperature for 30 min. Absorbance was taken at 420 nm. Quantification was done with respect to the standard of gallic acid at different concentrations (1, 0.5, 0.25, 0.125, 0.063, and 0.031 mg/mL). The total phenolic content was calculated based on a standard curve prepared using gallic acid, and expressed as milligrams of gallic acid equivalent (GAE) per gram of sample.

2.6. Determination of Pro-Anthocyanidin Content (PAC)

The vanillin–HCl method was used for the quantitative determination of condensed tannins (proanthocyanidins) [26]. In this method, 3 mL of 4% vanillin in methanol, and 1.5 mL of hydrochloric acid (HCl) was added to 0.5 mL of extract/fractions (1 mg/mL). The mixture was vortexed thoroughly and allowed to stand for 15 min at room temperature. Absorbance was read at 500 nm. A calibration curve was prepared using a standard gallic acid solution. All of the results were expressed as mg gallic acid equivalents (GAE) per gram of sample.

2.7. DPPH (2,2-Diphenyl-1-Picrylhydrazyl Hydrate) Radical Scavenging Assay

The radical scavenging ability of the fractions was determined using the stable radical DPPH (2,2-diphenyl-1-picrylhydrazyl hydrate), as previously described [27]. In this assay, 1 ml of 0.1 mM DPPH was mixed with 1 mL of crude extract and fractions of M. longistyla at different concentrations (1, 0.5, 0.25, 0.125, 0.063, and 0.031 mg/mL), as well as the positive controls (ascorbic acid and 2,6-di-tert-butyl-4-methylphenol (DDM)) at different concentrations (1, 0.5, 0.25, and 0.125 mg/mL). The reaction was vortexed and left in the dark at room temperature for 30 min, after which the absorbance was taken at 517 nm. The percentage inhibition was calculated as follows:
I% = [(Ablank − Asample)/Ablank] × 100
where Ablank is the absorbance of the control reaction (containing all reagents except the test compound), and Asample is the absorbance of the test compound. The sample concentration providing 50% inhibition (IC50) was also calculated.

2.8. Nitric Oxide (NO) Scavenging Assay

The nitric oxide scavenging assay was carried out as previously described [28]. First, 2 mL of sodium nitroprusside was mixed with 0.5 mL of phosphate buffer pH 7.4 and 0.5 mL of different concentrations of extract (0.0031–1.0 mg/mL). The mixture was incubated at 25 °C for 150 min, and an initial absorbance (A0) was taken at 540 nm. Thereafter, 0.5 mL of the incubated mixture was mixed with 1 mL of a sulfanilic acid reagent and 1 mL of naphthylethylenediamine dichloride (0.1% w/v), and incubated at room temperature for 30 min, before another absorbance (A1) was taken at 540 nm. The same reaction mixture without the extract but with the equivalent amount of methanol served as the negative control. Ascorbic acid and DDM at various concentrations were used as the standard. All of the experiments were in triplicates. The percentage nitrite radical scavenging activity of the extracts and standard were calculated using the following formula:
% inhibition of NO = [A0 − A1]/A0 × 100
where A0 is the absorbance before the reaction, and A1 is the absorbance after the reaction.

2.9. Ferric Reducing Antioxidant Assay

The reducing power was determined according to the method of Oyaizu [29]. Substances with a reducing ability react with potassium ferricyanide (Fe3+) to form potassium ferrocyanide (Fe2+), which then reacts with ferric chloride to form a ferric ferrous complex that has an absorption maximum at 700 nm. Briefly, 0.2 mL of various concentrations of plant extract and fractions was mixed with 0.2 mL of phosphate buffer and 0.2 mL of potassium ferricyanide. The mixture was vortexed and incubated at 50 °C for 20 min. After cooling, 0.2 mL of 10% trichloroacetic acid (TCA) was then added to the mixture and centrifuged at 4500 rpm for 10 min. Then, 100 µL of the upper solution was mixed with 20 µL of the ferric chloride solution and 100 µL of distilled water. The absorbance was taken at 700 nm. The control was prepared in a similar manner, but without the test sample. Ascorbic acid and DDM at various concentrations were used as the standard. The experiments were done in triplicates.

2.10. Cholinesterase Inhibitory Assay

Acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) inhibitions were determined spectrophotometrically using acetylcholine iodide and butrrylcholine iodide as substrates, respectively, by a modified method of Ellman [30]. The serial dilutions of the fractions were subjected to this test using eserin and donepezil as the positive control. Then, 5 mg of both the extract and fractions were dissolved in 1 mL of methanol. Serial dilutions of each sample were done in order to obtain the final concentrations of 1, 0.5, 0.25, 0.125, and 0.0625 mg/mL, while the positive controls (eserin and donepezil) were also diluted serially to obtain the final concentrations of 0.1, 0.05, 0.025, 0.0125, 0.00625, and 0.003125 mg/mL. Thereafter, 20 µL of each concentration was pipetted into the micro plates, followed by 240 µL of the phosphate buffer (pH 8) and 20 µL of the enzyme, which was then vortexed. The plates were then incubated at 37 ºC for 30 min. After incubation, 20 µL of 25 mM of the substrate (acetylthiocholinecholine iodide (ATChI) or butyrylthiocholine chloride (BTChCl)) was added to the reaction mixture, followed by the addition of 20 µL of 10 mM 5, 5ʹ-Dithiobis-2-nitrobenzoic acid (DTNB). The hydrolysis of acetylcholine iodide or butryrylthiocholine chloride was determined spectrophotometrically at 412 nm. The assay was carried out in triplicates, with methanol as the negative control. The percentage inhibition was computed using the following formula:
Δ a Δ b Δ a × 100
where Δa is the change in absorbance of the negative control, and Δb is the change in absorbance of the sample.

2.11. Gas Chromatography Mass Spectrometry (GCMS) Analysis

One microliter (1 µL) of the sample diluted in hexane was analyzed on a Bruker 450 gas chromatography-300 mass spectrometer (GCMS) system operating in EI mode at 70 eV, equipped with a HP-5 MS fused silica capillary system with a 5% phenylmethylsiloxane stationary phase. The capillary column parameter was 30 m by 0.25 mm, while the film thickness was 0.25 µm. The initial temperature of the column was set at 70 °C, and heated to 240 °C at a rate of 5 °C/min, with the final temperature kept at 450 °C. The run time was 66.67 min, and helium was used as the carrier gas at a flow rate of 1 min/min. The split ratio was 100:1. The scan time was 78 min, with a scanning range of 35 to 450 amu.

