Journal Pre-proof Bioactive constituents, vitamin analysis, antioxidant capacity and α-glucosidase inhibition of Canna indica L. rhizome extracts S. Ayusman, P. Duraivadivel, H.G. Gowtham, S. Sharma, P. Hariprasad PII:
S2212-4292(18)30802-2
DOI:
https://doi.org/10.1016/j.fbio.2020.100544
Reference:
FBIO 100544
To appear in:
Food Bioscience
Received Date: 22 August 2018 Revised Date:
10 February 2020
Accepted Date: 10 February 2020
Please cite this article as: Ayusman S., Duraivadivel P., Gowtham H.G., Sharma S. & Hariprasad P., Bioactive constituents, vitamin analysis, antioxidant capacity and α-glucosidase inhibition of Canna indica L. rhizome extracts, Food Bioscience (2020), doi: https://doi.org/10.1016/j.fbio.2020.100544. This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain. © 2020 Published by Elsevier Ltd.
Author Contribution Ayusman Swain: Conceptualization, Methodology, Validation, Investigation, Formal analysis, Funding acquisition, Roles/Writing - original draft, Writing - review & editing. Duraivadivel P.: Methodology, Validation. Gowtham HG: Methodology, Validation. Satyawati Sharma: Supervision. Hariprasad P.: Conceptualization, Resources, Formal Analysis, Project administration, Supervision, Writing - review & editing.
Conflict of interest The authors declare that there is no conflict of interest
1
Bioactive constituents, vitamin analysis, antioxidant capacity and α-glucosidase inhibition
2
of Canna indica L. rhizome extracts
3
Ayusman S1., Duraivadivel P1., Gowtham H. G2., Sharma S1., and Hariprasad P.1*
4 5 6 7
1
8
2
Center for Rural Development and Technology, Indian Institute of Technology Delhi, New Delhi, India 110016 Department of Studies in Biotechnology, University of Mysore, Karnataka, India 570006
9 10 11 12
First Author: Dr. Ayusman Swain
13
*Address to Whom Correspondence
14
Dr. P. Hariprasad
15
Assistant Professor
16
Centre for Rural Development and Technology,
17
Indian Institute of Technology Delhi,
18
Hauz Khas, New Delhi, India
19
Email:
[email protected]
20
[email protected]
21 22
Phone : +91 11 2659 1195
23
Mobile: +91 8373904447
24
Fax: +91 11 2658 2037
25 26 27 28
Running Title: Canna indica rhizomes - Bioactive constituents and biological properties
29 30 31 1
32
Abstract
33
Rhizomes of Canna indica were studied to determine their potential use as a functional food, a
34
source of vitamins, nutritional and nutraceutical ingredients. Biomass and nutrient
35
characterization showed the rhizomes were a good source of fiber (25.1%), starch (28.5%), crude
36
protein (4.72%), and lipids (5.75%) with a total estimated caloric value of 423 Kcal/100 g dry
37
weight. The rhizome also had considerable amounts of minerals and vitamins. Acetone extracts
38
of rhizomes showed significantly higher antioxidant properties. The IC50 values with DPPH,
39
ABTS+. and O2.- radicals were 21, 23 and 170 µg/ml, respectively. The reducing properties
40
(FRAP and CUPRAC) and DNA protection assay were correlated with the total phenolic and
41
flavonoid content of the rhizome extracts. Acetone and methanol extracts showed protection
42
against free radical-induced DNA and protein degradation. In a β-carotene-linoleic acid model,
43
the acetone extracts significantly decreased the bleaching of β-carotenoids. In a meat model
44
system, the acetone extracts minimized the thiobarbituric acid reactive substance of ground pork
45
meat. α-Glucosidase activity was significantly inhibited using water extracts (IC50 2.35 µg/ml)
46
and acetone extracts (IC50 27.1 µg/ml). HR-LCMS/MS analysis of different extracts showed the
47
occurrence of different bioactive compounds such as rosmarinic acid, psoromic acid, usnic acid,
48
isoeugenitol, ellagic acid, coumaric acid and swietenine. The results suggested that C. indica
49
rhizomes might be a potential source of nutrients and metabolites with health benefits.
50 51
Keywords: Canna indica, Rhizome, α-Glucosidase inhibitors. Rosmarinic acid, Psoromic acid,
52
Usnic acid.
53
2
54
1. Introduction
55
"Let food be thy medicine and medicine be thy food" is the famous quote of Hippocrates.
