You are on page 1of 6

SAJB-01948; No of Pages 6

South African Journal of Botany xxx (2017) xxx–xxx

Contents lists available at ScienceDirect

South African Journal of Botany

journal homepage: www.elsevier.com/locate/sajb

Mangiferin from Mangifera indica fruits reduces post-prandial glucose


level by inhibiting α-glucosidase and α-amylase activity
V. Sekar, S. Chakraborty, S. Mani, V.K. Sali, H.R. Vasanthi ⁎
Natural Products Research Laboratory, Department of Biotechnology, Pondicherry University, Puducherry 605014, India

a r t i c l e i n f o a b s t r a c t

Article history: Background: Mangiferin, is a xanthone glycoside mainly present in Mangifera indica fruits and few other
Received 20 November 2017 medicinal plants that are cultivated in the tropical and sub-tropical regions. Mangiferin has a wide spectrum of
Received in revised form 17 January 2018 biomedical properties from anti-microbial to anti-cancer activities.
Accepted 1 February 2018 Aim: In the present study, the mangiferin content in the methanolic extract of unripe and ripe mango pulp
Available online xxxx
was compared and quantified by Reverse Phase HPLC method. Further, inhibitory action of mangiferin on α-
Edited by Kannan Ragupathi Raja Rengasamy
glucosidase and α-amylase enzymes which play a vital role in the regulation of serum glucose level was studied.
Materials and methods: The enzyme inhibitory nature of mangiferin was tested using in silico docking analysis
Keywords: with Autodock software and was further confirmed by in vitro α-glucosidase and α-amylase biochemical assays.
Mangiferin Results: The results of this study reveal that, mangiferin content was higher in ripe methanolic extract than in
RP HPLC unripe. Further, mangiferin exhibited better enzyme inhibitory action in silico with α-glucosidase with a binding
α-Glucosidase energy of −7.4 kcal/mol than α-amylase. This was concurred with α-glucosidase in vitro assays wherein the IC50
α-Amylase of mangiferin was 36.84 μg/ml and that of ripe mango extract was 112.8 μg/ml when compared to the standard
Mangifera indica acarbose 21.33 μg/ml.
Conclusion: Evidently, it can be concluded that mangiferin from Mangifera indica fruits slows down the glucose
metabolism and thereby could be used as a possible hypoglycemic agent owing to its enzyme inhibitory
properties.
© 2017 SAAB. Published by Elsevier B.V. All rights reserved.

1. Introduction affects the pathophysiology and ensures complications like nephropa-


thy, retinopathy or neuropathy (Brownlee, 2001; Goh and Cooper,
The recent statistics on the steady increase in the number of 2008).
diabetics across the world poses a major challenge for biomedical The therapeutic strategies for managing diabetes encompasses a
researchers and health care professionals. Diabetes mellitus (DM) is a range of drugs, including biguanides, prescribed as first line of medica-
metabolic disorder characterized by chronic hyperglycemia due to tion, to decrease glucose production in the liver. Also, the sulfonylureas
abnormality in insulin secretion or insulin receptor or defects in metab- stimulates the pancreatic β cells for insulin release (Gupta et al., 2016)
olism of carbohydrates in association with liver or kidney or pancreatic One such therapeutic option to subside the post-prandial hyperglyce-
damage. In addition to these factors, DM can also be very much influ- mia is the enzyme inhibitors and acarbose is the most commonly
enced by genetic susceptibility, sedentary and other harmful lifestyle used enzyme inhibitor (Obih et al., 2016). Alpha-glucosidase and
practices such as alcoholism, smoking, etc. (Marx, 2002). The persistent α-amylase are the two enzymes responsible for the hydrolysis of
factor which propels DM as a challenging disease is the elevated blood long chain complex carbohydrates into simple chain glucose molecules
glucose level or hyperglycemia. Chronic hyperglycemia, besides being to facilitate their transport inside the cells. Acarbose acts as an inhibitor
deleterious to the metabolism, on a cellular level reacts with structural to these enzymes, hence ensures the delay in the increase of post
or plasma proteins – glycation reaction which results in advanced prandial hyperglycemic state after a meal (Baron, 1998; Chiasson
glycated end products (AGE). The AGEs modulates the structural et al., 2002). Although, acarbose is a promising therapeutic option for
integrity, stability and function of the proteins and thereby promptly diabetics, but in the long run, like any other therapeutic drug it does
have a few setbacks that cannot be overlooked. Continous intake of
acarbose are proven to exhibit side effects like stomach distention,
flatulence, diarrhoea, etc. (Chiasson et al., 2002).
⁎ Corresponding author at: Department of Biotechnology, School of Life Sciences,
Pondicherry University, Puducherry 605014, India.
It is not an entirely a new concept of phytochemical intervention
E-mail addresses: hannah.dbt@pondiuni.edu.in, hrvasanthi@gmail.com when modern therapeutics takes a step down in its full potential to
(H.R. Vasanthi). treat a disease, as phytomedicines have been used extensively for