2.12. Statistical Analysis

All of the data were analyzed using GraphPad Prism 6.0, and were expressed as mean ± standard error of the mean (SEM). The correlation and regression analysis of the activities (Y) versus the total phytochemical content (X) were carried out using the online Quest Graph™ Linear, Logarithmic, Semi-Log Regression Calculator [31].

3. Results and Discussion

M. longistyla has been reportedly used for managing different ailments in traditional medicine [13,14,15,16,17,18]. In an ethnomedical survey carried out by us, the plant was mentioned as a memory enhancer. Thus, the present study was carried out to investigate its phytochemical content, as well as evaluate the antioxidant and cholinesterase inhibitory activities of the extracts and partitioned fractions.
The preliminary phytochemical screening of the methanol extract revealed the presence of tannins, flavonoids, phenolics, terpenoids, and saponins. Anthraquinones and alkaloids were, however, found absent in the plant (Table 1).
The percentage yield of the extract and fractions (expressed as weight of extract/fraction relative to the weight of the initial plant material) ranged from 4.70% to 40.00%, with the highest being the aqueous fraction (Table 2). This suggests that the polar solvent was able to extract more constituents, probably because of the solubility of the polar compounds present in the plant material.
Furthermore, the content of the phenols (TPC), flavonoids (TFC), and the tannins (PAC) was estimated quantitatively. The TPC, as determined by the Folin–Ciocalteu method, ranged from 7.56 ± 0.12 to 18.30 ± 0.04 mg GAE/g of extract (Table 2). Both the crude extract and the various fractions had an appreciable total phenolic content, with the methanol extract and the ethyl acetate fractions having the highest TPC, while the n-hexane fraction had the least TPC. The total flavonoid and proauthocyanidin contents, also reported as mg GAE/g of extract, showed that the TFC ranged from 5.02 ± 0.01 to 16. 07 ± 0.14 mg GAE/g of extract, while the PAC ranged from 2.99 ± 0.06 to 26.11 ± 0.02. In both cases, the hexane fraction had the least amount (Table 2).
Phenolic compounds are present in plant tissues and serve as antioxidants [32], because of the presence of hydroxyl groups, which are responsible for their scavenging ability. Thus, they are capable of reacting with active oxygen radicals such as hydroxyl radicals [33]. Flavonoids are polyphenolic compounds, and are responsible for some of the health benefits of vegetable and fruits [34]. They are known to play an active role in the quenching of free radicals, because of their redox properties [35]. Tannins, however, are a high molecular weight polyphenolic that have also been implicated as antioxidants [36].
The antioxidant activity of the extract and fractions was evaluated by the DPPH and NO radical scavenging activity, as well as the ferric reducing power, while the AChE inhibitory activity was evaluated by Ellman’s colorimetric assay.
DPPH is usually reduced by a hydrogen donating compound, leading to its change in color, from deep violet to light yellow, which can be monitored spectrophotometrically [37]. The DPPH radical scavenging activity results are as shown in Figure 1, while the IC50—the concentration of antioxidant (extract/fractions) required for 50% scavenging of DPPH radicals—values are given in Table 3. From the results, the ethyl acetate fraction had the highest activity, with an IC50 value of 0.078.
Nitric oxide is important in the regulation of several physiological processes, and several diseases have been associated with a high concentration of NO [38]. The nitric oxide scavenging activity can be determined by estimating for nitrate and nitrite, using the Greiss Illosvoy reaction [39]. At a physiological pH (7.2), sodium nitroprusside decomposes in an aqueous solution to produce NO, which reacts with oxygen to form stable products—nitrate and nitrite. Scavengers of NO compete with oxygen, leading to a reduced production of nitrite ions [40].
In the nitric oxide scavenging assay, all of the extract and fractions exhibited a good scavenging effect, with the methanol extract having the best scavenging effect (IC50 = 0.008), followed by an aqueous fraction (IC50 = 0.010) and then the ethyl acetate fraction (IC50 = 0.056; Figure 2 and Table 3).
Ferric reducing power is well linked with antioxidant activity [41], and compounds with a reducing effect are usually electron donors that can reduce oxidized intermediates of lipid peroxidation processes, thus acting as primary or secondary antioxidants [33]. In the ferric reducing antioxidant assay, the methanol extract, aqueous fraction, and the ethyl acetate fraction had good reducing activity (Figure 3), with IC50 values of 0.051, 0.009, and 0.078 respectively.
On the whole, a better antioxidant activity was observed in the polar fractions, and this could be because of the abundant presence of major secondary metabolites, such as tannins and flavonoids, in these fractions, as supported by the higher TPC and TFC in these fractions. Phenolics are free-radical terminators [33], thus having protective effects against many infectious and neuro degenerative diseases such AD [42].
The inhibition of cholinesterase enzymes is considered promising in the management of neurological and neurodegenerative disorders such as AD, senile dementia, ataxia, and myasthenia gravis, where a deficit in cholinergic neurotransmission is often observed [43,44]. Compounds with a dual inhibitory effect on AChE and BuChE are also considered better, as BuChE also plays a minor role in the regulation of AChE [45,46].
In this study, the methanol extract inhibited the acetylcholinesterase enzyme the most, followed by the ethyl acetate and aqueous fractions, with respective percentage inhibitions of 81.629 ± 0.02, 76.985 ± 0.04, and 71.778 ± 0.01 (Figure 4). The hexane fraction had the least inhibitory action, suggesting that the active constituents are likely to be polar. The study also suggests a better inhibition of AChE as compared to BuChE, as both the crude extract and the various fractions had a lower percentage inhibition and higher IC50 values in the later enzyme (Figure 5 and Table 4).
The cholinesterase inhibitory activity of several medicinal plants has been reported in the literature [47,48,49,50,51]. Also, antioxidants such as vitamin E and vitamin C have been reportedly associated with a decrease in AD incidence and prevalence, [52] and AD patients on high doses of antioxidants were reported to have a slower rate of cognitive deterioration [53]. Thus, the good antioxidant and anticholinesterase activities of polar fractions in this study suggest that these fractions are good sources of phenolic compounds, with potential cholinesterase inhibitory and antioxidant properties that may find usefulness in the management of AD. This is the first report of such activities in Macrosphyra longistyla.
We also correlated the phytochemical content with the observed activities of the plant. Several pharmacological effects of the plant extract such, as being anti-inflammatory, antioxidant, and antimicrobial, have also been associated with the presence of phenolic compounds [54,55], and r2 values have been used to show the relationship between the phytochemical constituents and activities of medicinal plants [56]. There was a moderate correlation between the total phenolic content and the DPPH and NO radical scavenging, as well as the AChE inhibitory activities (r2 = 0.439, 0.430, and 0.439, respectively). However, a better correlation was observed between the flavonoid content and these activities (r2 = 0.695, 0.724, and 0.730, respectively), while the ferric reducing antioxidant power correlated with the proautocyanidin content (r2 = 0.801; Table 5).
Finally, the identification of possible compounds in the non-polar (hexane) fraction using GC-MS revealed the presence of twenty-three compounds (Table 6). The most abundant was decanoic acid methyl ester (24.303%), followed by 11,14-eicosadienoic acid methyl ester (16.788%), linoelaidic acid (10.444%), pentadecanoic acid (9.300%), and 2-methyl-hexadecanal (9.285%).