56
Ayurveda and traditional medicine systems followed across the world suggest that people
57
consume a nutrient-dense diet to prevent or cure diseases. The traditional Indian system of
58
medicine uses different plants to cure many diseases and disorders. However, the mechanistic
59
basis of their functioning is yet to be studied for several plant species. Canna indica L. (CI)
60
(family: Cannaceae) is a tropical perennial rhizomatous herb. It grows in almost all agro-climatic
61
zones of India and is commonly known as Indian shot or Sarvajaya or Canna lily (Nirmal et al.,
62
2007; Van Jaarsveld et al., 2006). Different plant parts of Canna have been consumed from
63
ancient times. The archeological remnants of Canna plants in the regions of Ecuador and Peru
64
indicated its dominance as a staple food in the prehistoric period (Gade, 1966). Tribes of
65
Lepchas, Bhutias and Nepalis on the Indian subcontinent were known to consume the rhizome of
66
CI as food (Mishra et al., 2011). Because of its difficulty in production and processing compared
67
to other plants such as potato, cassava and maize, Canna has been underutilized. In various
68
regions of China and Vietnam, C. edulis is primarily used to extract starch and prepare
69
transparent noodles (Piyachomkwan et al., 2002; Tonwitowat, 1994). Other than starch
70
extraction, utilization of Canna rhizomes as a functional food such as a source of antioxidants,
71
vitamins, minerals, and proteins has not been studied. In folklore medicine, the medicinal values
72
of each plant part of Canna have long been documented as a diaphoretic and diuretic with fever,
73
with dropsy as a demulcent and to treat suppuration, malaria, diarrhea, rheumatism, dysentery,
74
bursitis and cuts (Duke, 1985; Odugbemi et al., 2008). However, the mechanism and metabolites
75
behind its biological function are yet to be identified.
3
76
Broadly the plant metabolites are categorized into terpenoids, phenolics and alkaloids.
77
Collectively these metabolites improve the fitness of the plant in its natural environment
78
(Harborne, 1998). They are involved in communication, reduction of abiotic stress, protection
79
against herbivory and microbial diseases (Akula & Ravishankar, 2011; Sudha & Ravishankar,
80
2002). Secondary metabolites of plant origin in crude or purified form are widely used and
81
studied for their biological function and application in managing many diseases in humans and
82
animals. In recent years the antioxidant potential of secondary metabolites, especially phenolics
83
of plant origin are extensively studied for their beneficial effects on humans and animals. Many
84
plant metabolites are the natural source of dietary ingredients, which support a healthy life.
85
Several natural antioxidants such as chlorogenic acid, caffeic acid, curcumin, gallic acid and
86
ferulic acid are reported as compounds of pharmacological importance which are used in drugs
87
for curing numerous diseases initiated by excessive reactive oxygen species (ROS) (Suhaj,
88
2006). Some plant metabolites have been shown to be lifesaving such as quinine and artemisinin
89
against the malarial pathogen (protozoan parasite) (White, 1997); oseltamivir phosphate
90
(Tamiflu) to treat influenza virus A and B infection (Ward et al., 2005); morphine used to
91
alleviate severe pain (Sneader 1996); apomorphine to treat the "hypomobility" phase of
92
Parkinson's disease (FDA, 2004); vinca alkaloids against various types of cancer (Moudi et al.,
93
2013) etc. Given the above, the present study aims to evaluate the potential of CI rhizome as a
94
source of nutrition, antioxidants and enzyme inhibitors of health importance.
95 96
2. Material and methods
97
2.1 Plant material
4
98
The rhizomes of naturally growing CI were collected from the riverbanks of the Cauvery,
99
Mysore, India. The plants were cultivated and maintained at a research field (moist soil and
100
monsoon-influenced humid climate), at the micromodel experimental field, Indian Institute of
101
Technology Delhi, India. The identification of the plant was done at the Botanical Survey of
102
India (BSI), Kolkata, West Bengal, India. The rhizomes were washed in tap water, blot-dried and
103
kept at 4°C for a maximum of 3 days. These rhizomes were used throughout the experiment
104
either as fresh or dried powder for the extraction of metabolites and phytochemical analysis. To
105
dry, the rhizomes were cut with a knife into small pieces of ~0.5 cm3 and dried in a hot air oven
106
(Acumen Labware, Haryana, India) at 45ºC for 4 days. The dried pieces of rhizomes were course
107
powdered in a mixer grinder (Zodiac MG 218 750-Watt, Preethi Kitchen Appliances Pvt. Ltd.,
108
Chennai, India).
109
2.2 Chemicals and reagents
110
The chemicals used in the study were of analytical grade. 2,2-Diphenyl-1-picryl-hydrazyl
111
(DPPH), 2,2-azinobis (3-ethyl benzothiazoline-6-sulfonic acid) diammonium salt (ABTS), 2,2-
112
azobis (2-amidinopropane) dihydrochloride (AAPH), quercetin (QCT), bovine serum albumin
113
(BSA), calf thymus DNA (CT DNA), β-carotene, linoleic acid, thiobarbituric acid (TBA) and
114
1,3,3-tetramethoxypropane and enzymes were purchased from Sigma Chemicals Co. (St. Louis,
115
MO, USA). Gallic acid (GA), butylated hydroxy anisole (BHA), butylated hydroxy toluene
116
(BHT), ascorbic acid and Folin-Ciocalteu reagent were from Hi-Media Laboratories (Mumbai,
117
India). 4-Nitrophenyl α-D-glucopyranoside (PNPG), hydrogen peroxide, 5,5-dithio-bis-(2-
118
nitrobenzoic acid (DTNB), acetylthiocholine iodide (ATCI), 2,4,6-tri(2-pyridyl)-s-triazine
119
(TPTZ) and all other chemicals and solvents were obtained from Sisco Research Laboratory
120
(Mumbai, India).