https://doi.org/10.1016/j.sajb.2018.02.001
0254-6299/© 2017 SAAB. Published by Elsevier B.V. All rights reserved.

Please cite this article as: Sekar, V., et al., Mangiferin from Mangifera indica fruits reduces post-prandial glucose level by inhibiting α-glucosidase
and α-amylase activity, South African Journal of Botany (2017), https://doi.org/10.1016/j.sajb.2018.02.001
2 V. Sekar et al. / South African Journal of Botany xxx (2017) xxx–xxx

metabolism related disorders since time immemorial. Several medicinal enzyme solution was pre-incubated with the test sample and standard
plants such as Tinospora cordifolia (Sangeetha et al., 2011), Salacia drug for 5 min. The reaction was initiated by adding 370 mM of sucrose,
reticulata (Li et al., 2008; Im et al., 2009) and many other Indian medic- after incubation at 37 °C for 30 min and terminated by heating at
inal plants (Grover et al., 2002) are being used to treat diabetes and its 90–100 °C. The glucose is measured by GOD-POD method at
complications. Mangifera indica known for its drupe fruit is also 546 nm using a semi auto-analyser. The enzyme activity is directly pro-
known for its anti-diabetic potential (Wauthoz et al., 2007). portional to the liberated glucose. Acarbose was used as a positive
It is interesting to note that mangiferin, a polyphenol present in control.
many medicinal plants which exhibits hypoglycemic potential is also
present in mangoes. Herein, we have evaluated the level of mangiferin 2.5.2. Alpha amylase assay
in both ripe and unripe Mangifera indica fruits and their potential as The α-amylase assay was performed by protocol of (Kusano et al.,
enzyme inhibitors of α-glucosidase and α-amylase using in silico and 2010) with slight modifications. Substrate was prepared by dissolving
in vitro assays. 200 mg starch in 25 ml of sodium hydroxide (0.4 M) by heating at
100 °C for 5 min. After cooling, pH was adjusted to 7.0 and the final
2. Materials and methods volume was made up to 100 ml using distilled water. Acarbose was
used as a positive control. Forty microliter of substrate solution was
2.1. Chemicals pre-incubated at 37 °C for 3 min with 20 μl of acarbose and test material
at varying concentrations, followed by 20 μl of 3 U/ml α-amylase
Alpha-amylase from Aspergillus oryzae and mangiferin were pur- (20 mM phosphate buffer with 6.7 mM Nacl, pH 6.9) and incubated at
chased from Sigma Aldrich, USA. Alpha-glucosidase from Saccharomyces 37 °C for 15 min. Termination of the reaction was carried out by adding
cervesiae and HPLC grade solvents were purchased from SRL Chemicals, 80 μl of HCl (0.1 M), followed by addition of 100 μl of iodine reagent
Mumbai, India. Acarbose was purchased from Santa Cruz Biotechnology, (2.5 mM). The absorbance was measured at 630 nm.
USA. All other solvents and chemicals used were of analytical grade.
2.6. Statistical analysis
2.2. Preparation of plant extracts
All the data were expressed as mean ± SEM of triplicates. IC50 values
Fresh unripe and ripe Totapuri mangoes were procured from a near-
were determined from dose-inhibition (curve fit) using non-linear re-
by organic farm in Puducherry. The pulp of ripe and unripe mangoes
gression in Graph Pad Prism Software, version 5 (Graph Pad Software,
were removed manually and crushed. The pulp was subjected to
San Diego, CA, USA).
Soxhlet extraction using 95% methanol for 6–8 h. The extract was con-
centrated using a rotary evaporator under reduced pressure
(Sellamuthu et al., 2009). This extract was used for further analysis. 3. Results and discussion