4. Conclusions

This study revealed the antioxidant and anticholinesterase activities of the compounds present in M. longistyla, and suggest the potential use of extracts from this plant for the management of neurodegenerative conditions. The polar fractions had the highest antioxidants and anticholinesterase constituents, which can be further exploited. Also, the GCMS analysis identified the compounds likely to contribute to the observed activities.

Author Contributions

Conceptualization, T.O.E.; data curation, C.G.C.; funding acquisition, T.O.E.; investigation, C.G.C.; methodology, T.O.E.; project administration, A.O.O.; resources, A.O.O.; supervision, T.O.E.; writing (original draft), C.G.C.; writing (review and editing), T.O.E. and A.O.O.

Funding

This work is based on the research supported in part by the National Research Foundation of South Africa (Grant number UID 110789) and the TETFUND Research Project (RP) intervention of Nigeria.

Acknowledgments

The authors hereby acknowledge the financial support of Walter Sisulu University.

Conflicts of Interest

Authors declare no conflict of interest.

References

  1. Amor, S.; Puentes, F.; Baker, D.; Van Der Valk, P. Inflammation in neurodegenerative diseases. Immunology 2010, 129, 154–169. [Google Scholar] [CrossRef]
  2. Chen, H.; Kwong, J.C.; Copes, R.; Tu, K.; Villeneuve, P.J.; Van Donkelaar, A.; Hystad, P.; Martin, R.V.; Murray, B.J.; Jessiman, B.; et al. Living near major roads and the incidence of dementia, Parkinson’s disease, and multiple sclerosis: A population-based cohort study. Lancet 2017, 389, 718–726. [Google Scholar] [CrossRef]
  3. Prince, M.; Guerchet, M.; Prina, M. The Epidemiology and Impact of Dementia: Current State and Future Trends; World Health Organization: Geneva, Switzerland, 2015. [Google Scholar]
  4. Solanki, I.; Parihar, P.; Mansuri, M.L.; Parihar, M.S. Flavonoid-Based Therapies in the Early Management of Neurodegenerative Diseases. Adv. Nutr. 2015, 6, 64–72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Tan, C.-C.; Yu, J.-T.; Tan, M.-S.; Jiang, T.; Zhu, X.-C.; Tan, L. Autophagy in aging and neurodegenerative diseases: Implications for pathogenesis and therapy. Neurobiol. Aging 2014, 35, 941–957. [Google Scholar] [CrossRef] [PubMed]
  6. Johri, A.; Beal, M.F. Mitochondrial Dysfunction in Neurodegenerative Diseases. J. Pharmacol. Exp. Ther. 2012, 342, 619–630. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Takalo, M.; Salminen, A.; Soininen, H.; Hiltunen, M.; Haapasalo, A. Protein aggregation and degradation mechanisms in neurodegenerative diseases. Am. J. Neurodegener. Dis. 2013, 2, 1–14. [Google Scholar]
  8. Chen, W.-W.; Zhang, X.; Huang, W.-J. Role of neuroinflammation in neurodegenerative diseases (Review). Mol. Med. Rep. 2016, 13, 3391–3396. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Schliebs, R.; Arendt, T. The cholinergic system in aging and neuronal degeneration. Behav. Brain Res. 2011, 221, 555–563. [Google Scholar] [CrossRef] [PubMed]
  10. Butterfield, D.A.; Swomley, A.M.; Sultana, R. Amyloid β-Peptide (1–42)-Induced Oxidative Stress in Alzheimer Disease: Importance in Disease Pathogenesis and Progression. Antioxid. Redox Signal. 2013, 19, 823–835. [Google Scholar] [CrossRef]
  11. Kumar, A.; Singh, A. A review on Alzheimer’s disease pathophysiology and its management: An update. Pharmacol. Rep. 2015, 67, 195–203. [Google Scholar] [CrossRef]
  12. Keay, R.W. Randia and Gardenia in West Africa; Bulletin du Jardin botanique de l’Etat, Bruxelles/Bulletin van den Rijksplantentuin: Brussel, Belgium, 1958; pp. 15–72. [Google Scholar]
  13. Klotoé, J.; Dougnon, T.V.; Koudouvo, K.; Atègbo, J.-M.; Loko, F.; Akoègninou, A.; Aklikokou, K.; Dramane, K.; Gbeassor, M. Ethnopharmacological Survey on Antihemorrhagic Medicinal Plants in South of Benin. Eur. J. Med. Plants 2013, 3, 40–51. [Google Scholar] [CrossRef]
  14. Olabanji, S.; Adebajo, A.; Omobuwajo, O.; Ceccato, D.; Buoso, M.; Moschini, G. PIXE analysis of some Nigerian anti-diabetic medicinal plants (II). Nucl. Instrum. Methods Phys. Res. Sect. B Beam Interact. Mater. Atoms 2014, 318, 187–190. [Google Scholar] [CrossRef]
  15. Konkon, N.G.; Ouatara, D.; Kpan, W.B.; Kouakou, T.H. Medicinal plants used for treatment of diabetes by traditional practitioners in the markets of Abidjan district in Côte d’Ivoire. J. Med. Plants Stud. 2017, 5, 39–48. [Google Scholar]
  16. Soladoye, M.O.; Ikotun, T.; Chukwuma, E.C.; Ariwaodo, J.O.; Ibhanesebor, G.A.; Agbo-Adediran, O.A.; Owolabi, S.M. Our plants, our heritage: Preliminary survey of some medicinal plant species of Southwestern University Nigeria Campus, Ogun State, Nigeria. Annu. Biol. Res. 2013, 4, 27–34. [Google Scholar]
  17. Allabi, A.C.; Busia, K.; Ekanmian, V.; Bakiono, F. The use of medicinal plants in self-care in the Agonlin region of Benin. J. Ethnopharmacol. 2011, 133, 234–243. [Google Scholar] [CrossRef]