2.3. Quantification of mangiferin by HPLC Dietary recommendations to cure and prevent DM include fresh
vegetables and fruits. Misconception, moderate awareness and
Reverse Phase High Performance Liquid Chromatography (RP-HPLC) inadequate research in functional foods have led the growing popula-
was performed to check and quantify the mangiferin content in both the tion to approach for synthetic therapy in the developing countries
ripe and unripe pulp extracts. C18-250X4.60 mm 5 μm, 100 Å column (Lock et al., 2005). Owing to the advancement of ripening in fruits,
was used to separate mangiferin. The mobile phase consisting of they contain low starch, high sugars, polyphenols, flavonoids etc.
HPLC grade water with potassium phosphate monobasic and ortho- (Moneruzzaman et al., 2008). In the present study in order to identify
phosphoric acid and the stationary phase was acetonitrile was used if mangoes can be considered as functional food, the quantity of
for the analysis. A flow rate of 0.5 ml/min was maintained and the mangiferin, the principle bioactive compound, was measured in
UV–visible detector wavelength was set at 254 nm (Geodakyan et al., ripe as well as unripe mangoes and identified its α-glucosidase and
1992). α-amylase inhibiting potential to act as an agent against post-prandial
hyperglycemia.
2.4. In silico docking analysis
3.1. Quantification of mangiferin in mango pulp
Molecular docking of mangiferin with the two enzymes α-amylase
and α-glucosidase was done to identify the binding of the protein Mangiferin is a C-glycosyl xanthone, chemically known as C2-β-D-
with the ligand. Hence, the details of α-glucosidase (PDB ID 5NN4.A) glucopyranosyl- 1,3,6,7-tetrahydroxyxanthone (Fig. 1) and Mangifera
and α-amylase (PDB ID 4X9Y) were retrieved from the Protein Data indica is the primary and easily available source of mangiferin. The
Bank (PDB) (www.pdb.org/pdb). Using Pymol, the structures of the content of the same is found in different parts of the tree which includes
receptors were prepared for docking by removing the co-crystallized the leaves, fruits, seed kernels, peels, heartwood, and stem bark and the
ligand and additional water molecules to make it as a nascent receptor.
Ligand structures were generated by Chemsketch (www.acdlabs.com)
in mol format and the generated structures were converted into
PDB format using Chimera (http://www.cgl.ucsf.edu/chimera). Subse-
quently, the generated PDB structures were used for docking by using
flexible docking protocol (Trott and Olson, 2010), Auto dock (Auto
dock tools- 1.5.4 version).

2.5. Enzyme inhibitory assays

2.5.1. Alpha glucosidase assay


Alpha-glucosidase inhibitory assay is based on the influence of
α-glucosidase for the breakdown of maltose to glucose and was
performed with few modifications (Li et al., 2004). Briefly, 0.6 U/ml of Fig. 1. Chemical structure of Mangiferin.

Please cite this article as: Sekar, V., et al., Mangiferin from Mangifera indica fruits reduces post-prandial glucose level by inhibiting α-glucosidase
and α-amylase activity, South African Journal of Botany (2017), https://doi.org/10.1016/j.sajb.2018.02.001
V. Sekar et al. / South African Journal of Botany xxx (2017) xxx–xxx 3

latter contains the highest mangiferin content comparatively (Barreto anti-diabetic agent, the mangiferin content in the ripe and unripe pulp
et al., 2008). methanolic extracts were checked and quantified. The chromatograms
As this study was mainly focused on the mangiferin present in the are shown in (Fig. 2) and the estimated amount based on the retention
pulps of ripe and unripe Mangifera indica fruits to consider it as an time and peak area are given in Table 1). Identification and comparison

A)

B)

C)

Fig. 2. HPLC Chromatogram. Representative HPLC profile A) standard mangiferin B) methanloic extract of unripe mango pulp (* mangiferin peak) C) methanolic extract of ripe mango pulp
(* mangiferin peak).

Please cite this article as: Sekar, V., et al., Mangiferin from Mangifera indica fruits reduces post-prandial glucose level by inhibiting α-glucosidase
and α-amylase activity, South African Journal of Botany (2017), https://doi.org/10.1016/j.sajb.2018.02.001
4 V. Sekar et al. / South African Journal of Botany xxx (2017) xxx–xxx

Table 1 its chemical structure obeys the Lipinski's rule of five (Lipinski et al.,
Quantification of mangiferin based on the HPLC chromatogram. 1997) and is vividly discussed (Saha et al., 2016). However, there are
Sample Retention time Area Quantity major concerns on the bioavailability of mangiferin both in vivo and
Mangiferin 7.237 30143678 100 μg/ml
in vitro. For instance, better than the pure mangiferin, mangiferin com-
Unripe mango pulp 7.639 24746181 1.32 μg/g plexed with phospholipids have better bioavailability in rats (Ma et al.,
Ripe mango pulp 7.127 363836699 3.06 μg/g 2014). Similarly, mangiferin with soya phospholipid complex was prov-
en to have 9.75 fold increase in the bioavailability in vivo (Bhattacharyya
et al., 2014). A study on the pharmacokinetics of mangiferin and its me-
of mangiferin present in the extracts were correlated with the chro- tabolite in rodent model by administration of Rhizoma Anemarrhenae
matogram of standard mangiferin (Fig. 2A). Based on the retention decoction yet again revealed that mangiferin's bioavailabilty could be
time and peak area, the concentration of mangiferin from ripe mango vastly improved by complex interactions with other compounds (Tian
pulp extract was 3.06 μg/g while in unripe mango pulp extract was et al., 2016). In addition, to the mangiferin complexes, researchers
1.32 μg/g. The exact reason why mangiferin content was higher in ripe have developed derivatives of parent mangiferin to improve the phar-
mango pulp extract than in unripe mango pulp extract remains to be macological activities. Esterified derivatives of mangiferin were studied
unclear as the biological synthesis of mangiferin is a multi-step process and identified to show improved hypoglycemic and lipid solubility
(Ehianeta et al., 2016). Mangiferin embodies as an orally active drug as properties (Xue-Jian et al., 2013). Earlier study on the inhibitory activity

A) B)

C) D)

Fig. 3. Docking analysis. In silico docking analysis A) mangiferin with α-glucosidase B) mangiferin with α-amylase C) acarbose with α-glucosidase D) acarbose with α-amylase.

Please cite this article as: Sekar, V., et al., Mangiferin from Mangifera indica fruits reduces post-prandial glucose level by inhibiting α-glucosidase
and α-amylase activity, South African Journal of Botany (2017), https://doi.org/10.1016/j.sajb.2018.02.001
V. Sekar et al. / South African Journal of Botany xxx (2017) xxx–xxx 5

Table 2
Binding energy and active site amino acids on docking mangiferin with respective proteins.