  18. Agbankpé, A.J.; Dougnon, T.V.; Bankolé, H.S.; Yèhouénou, B.; Yédomonhan, H.; Lègonou, M.; Dougnon, T.J. Etude ethnobotanique des légumes feuilles thérapeutiques utilisés dans le traitement des diarrhées au sud-Bénin (Afrique de l’Ouest). Int. J. Biol. Chem. Sci. 2014, 8, 1784–1795. [Google Scholar] [CrossRef]
  19. Achigan-Dako, E.G.; Pasquini, M.W.; Assogba Komlan, F.; N’danikou, S.; Yédomonhan, H.; Dansi, A.; Ambrose-Oji, B. Traditional Vegetables in Benin; Institut National des Recherches Agricoles du Bénin, Imprimeries du CENAP: Cotonou, Benin, 2010; p. 174. [Google Scholar]
  20. Atato, A.; Wala, K.; Batawila, K.; Lamien, N.; Akpagana, K. Edible wild fruit highly consumed during food shortage period in Togo: State of knowledge and conservation status. J. Life Sci. 2011, 5, 1046–1057. [Google Scholar]
  21. Chadare, F.J.; Madode, Y.E.; Fanou-Fogny, N.; Kindossi, J.M.; Ayosso, J.O.; Honfo, S.H.; Kayodé, A.P.; Linnemann, A.R.; Hounhouigan, D.J. Indigenous food ingredients for complementary food formulations to combat infant malnutrition in Benin: A review. J. Sci. Food Agric. 2018, 98, 439–455. [Google Scholar] [CrossRef]
  22. Sofowora, A. Screening Plants for Bioactive Agents. In Medicinal Plants and Traditional Medicinal in Africa, 2nd ed.; Spectrum Books Ltd.: Ibadan, Nigeria, 1993; pp. 134–156. [Google Scholar]
  23. Trease, G.E.; Evans, W.C. Pharmacognosy, 15th ed.; Saunders Publishers: London, UK, 2002; pp. 221–393. [Google Scholar]
  24. Singleton, V.L.; Rossi, J.A. Colorimetric of total phenolics with phosphomolybdic-phosphotungstic acid reagents. Am. J. Enol. Vinic. 1965, 16, 144–158. [Google Scholar]
  25. Köksal, E.; Gülçin, I. Antioxidant activity of cauliflower (Brassica oleracea L.). Turk. J. Agric. For. 2008, 32, 65–78. [Google Scholar]
  26. Broadhurst, R.B.; Jones, W.T. Analysis of condensed tannins using acidified vanillin. J. Sci. Food Agric. 1978, 29, 788–794. [Google Scholar] [CrossRef]
  27. Brand-Williams, W.; Cuvelier, M.; Berset, C. Use of a free radical method to evaluate antioxidant activity. LWT 1995, 28, 25–30. [Google Scholar] [CrossRef]
  28. Aiyegoro, O.A.; Okoh, A.O. Preliminary phytochemical screening and In vitro antioxidant activities of the aqueous extract of Helichrysum longifolium DC. BMC Complement. Altern. Med. 2010, 10, 21. [Google Scholar] [CrossRef] [PubMed]
  29. Oyaizu, M. Studies on products of browning reaction. Antioxidative activities of products of browning reaction prepared from glucosamine. Jpn. J. Nutr. Diet. 1986, 44, 307–315. [Google Scholar] [CrossRef]
  30. Ellman, G.L.; Courtney, K.; Andres, V.; Featherstone, R.M. A new and rapid colorimetric determination of acetylcholinesterase activity. Biochem. Pharmacol. 1961, 7, 88–95. [Google Scholar] [CrossRef]
  31. Quest Graph™. Linear, Logarithmic, Semi-Log Regression Calculator; AAT Bioquest, Inc.: Sunnyvale, CA, USA; Available online: https://www.aatbio.com/tools/linear-logarithmic-semi-log-regression-online-calculator (accessed on 20 May 2019).
  32. Shahidi, F.; Ambigaipalan, P. Phenolics and polyphenolics in foods, beverages and spices: Antioxidant activity and health effects—A review. J. Funct. Foods 2015, 18, 820–897. [Google Scholar] [CrossRef]
  33. Brewer, M. Natural Antioxidants: Sources, Compounds, Mechanisms of Action, and Potential Applications. Compr. Rev. Food Sci. Food Saf. 2011, 10, 221–247. [Google Scholar] [CrossRef]
  34. Han, R.-M.; Zhang, J.-P.; Skibsted, L.H. Reaction Dynamics of Flavonoids and Carotenoids as Antioxidants. Molecules 2012, 17, 2140–2160. [Google Scholar] [CrossRef] [Green Version]
  35. Wolfe, K.; Wu, X.; Liu, R.H. Antioxidant Activity of Apple Peels. J. Agric. Food Chem. 2003, 51, 609–614. [Google Scholar] [CrossRef]
  36. Koleckar, V.; Kubikova, K.; Řeháková, Z.; Kuca, K.; Jun, D.; Jahodář, L.; Opletal, L. Condensed and Hydrolysable Tannins as Antioxidants Influencing the Health. Mini-Rev. Med. Chem. 2008, 8, 436–447. [Google Scholar] [CrossRef]
  37. Kedare, S.B.; Singh, R.P. Genesis and development of DPPH method of antioxidant assay. J. Food Sci. Technol. 2011, 48, 412–422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Pacher, P.; Beckman, J.S.; Liaudet, L. Nitric Oxide and Peroxynitrite in Health and Disease. Physiol. Rev. 2007, 87, 315–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Garrat, D.C. The Quantitative Analysis of Drugs; Chapman and Hall Ltd.: Tokyo, Japan, 1964; Volume 3, pp. 456–458. [Google Scholar]
  40. Al-Majedy, Y.K.; Al-Amiery, A.A.; Kadhum, A.A.H.; Mohamad, A.B. Antioxidant Activities of 4-Methylumbelliferone Derivatives. PLoS ONE 2016, 11, 0156625. [Google Scholar] [CrossRef] [PubMed]
  41. Benzie, I.F.; Strain, J. Ferric reducing/antioxidant power assay: Direct measure of total antioxidant activity of biological fluids and modified version for simultaneous measurement of total antioxidant power and ascorbic acid concentration. In Methods in Enzymology; Elsevier BV: Amsterdam, The Netherlands, 1999; Volume 299, pp. 15–27. [Google Scholar]