S.No. Ligand-protein Binding energy (kcal/mol) Amino acids

1. Mangiferin Vs α-Glucosidase −7.4 Asp-1157, Asp-1279, Arg-1510,Lys-1460 and His-1584


2. Mangiferin Vs α-Amylase −5.67 `Asp-195, Asp-197, Glu-233, Glu-240 and Asp-300
3 Acarbose Vs α-Glucosidase −7.3 Arg- 608, Tyr- 360, His- 717
4 Acarbose Vs α-Amylase −7.1 Ser- 19, Asn-20, Arg-413, Gly-349

against protein tyrosine phosphatase 1B (PTP1B), to study the anti-dia- The IC50 values of each sample corresponding to the assays are tabulated
betic and obesity activity of benzyl derivatives of mangiferin revealed in (Table 3). As expected, mangiferin showed better enzyme inhibitory
better activity than the mangiferin molecule (Hu et al., 2007). These activity with IC50 of α-glucosidase 36.84 μg/ml and that of α-amylase is
studies on the derivatives provide better insight on the specific pharma- 63.57 μg/ml. Ripe mango extracts exhibited high enzyme inhibitory
cological actions and better understanding on the structure–activity re- potential against α-glucosidase with an IC50 112.8 μg/ml and that of
lationships. The bioavailability and liposolubility concern of mangiferin α-amylase is 287.6 μg/ml. The unripe mango pulp extract exhibited an
in the mammalian system has to be evaluated to consider the ripe enzyme inhibition of α-glucosidase corresponding to 127.0 μg/ml and
Mangifera indica fruits as an anti-diabetic agent. that of α-amylase as 318.1 μg/ml. It is interesting to note that, several
in vitro and in silico studies have been carried out in mangiferin from
3.2. Enzyme inhibitory activity by in silico analysis various sources (Picot et al., 2017). However, our team mainly focused
on mangiferin, from ripe and unripe Mangifera indica fruit pulps, to
Alpha-glucosidase and α-amylase are two key enzymes in justify the claim that they serve as an anti-diabetic functional food.
dietary carbohydrate metabolism. Alpha-amylase (1,4-α-D-glucan- To aid in food selection for diabetic population, the Glycemic Index
glucanohydrolase) is a starch metabolizing enzyme which hydrolyses (GI) is formulated. GI is the post-prandial increase in blood glucose
the polysaccharides to glucose and maltose oligosaccharides after the consumption of a food item, with pure glucose as reference,
(Koukiekolo et al., 2001). These hydrolysed oligosaccharides are then GI of foods can be classified as, low (0–55), medium (55–69) and high
acted upon by α-glucosidase which is a membrane bound enzyme (≥70) (Jenkins et al., 2002). Based on our study, the mangiferin content
present in the epithelium of brush borders in the small intestines. This and enzyme inhibitory activity was more pronounced in the ripe mango
enzyme hydrolyses at the non-reducing links to release the bound methanolic extract. Mangoes fall under the low GI category which cor-
alpha-glucose and thus increasing the glucose level in the serum responds to the GI value of 51 (Atkinson et al., 2008). Thus from the
(Kimura, 2000). For the effective management of DM, inhibiting the above facts it can be inferred that ripe mangoes with more mangiferin
enzymes responsible for carbohydrate digestion ensures delay in the in- content can be considered as a potential functional food for diabetics.
crease of post prandial glucose in the serum (Bischoff, 1995). As long However, further in vivo studies are warranted to check the influence