  42. Aliev, G.; Obrenovich, M.E.; Reddy, V.P.; Shenk, J.C.; Moreira, P.I.; Nunomura, A.; Zhu, X.; Smith, M.A.; Perry, G. Antioxidant Therapy in Alzheimer’s Disease: Theory and Practice. Mini-Rev. Med. Chem. 2008, 8, 1395–1406. [Google Scholar] [CrossRef] [PubMed]
  43. Schneider, L.S. Treatment of Alzheimer’s disease with cholinesterase inhibitors. Clin. Geriatr. Med. 2001, 17, 337–358. [Google Scholar] [CrossRef]
  44. Ranjan, N.; Kumari, M. Acetylcholinesterase inhibition by medicinal plants: A Review. Ann. Plant Sci. 2017, 6, 1640–1644. [Google Scholar] [CrossRef] [Green Version]
  45. Venneri, A.; McGeown, W.J.; Shanks, M.F. Empirical evidence of neuroprotection by dual cholinesterase inhibition in Alzheimer’s disease. NeuroReport 2005, 16, 107–110. [Google Scholar] [CrossRef]
  46. Khan, I.; Nisar, M.; Khan, N.; Saeed, M.; Nadeem, S.; Rehman, F.U.; Ali, F.; Karim, N.; Kaleem, W.A.; Qayum, M.; et al. Structural insights to investigate Conypododiol as a dual cholinesterase inhibitor from Asparagus adscendens. Fitoterapia 2010, 81, 1020–1025. [Google Scholar] [CrossRef]
  47. Mathew, M.; Subramanian, S. In Vitro Screening for Anti-Cholinesterase and Antioxidant Activity of Methanolic Extracts of Ayurvedic Medicinal Plants Used for Cognitive Disorders. PLoS ONE 2014, 9, e86804. [Google Scholar] [CrossRef]
  48. Owokotomo, I.; Ekundayo, O.; Abayomi, T.; Chukwuka, A. In-vitro anti-cholinesterase activity of essential oil from four tropical medicinal plants. Toxicol. Rep. 2015, 2, 850–857. [Google Scholar] [CrossRef] [Green Version]
  49. Samaradivakara, S.P.; Samarasekera, R.; Handunnetti, S.M.; Weerasena, O.J. Cholinesterase, protease inhibitory and antioxidant capacities of Sri Lankan medicinal plants. Ind. Crop. Prod. 2016, 83, 227–234. [Google Scholar] [CrossRef]
  50. Sheeja, M.D.; Beema, S.R.; Karutha, P.S.; Pandima, D.K. Cholinesterase inhibitory, anti-amyloidogenic and neuroprotective effect of the medicinal plant Grewia tiliaefolia—An in vitro and in silico study. Pharm. Biol. 2017, 55, 381–393. [Google Scholar] [CrossRef] [PubMed]
  51. Ovais, M.; Ayaz, M.; Khalil, A.T.; Shah, S.A.; Jan, M.S.; Raza, A.; Shahid, M.; Shinwari, Z.K. HPLC-DAD finger printing, antioxidant, cholinesterase, and α-glucosidase inhibitory potentials of a novel plant Olax nana. BMC Complement. Altern. Med. 2018, 18, 1. [Google Scholar] [CrossRef] [PubMed]
  52. Steele, M.; Stuchbury, G.; Münch, G. The molecular basis of the prevention of Alzheimer’s disease through healthy nutrition. Exp. Gerontol. 2007, 42, 28–36. [Google Scholar] [CrossRef] [PubMed]
  53. Mecocci, P.; Polidori, M.C. Antioxidant clinical trials in mild cognitive impairment and Alzheimer’s disease. Biochim. Biophys. Acta Mol. Basis Dis. 2012, 1822, 631–638. [Google Scholar] [CrossRef] [PubMed]
  54. Zhang, L.; Ravipati, A.S.; Koyyalamudi, S.R.; Jeong, S.C.; Reddy, N.; Smith, P.T.; Bartlett, J.; Shanmugam, K.; Münch, G.; Wu, M.J. Antioxidant and Anti-inflammatory Activities of Selected Medicinal Plants Containing Phenolic and Flavonoid Compounds. J. Agric. Food Chem. 2011, 59, 12361–12367. [Google Scholar] [CrossRef] [PubMed]
  55. Gülçin, İ.; Huyut, Z.; Elmastaş, M.; Aboul-Enein, H.Y. Radical scavenging and antioxidant activity of tannic acid. Arab. J. Chem. 2010, 3, 43–53. [Google Scholar] [CrossRef] [Green Version]
  56. Babbar, N.; Oberoi, H.S.; Uppal, D.S.; Patil, R.T. Total phenolic content and antioxidant capacity of extracts obtained from six important fruit residues. Food Res. Int. 2011, 44, 391–396. [Google Scholar] [CrossRef]
  57. Hussain, A.; Tian, M.-Y.; He, Y.-R.; Bland, J.M.; Gu, W.-X. Behavioral and electrophysiological responses of Coptotermes formosanus Shiraki towards entomopathogenic fungal volatiles. Biol. Control 2010, 55, 166–173. [Google Scholar] [CrossRef]
  58. Choi, H.-S. Analysis of the Terpenoids from Syneilesis palmata Essential Oil and the Variation of the Sesquiterpene Compounds by Harvest Year. Korean J. Food Nutr. 2013, 26, 287–294. [Google Scholar] [CrossRef]
  59. Palani, S.; Raja, S.; Kumar, R.P.; Selvaraj, R.; Kumar, B.S. Evaluation of phytoconstituents and anti-nephrotoxic and antioxidant activities of Monochoria vaginalis. Pak. J. Pharm. Sci. 2011, 24, 293–301. [Google Scholar] [PubMed]
  60. Ntalli, N.G.; Manconi, F.; Leonti, M.; Maxia, A.; Caboni, P. Aliphatic Ketones from Ruta chalepensis (Rutaceae) Induce Paralysis on Root Knot Nematodes. J. Agric. Food Chem. 2011, 59, 7098–7103. [Google Scholar] [CrossRef] [PubMed]
  61. Walia, M.; Mann, T.S.; Kumar, D.; Agnihotri, V.K.; Singh, B. Chemical Composition and In Vitro Cytotoxic Activity of Essential Oil of Leaves of Malus domestica Growing in Western Himalaya (India). Evid. Based Complement. Altern. Med. 2012, 2012, 649727. [Google Scholar] [CrossRef] [PubMed]
  62. Innocent, E.; Gikonyo, N.K.; Nkunya, M.H. Repellency property of long chain aliphatic methyl ketones against Anopheles gambiae s.s. Tanzan. J. Health Res. 2008, 10, 50–54. [Google Scholar]
  63. Manosroi, A.; Jantrawut, P.; Sainakham, M.; Manosroi, W.; Manosroi, J. Anticancer activities of the extract from Longkong (Lansium domesticum) young fruits. Pharm. Biol. 2012, 50, 1397–1407. [Google Scholar] [CrossRef] [PubMed]
  64. Hernández-Villegas, M.; Borges-Argáez, R.; Rodríguez-Vivas, R.; Torres-Acosta, J.; Méndez-González, M.; Cáceres-Farfán, M.; Torres-Acosta, J.F.D.J. In vivo anthelmintic activity of Phytolacca icosandra against Haemonchus contortus in goats. Veter Parasitol. 2012, 189, 284–290. [Google Scholar] [CrossRef] [PubMed]
  65. Hamza, F.; Zinjarde, S. Marine Biodiversity As a Resource for Bioactive Molecules As Inhibitors of Microbial Quorum Sensing Phenotypes. In Biotechnological Applications of Quorum Sensing Inhibitors; Springer Science and Business Media LLC: Berlin/Heidelberg, Germany, 2018; pp. 329–350. [Google Scholar]
  66. Carretero, M.; López-Pérez, J.L.; Abad, M.; Bermejo, P.; Tillet, S.; Israel, A.; Noguera, P.B. Preliminary study of the anti-inflammatory activity of hexane extract and fractions from Bursera simaruba (Linneo) Sarg. (Burseraceae) leaves. J. Ethnopharmacol. 2008, 116, 11–15. [Google Scholar] [CrossRef]
  67. Silici, S.; Kutluca, S. Chemical composition and antibacterial activity of propolis collected by three different races of honeybees in the same region. J. Ethnopharmacol. 2005, 99, 69–73. [Google Scholar] [CrossRef]
  68. Conley, A.J.; Kabara, J.J. Antimicrobial Action of Esters of Polyhydric Alcohols. Antimicrob. Agents Chemother. 1973, 4, 501–506. [Google Scholar] [CrossRef] [Green Version]
  69. Santos, C.C.; Salvadori, M.S.; Mota, V.G.; Costa, L.M.; de Almeida, A.A.; de Oliveira, G.A.; Costa, J.P.; de Sousa, D.P.; de Freitas, R.M.; de Almeida, R.N. Antinociceptive and antioxidant activities of phytol in vivo and in vitro models. Neurosci. J. 2013, 2013, 949452. [Google Scholar] [CrossRef]
  70. Elufioye, T.O.; Obuotor, E.M.; Agbedahunsi, J.M.; Adesanya, S.A. Cholinesterase inhibitory activity and structure elucidation of a new phytol derivative and a new cinnamic acid ester from Pycnanthus angolensis. Rev. Bras. Farm. 2016, 26, 433–437. [Google Scholar] [CrossRef] [Green Version]
  71. Ghalib, R.M.; Hashim, R.; Sulaiman, O.; Mehdi, S.H.; Anis, Z.; Rahman, S.Z.; Ahamed, B.K.; Abdul Majid, A.M. Phytochemical analysis, cytotoxic activity and constituents–activity relationships of the leaves of Cinnamomum iners (Reinw. ex Blume-Lauraceae). Nat. Prod. Res. 2012, 26, 2155–2158. [Google Scholar] [PubMed]
  72. Gecibesler, I.H.; Yaglıoglu, A.S.; Gul, F.; Temirturk, M.; Demirtas, I. Phytochemicals of Chrysophthalmum montanum (DC.) Boiss. Roots and their antiproliferative activities Against HeLa and C6 Cell Lines. Proc. Natl. Acad. Sci. India Sect. B Biol. Sci. 2019, 89, 145–154. [Google Scholar] [CrossRef]
  73. El-Din, S.M.M.; Mohyeldin, M.M. Component Analysis and Antifungal Activity of the Compounds Extracted from Four Brown Seaweeds with Different Solvents at Different Seasons. J. Ocean Univ. China 2018, 17, 1178–1188. [Google Scholar] [CrossRef]
  74. Conforti, F.; Menichini, F.; Loizzo, M.R.; Statti, A.G.; Rapisarda, A.; Menichini, F.; Houghton, P.J. Antioxidant, α-amylase inhibitory and brine-shrimp toxicity studies on Centaurea centaurium L. methanolic root extract. Nat. Prod. Res. 2008, 22, 1457–1466. [Google Scholar] [CrossRef]
  75. Vakharia, P.P.; Silverberg, J.I. New therapies for atopic dermatitis: Additional treatment classes. J. Am. Acad. Dermatol. 2018, 78, S76–S83. [Google Scholar] [CrossRef]
  76. Jíchová, Š.; Kopkan, L.; Husková, Z.; Doleželová, Š.; Neckář, J.; Kujal, P.; Vernerová, Z.; Kramer, H.J.; Sadowski, J.; Kompanowska-Jezierska, E.; et al. Epoxyeicosatrienoic acid analog attenuates the development of malignant hypertension, but does not reverse it once established: A study in Cyp1a1-Ren-2 transgenic rats. J. Hypertens. 2016, 34, 2008–2025. [Google Scholar] [CrossRef]
  77. Sales, D.L.; Oliveira, O.P.; Cabral, M.E.; Dias, D.Q.; Kerntopf, M.R.; Coutinho, H.D.; Costa, J.G.; Freitas, F.R.; Ferreira, F.S.; Alves, R.R.; et al. Chemical identification and evaluation of the antimicrobial activity of fixed oil extracted from Rhinella jimi. Pharm. Biol. 2015, 53, 98–103. [Google Scholar] [CrossRef]
  78. Nagarjunakonda, S.; Amalakanti, S.; Dhishana, S.R.; Ramaiah, M.; Rajanala, L. GC-MS Analysis of Indrakeeladri Native Medicine used in the Treatment of Stroke. Pharmacogn. J. 2017, 9, 102–106. [Google Scholar] [CrossRef]
  79. Sandoval-Montemayor, N.E.; García, A.; Elizondo-Treviño, E.; Garza-González, E.; Alvarez, L.; Camacho-Corona, M.D.R. Chemical Composition of Hexane Extract of Citrus aurantifolia and Anti-Mycobacterium tuberculosis Activity of Some of Its Constituents. Molecules 2012, 17, 11173–11184. [Google Scholar] [CrossRef]
  80. Mormile, R.; Vittori, G.; De Michele, M.; Squarcia, U.; Quaini, F. Linoleic acid and colorectal cancer cell growth suppression: Is the deregulation of mitochondrial surviving the key factor? Int. J. Colorectal Dis. 2012, 27, 1383–1384. [Google Scholar] [CrossRef] [PubMed]