term consumption of acarbose leads to certain complications in the di- of mangiferin and Mangifera indica fruits in the proteomic level.
gestive system, medicinal plant extracts and their bioactive components
are evaluated for enzyme inhibitory properties (Tundis et al., 2010). In 4. Conclusion
this study, mangiferin molecule was docked with the protein ligands
α-glucosidase and α-amylase. With α-glucosidase, mangiferin was In this study, mangiferin, a phytocompound with various biomedical
docked in the catalytic site and interacted with amino acids with a properties was quantified in unripe and ripe Mangifera indica fruit pulp
binding energy of −7.4 kcal/mol showing positive binding and poten- methanolic extracts and it was found that the ripe extract had a slightly
tial inhibition (Fig. 3A, Table 2). In the case of α-amylase, mangiferin higher concentration of mangiferin than the unripe extract. The enzyme
was bound with a binding energy of −5.67 kcal/mol showing positive inhibitory nature of mangiferin was evaluated both by in silico and
binding and potential inhibition (Fig. 3B, Table 2). When the docking in vitro approach employing docking analysis and enzymatic assays
interaction of mangiferin with both the enzymes were compared, the and the results revealed that mangiferin showed better alpha-
results reveal mangiferin showed better α-glucosidase inhibition than glucosidase inhibitory activity. Based on the results, it can be concluded
α-amylase as the former had higher intermolecular energy. These that the enzyme inhibitory nature of mangiferin could be an added
interactions of mangiferin with α-glucosidase and α-amylase were reason to further explore mangiferin's influence on the metabolism in
compared with the interaction of acarbose with α-glucosidase a genomic and proteomic level.
and α-amylase whose binding energies were − 7.3 kcal/mol and
− 7.1 kcal/mol, respectively. It is evident that, mangiferin's enzyme Conflict of interest
inhibitory energy is comparable to acarbose. Docking analysis results
prompted us to further explore the enzyme inhibitory activity via The authors declare that there are no conflicts of interest.
biochemical assays.
Acknowledgement
3.3. Enzyme inhibitory activity by in vitro assays
The authors would like to express their gratitude to UGC (SAP) and
To establish a relationship between enzyme inhibiting activity of DST (FIST) grants for infrastructural facilities in the Department of
mangiferin and the methanolic extracts of ripe and unripe mango Biotechnology, Pondicherry University to execute the present work.
pulp, α-glucosidase and α-amylase inhibitory assays were performed We are also grateful for the Pondicherry University Fellowship to VS
with all three samples and acarbose was used as a positive control. from UGC, DBT fellowship to SC, DST fellowship to SM and RGNF
fellowship from UGC to VKS.
Table 3
Inhibitory concentration of extracts, mangiferin and standard drug acarbose. References
IC50 (μg) Acarbose Mangiferin Mango (Ripe) Mango (Unripe) Atkinson, F.S., Foster-Powell, K., Brand-Miller, J.C., 2008. International tables of glycemic
α- amylase 11.18 μg/ml 63.57 μg/ml 287.6 μg/ml 318.1 μg/ml index and glycemic load values: 2008. Diabetes Care 31, 2281–2283.
Baron, A.D., 1998. Postprandial hyperglycaemia and α-glucosidase inhibitors. Diabetes
α- glucosidase 21.33 μg/ml 36.84 μg/ml 112.8 μg/ml 127.0 μg/ml
Res. Clin. Pract. 40, S51–S55.