  81. Kim, Y.-S.; Lee, S.-J.; Hwang, J.-W.; Kim, E.-K.; Kim, S.-E.; Kim, E.-H.; Moon, S.-H.; Jeon, B.-T.; Park, P.-J. In vitro protective effects of Thymus quinquecostatus Celak extracts on t-BHP-induced cell damage through antioxidant activity. Food Chem. Toxicol. 2012, 50, 4191–4198. [Google Scholar] [CrossRef] [PubMed]
  82. Gok, M.; Zeybek, N.D.; Bodur, E. Butyrylcholinesterase expression is regulated by fatty acids in HepG2 cells. Chem. Interact. 2016, 259, 276–281. [Google Scholar] [CrossRef] [PubMed]
  83. Agboke, A.A.; Attama, A.A. Bioactive components and antibacterial activities of n-hexane extract of Moringa oleifera root bark on clinical isolates of methicilin resistant Staphylococcus aureus. Int. J. Curr. Res. Chem. Pharm. Sci. 2016, 3, 1–9. [Google Scholar]
  84. Avis, T.J.; Boulanger, R.R.; Belanger, R.R. Synthesis and Biological Characterization of (Z)-9-Heptadecenoic and (Z)-6-Methyl-9-Heptadecenoic Acids: Fatty Acids with Antibiotic Activity Produced by Pseudozyma flocculosa. J. Chem. Ecol. 2000, 26, 987–1000. [Google Scholar] [CrossRef]
  85. Wang, J.; Liu, H.; Gao, H.; Zhao, J.; Zhou, L.; Han, J.; Yu, Z.; Yang, F. Antimicrobial and antioxidant activities of the flower essential oil of Halimodendron halodendron. Nat. Prod. Commun. 2011, 6. [Google Scholar] [CrossRef]
Figure 1. 1,1-diphenyl-2-picryl-hydrazyl (DPPH) radical scavenging activity of extract and fractions of M. longistyla. Data are expressed as mean ± standard deviation (SD; n = 3). ME—methanol extract; HF—hexane fraction; EF—ethyl acetate fraction; AF—aqueous fraction; DDM—2,6-di-tert-butyl-4-methylphenol.
Figure 1. 1,1-diphenyl-2-picryl-hydrazyl (DPPH) radical scavenging activity of extract and fractions of M. longistyla. Data are expressed as mean ± standard deviation (SD; n = 3). ME—methanol extract; HF—hexane fraction; EF—ethyl acetate fraction; AF—aqueous fraction; DDM—2,6-di-tert-butyl-4-methylphenol.
Antioxidants 08 00400 g001
Figure 2. Nitic oxide (NO) radical scavenging activity of the extract and fractions of M. longistyla. Data are expressed as mean ± SD (n = 3). ME—methanol extract; HF—hexane fraction; EF—ethyl acetate fraction; AF—aqueous fraction; DDM—2,6-di-tert-butyl-4-methylphenol.
Figure 2. Nitic oxide (NO) radical scavenging activity of the extract and fractions of M. longistyla. Data are expressed as mean ± SD (n = 3). ME—methanol extract; HF—hexane fraction; EF—ethyl acetate fraction; AF—aqueous fraction; DDM—2,6-di-tert-butyl-4-methylphenol.
Antioxidants 08 00400 g002
Figure 3. Ferric reducing activity of the extract and fractions of M. longistyla. Data are expressed as mean ± SD (n = 3). ME—methanol extract; HF—hexane fraction; EF—ethyl acetate fraction; AF—aqueous fraction; DDM—2,6-di-tert-butyl-4-methylphenol.
Figure 3. Ferric reducing activity of the extract and fractions of M. longistyla. Data are expressed as mean ± SD (n = 3). ME—methanol extract; HF—hexane fraction; EF—ethyl acetate fraction; AF—aqueous fraction; DDM—2,6-di-tert-butyl-4-methylphenol.
Antioxidants 08 00400 g003
Figure 4. Acetylcholinesterase inhibitory activity of the extract and fractions of M. longistyla. Data are expressed as mean ± SD (n = 3). ME—methanol extract; HF—hexane fraction; EF—ethyl acetate fraction; AF—aqueous fraction.
Figure 4. Acetylcholinesterase inhibitory activity of the extract and fractions of M. longistyla. Data are expressed as mean ± SD (n = 3). ME—methanol extract; HF—hexane fraction; EF—ethyl acetate fraction; AF—aqueous fraction.
Antioxidants 08 00400 g004
Figure 5. Butryrylcholinesterase inhibitory activity of the extract and fractions of M. longistyla. Data are expressed as mean ± SD (n = 3). ME—methanol extract; HF—hexane fraction; EF—ethyl acetate fraction; AF—aqueous fraction.
Figure 5. Butryrylcholinesterase inhibitory activity of the extract and fractions of M. longistyla. Data are expressed as mean ± SD (n = 3). ME—methanol extract; HF—hexane fraction; EF—ethyl acetate fraction; AF—aqueous fraction.
Antioxidants 08 00400 g005
Table 1. Phytochemical screening results of Macrosphyra longistyla.
Table 1. Phytochemical screening results of Macrosphyra longistyla.
TestsObservationsInferences
1. Alkaloids
 a. DragendorffDeep yellow colorAlkaloid absent
 b. Wagner testOrange colorAlkaloid absent
2. Anthraquinones
a. Borntrager’s testMilky color Anthraquinone absent
3. FlavonoidsYellow coloration Flavonoids present
4. PhenolsDark colorationPhenols present
5. TanninsBlue black colorationTannin present
6. SaponinFrothing which disappear after sometimeSaponin present
7. TerpenoidDark green colorationTerpenoids present
Table 2. The total phenolics, flavonoids, and authocyanidins content in the extract and fractions of M. longistyla.
Table 2. The total phenolics, flavonoids, and authocyanidins content in the extract and fractions of M. longistyla.
AssaysMEHFEFAF
% Yield6.184.707.1140.00
Total phenolics (mg GAE/g)18.30 ± 0.047.56 ± 0.1216.06 ± 0.139.02 ± 0.02
Total flavonoids (mg GAE/g)16. 07 ± 0.145.02 ± 0.0110.49 ± 0.01411.62 ± 0.01
Total tannins (mg GAE/g)24. 44 ± 0.322.99 ± 0.069.12 ± 0.1726.11 ± 0.02