Please cite this article as: Sekar, V., et al., Mangiferin from Mangifera indica fruits reduces post-prandial glucose level by inhibiting α-glucosidase
and α-amylase activity, South African Journal of Botany (2017), https://doi.org/10.1016/j.sajb.2018.02.001
6 V. Sekar et al. / South African Journal of Botany xxx (2017) xxx–xxx

Barreto, J.C., Trevisan, M.T.S., Hull, W.E., Erben, G., de Brito, E.S., Pfundstein, B., Würtele, G., Li, Y., Huang, T.H.-W., Yamahara, J., 2008. Salacia root, a unique Ayurvedic medicine,
Spiegelhalder, B., Owen, R.W., 2008. Characterization and quantitation of polyphenolic meets multiple targets in diabetes and obesity. Life Sci. 82, 1045–1049.
compounds in bark, kernel, leaves, and peel of mango (Mangifera indica L.). J. Agric. Lipinski, C.A., Lombardo, F., Dominy, B.W., Feeney, P.J., 1997. Experimental and
Food Chem. 56, 5599–5610. computational approaches to estimate solubility and permeability in drug discovery
Bhattacharyya, S., Ahmmed, S.M., Saha, B.P., Mukherjee, P.K., 2014. Soya phospholipid and development settings. Adv. Drug Deliv. Rev. 23, 3–25.
complex of mangiferin enhances its hepatoprotectivity by improving its bioavailability Lock, K., Pomerleau, J., Causer, L., Altmann, D.R., McKee, M., 2005. The global burden of
and pharmacokinetics. J. Sci. Food Agric. 94, 1380–1388. disease attributable to low consumption of fruit and vegetables: implications for
Bischoff, H., 1995. The mechanism of alpha-glucosidase inhibition in the management of the global strategy on diet. Bull. World Health Organ. 83, 100–108.
diabetes. Clin. Invest. Med. 18, 303–311. Ma, H., Chen, H., Sun, L., Tong, L., Zhang, T., 2014. Improving permeability and oral
Brownlee, M., 2001. Biochemistry and molecular cell biology of diabetic complications. absorption of mangiferin by phospholipid complexation. Fitoterapia 93, 54–61.
Nature 414, 813–820. Marx, J., 2002. Unraveling the Causes of Diabetes.
Chiasson, J.-L., Josse, R.G., Gomis, R., Hanefeld, M., Karasik, A., Laakso, M., Group, S.-N.T.R, Moneruzzaman, K.M., Hossain, A., Sani, W., Saifuddin, M., 2008. Effect of stages of
2002. Acarbose for prevention of type 2 diabetes mellitus: the STOP-NIDDM maturity and ripening conditions on the biochemical characteristics of tomato. Am.
randomised trial. Lancet 359, 2072–2077. J. Biochem. Biotechnol. 4, 336–344.
Ehianeta, T.S., Laval, S., Yu, B., 2016. Bio-and chemical syntheses of mangiferin and Obih, P.O., Nguyen, A., Obih, J.-C.A., 2016. Alpha-glucosidase inhibitory activity of
congeners. Biofactors 42, 445–458. Mormodica charantia. FASEB J. 30, 607–613.
Geodakyan, S.V., Voskoboinikova, I.V., Kolesnik, J.A., Tjukavkina, N.A., Vasiliy, L.I., Glyzin, Picot, M.C.N., Zengin, G., Mollica, A., Stefanucci, A., Carradori, S., Mahomoodally, M., 2017.
V.I., 1992. High-performance liquid chromatographic method for the determination In vitro and in silico studies of mangiferin from Aphloia theiformis on key enzymes
of mangiferin, likviritin and dihydroquercetin in rat plasma and urine. linked to diabetes type 2 and associated complications. Med. Chem. (Los Angeles)
J. Chromatogr. B Biomed. Sci. Appl. 577, 371–375. 13, 633–640.
Goh, S.-Y., Cooper, M.E., 2008. The role of advanced glycation end products in progression Saha, S., Sadhukhan, P., Sil, P.C., 2016. Mangiferin: a xanthonoid with multipotent anti-
and complications of diabetes. J. Clin. Endocrinol. Metab. 93, 1143–1152. inflammatory potential. Biofactors 42, 459–474.
Grover, J.K., Yadav, S., Vats, V., 2002. Medicinal plants of India with anti-diabetic potential. Sangeetha, M.K., Raghavendran, H.R.B., Gayathri, V., Vasanthi, H.R., 2011. Tinospora
J. Ethnopharmacol. 81, 81–100. cordifolia attenuates oxidative stress and distorted carbohydrate metabolism in
Gupta, P., Bala, M., Gupta, S., Dua, A., Dabur, R., Injeti, E., Mittal, A., 2016. Efficacy and experimentally induced type 2 diabetes in rats. J. Nat. Med. 65, 544.
risk profile of anti-diabetic therapies: conventional vs traditional drugs—a mechanistic Sellamuthu, P.S., Muniappan, B.P., Perumal, S.M., Kandasamy, M., 2009. Antihyperglycemic
revisit to understand their mode of action. Pharmacol. Res. 113, 636–674. effect of mangiferin in streptozotocin induced diabetic rats. J. Health Sci. 55, 206–214.
Hu, H., Wang, M., Zhao, Q., Liao, H., Cai, L., Song, Y., Zhang, J., Yu, S., Chen, W., Liu, C., 2007. Tian, X., Gao, Y., Xu, Z., Lian, S., Ma, Y., Guo, X., Hu, P., Li, Z., Huang, C., 2016. Pharmacoki-
Synthesis of mangiferin derivatives as protein tyrosine phosphatase 1B inhibitors. netics of mangiferin and its metabolite—Norathyriol, Part 1: systemic evaluation of
Chem. Nat. Compd. 43, 663–666. hepatic first-pass effect in vitro and in vivo. Biofactors 42, 533–544.
Im, R., Mano, H., Matsuura, T., Nakatani, S., Shimizu, J., Wada, M., 2009. Mechanisms of Trott, O., Olson, A.J., 2010. AutoDock Vina: improving the speed and accuracy of docking
blood glucose-lowering effect of aqueous extract from stems of Kothala himbutu with a new scoring function, efficient optimization, and multithreading. J. Comput.
(Salacia reticulata) in the mouse. J. Ethnopharmacol. 121, 234–240. Chem. 31, 455–461.
Jenkins, D.J.A., Kendall, C.W.C., Augustin, L.S.A., Franceschi, S., Hamidi, M., Marchie, A., Tundis, R., Loizzo, M.R., Menichini, F., 2010. Natural products as α-amylase and
Jenkins, A.L., Axelsen, M., 2002. Glycemic index: overview of implications in health α-glucosidase inhibitors and their hypoglycaemic potential in the treatment of
and disease. Am. J. Clin. Nutr. 76, 266S–273S. diabetes: an update. Mini-Rev. Med. Chem. 10, 315–331.
Kimura, A., 2000. Molecular anatomy of α-glucosidase. Trends Glycosci. Glycotechnol. 12, Wauthoz, N., Balde, A., Balde, E.S., Van Damme, M., Duez, P., 2007. Ethnopharmacology of
373–380. Mangifera indica L. bark and pharmacological studies of its main C-glucosylxanthone,
Koukiekolo, R., Desseaux, V., Moreau, Y., Marchis-Mouren, G., Santimone, M., 2001. mangiferin. Int. J. Biomed. Pharm. Sci. 1, 112–119.
Mechanism of porcine pancreatic α-amylase. FEBS J. 268, 841–848. Xue-Jian, L.I., Zheng-Cai, D.U., Huang, Y., Bu-Ming, L.I.U., Wen-Ji, H.U., Wen-Jie, L.U., Jia-Gang,
Kusano, R., Ogawa, S., Matsuo, Y., Tanaka, T., Yazaki, Y., Kouno, I., 2010. α-Amylase and D., 2013. Synthesis and hypoglycemic activity of esterified-derivatives of mangiferin.
lipase inhibitory activity and structural characterization of acacia bark Chin. J. Nat. Med. 11, 296–301.
proanthocyanidins. J. Nat. Prod. 74, 119–128.
Li, Y., Peng, G., Li, Q., Wen, S., Huang, T.H.-W., Roufogalis, B.D., Yamahara, J., 2004. Salacia
oblonga improves cardiac fibrosis and inhibits postprandial hyperglycemia in obese
Zucker rats. Life Sci. 75, 1735–1746.

Please cite this article as: Sekar, V., et al., Mangiferin from Mangifera indica fruits reduces post-prandial glucose level by inhibiting α-glucosidase
and α-amylase activity, South African Journal of Botany (2017), https://doi.org/10.1016/j.sajb.2018.02.001

You might also like