Data are expressed as mean ± standard error of the mean (SEM; n = 3). GAE—gallic acid equivalent; ME—methanol extract; HF—hexane fraction; EF—ethyl acetate fraction; AF—aqueous fraction.
Table 3. The concentration providing 50% inhibition (IC50) values of the different antioxidant assays.
Table 3. The concentration providing 50% inhibition (IC50) values of the different antioxidant assays.
AssaysIC50
MEHFEFAFAscorbic AcidDDM
DPPH scavenging0.0900.3630.0790.0890.0060.050
NO scavenging0.0085.6780.0560.0100.0720.063
Ferric reducing0.0510.0870.0780.0090.0530.003
ME—methanol extract; HF—hexane fraction; EF—ethyl acetate fraction; AF—aqueous fraction; DDM—2,6-di-tert-butyl-4-methylphenol; DPPH—1,1-diphenyl-2-picryl-hydrazyl; NO—nitric oxide.
Table 4. IC50 values for the cholinesterase inhibitory assay. AChE—acetylcholinesterase; BuChE—butyrylcholinesterase.
Table 4. IC50 values for the cholinesterase inhibitory assay. AChE—acetylcholinesterase; BuChE—butyrylcholinesterase.
AssaysIC50
MEHFEFAFEserinDonepezil
AChE0.55625.8710.9140.8460.0020.001
BuChE5.54111.95723.338ND0.0020.001
ND: Not determined.
Table 5. Correlation of the total phenolic, total flavonoid, and proautocyanidin contents with antioxidant and anticholinesterase activities.
Table 5. Correlation of the total phenolic, total flavonoid, and proautocyanidin contents with antioxidant and anticholinesterase activities.
Assaysr2 Values
Total PhenolicsTotal FlavonoidsProautocyanidin
DPPH scavenging0.4390.6950.515
NO scavenging0.4300.7240.558
Ferric reducing0.0120.2760.801
AChE inhibition0.4390.7300.557
BuChE inhibition0.001540.1310.325
Table 6. Compounds identified through gas chromatography mass spectrometry (GCMS).
Table 6. Compounds identified through gas chromatography mass spectrometry (GCMS).
S/NName of Identified CompoundsRetention Time (min)% AbundanceMolecular FormulaClass of CompoundReported Biological EffectReferences
12,6,8-trimethyl-decane27.0840.084C13H28AlkaneAntifungal[57]
22-methyl-hexadecanal30.8339.285C17H34O AldehydeAntifungal[58]
3Z,Z,Z-1,4,6,9-nonadecatetraene34.1470.349C19H32AlkeneAntioxidant[59]
42-dodecanone34.8310.357C12H24OAliphatic ketones Nematocidal [60]
52-pentadecanone35.5911.630C15H30OKetoneCytotoxic and repellant[61,62]
617-octadecanal39.2110.119C18H36OLong-chain aldehydeNRNR
7Hexadecanoic acid39.5640.152C16H32O2Saturated fatty acid Anticancer and anthelmintic[63,64]
82-methyl-dodecanoic acid43.0710.283C11H22O2Fatty acidAntimicrobial[65]
9Neophytadiene43.4820.109C20H38SesquiterpeneAnti-inflammatory[66]
102-nonadecanone43.6111.942C19H38OAlkanoneAntimicrobial [67]
11Decanoic acid methyl ester46.41424.303C11H22O2Fatty acid esterAntimicrobial[68]
12Phytol47.0530.202C20H40ODiterpene alcoholAntinociceptive, antioxidant, and anticholinesterase[69,70]
13Eicosanoic acid ethyl ester48.5855.265C22H44O2Fatty acidAnticancer[71]
14Pentadecanoic acid48.5929.300C15H30O2Saturated fatty acidAnthelmintic [64]
15tetradecanoic acid-12-methyl-methyl ester49.5800.048C16H32O2Fatty acidAnticancer and antifungal[72,73]
1611,14-eicosadienoic acid methyl ester51.56216.788C21H38O2Fatty acidAntioxidant and anti-amylase[74]
178,11,14-ecosatrienoic acid51.7122.299C20H34O2Omega fatty acidAtopic dermatitis and malignant hypertension[75,76]
18Z-methyl-hexadec-11-enoate51.9634.204C17H32O2Fatty acid methyl esterAntimicrobial[77]
19Dodecanoic acid-10-methyl-methyl ester52.6431.957C14H28O2Fatty acid methyl esterAnticoagulant[78]
20Linoelaidic acid53.55910.444C18H32O2 Omega-6 trans fatty acid Anticholinesterase, anti-mycobacterium, anticancer, and antioxidant [79,80,81,82]
21Z,E-3,13-octadecadien-1-ol53.7632.707C18H34OFatty alcoholAntimicrobial[83]
22(Z)-methyl-Heptadec-9-enoate53.9530.179C18H34O2Fatty acidAntibiotic[84]
23Hexadecanoic acid-2-methyl-methyl ester54.6151.139C18H36O2Fatty acid methyl esters Antimicrobial and antioxidant[85]
NR: Not reported. The various identified compounds have been reported to have different biological effects, such as being antimicrobial, antioxidant, anticoagulant, anticholinesterase, anticancer, and anthelmintic. All of these ultimately contribute to the overall activity of the plant.

Share and Cite

MDPI and ACS Style

Elufioye, T.O.; Chinaka, C.G.; Oyedeji, A.O. Antioxidant and Anticholinesterase Activities of Macrosphyra Longistyla (DC) Hiern Relevant in the Management of Alzheimer’s Disease. Antioxidants 2019, 8, 400. https://doi.org/10.3390/antiox8090400

AMA Style

Elufioye TO, Chinaka CG, Oyedeji AO. Antioxidant and Anticholinesterase Activities of Macrosphyra Longistyla (DC) Hiern Relevant in the Management of Alzheimer’s Disease. Antioxidants. 2019; 8(9):400. https://doi.org/10.3390/antiox8090400

Chicago/Turabian Style

Elufioye, Taiwo O., Chidimma G. Chinaka, and Adebola O. Oyedeji. 2019. "Antioxidant and Anticholinesterase Activities of Macrosphyra Longistyla (DC) Hiern Relevant in the Management of Alzheimer’s Disease" Antioxidants 8, no. 9: 400. https://doi.org/10.3390/antiox8090400

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

Article Metrics

Back to TopTop