5
121
2.3 Biomass characterization
122
The proximate analysis was done including total lipid (Soxhlet extract) (AOAC, 2000)
123
and crude fiber (AOAC, 1990) contents. Free starch was determined following an earlier method
124
(Jeong et al., 2010) with some modifications. Briefly, 100 mg dry weight (dw) of powdered
125
rhizome sample was mixed with 3 ml 80% ethanol and sonicated, 150 W (Ultrasonic bath,
126
Guyson International, Leeds, UK) for 10 min at room temperature (30±2ºC). Then the sample
127
was centrifuged (CPR-24 Plus, Remi Laboratory Instruments, Maharashtra, India) at 13000 g for
128
10 min at 4°C. The supernatant was discarded and the process was repeated thrice. The pellet
129
was dried and dissolved in 2 ml distilled water. To this solution, 100 µl 2 M sodium acetate and
130
100 µl 25 U/ml α-amylase solution were added and incubated at 60°C for 2 h. The reducing
131
sugars in the supernatant were separated by centrifuging at 13000 g for 10 min at 4°C and
132
quantified using DNS reagent (Miller, 1959). Briefly, the test solution was mixed with DNS
133
reagent at 1:1 (v/v) ratio in a test tube and placed on the boiling water bath for 15 min. The tubes
134
were cooled to room temperature, 250 µl of solution was transferred to 96 well microtiter plate
135
and absorbance was read at 540 nm (Goncalve et al., 2010). The percentage of free starch was
136
determined using a standard curve of enzymatic starch (corn) hydrolysis.
137
Calorific value (CV) of dried powder of CI rhizome was estimated using a bomb
138
calorimeter (Model RSKT-6, Rico Scientific Industries, New Delhi, India). Briefly, the equation
139
used to calculate the CV is as follows. =
×
− .
ℎ
+
140
Carbon, hydrogen, nitrogen contents of the rhizome were determined using a CHN
141
analyzer (Elementar Analysensysteme GmbH, Euro-EA 3000, Langenselbold, Germany). The
142
samples were weighed (5 mg) in small tin containers in triplicate. The containers were closed 6
143
and folded over the edge and placed in the elemental analyzer. Different elements were measured
144
as a percentage of initial starting weight using the elemental analyzer software. The crude protein
145
content in the rhizome was estimated by using a Kjeldahl conversion factor of 6.26 (Tucker &
146
Debusk, 1981). Quantitative analysis of inorganic elements (essential metals) such as Na, K, Fe,
147
Ca, Mg, Mn, Cu, Co and Zn were determined using inductively coupled plasma mass
148
spectrometry (Agilent 7900 ICP-MS, Agilent Technologies, Santa Clara, CA, USA). The
149
powdered sample was digested with HNO3 (ultra-pure grade) using closed-vessel microwave
150
digestion (Titan MPS microwave sample preparation system, PerkinElmer, Inc., Waltham, MA,
151
USA). Commercially available standard solution (ICP multi-element standard solution XXI for
152
MS, Merck, Darmstadt, Germany) was used to prepare the calibration curves.
153
2.4 Analysis of vitamins
154
2.4.1 Preparation of samples
155
Vitamins (water and fat-soluble) in the dried rhizome was determined using HPLC and
156
LC-MS/MS. For HPLC, standard solutions of vitamins, A (retinoic acid), D (cholecalciferol and
157
cholecalcitriol), and E (α-tocopherol and α-tocopheryl acetate) were prepared in methanol and
158
(L-ascorbic acid) was prepared in water. Similarly, the rhizome powders (mortar ground) were
159
directly extracted with methanol and water. Vitamin standards and rhizome extracts (RE) were
160
run in the HPLC system with an RP C18 column (Agilent-1260, Agilent Technologies, Santa
161
Clara CA, USA). The specific HPLC conditions are provided in Table 1.
162
LC-MS/MS analysis was done in a Waters Acquity PDA system with an Accucore RP
163
C18 150 x 2.1 mm, 2.6 µm column with an auto-sampler (Waters UPLC-TQD, Waters India Pvt.
164
Ltd., Bangalore, India). The crude methanol and hexane extracts reconstituted with methanol and
165
were tested for fat-soluble vitamins (A, D and E). The water extract was tested for water-soluble
7
166
vitamins (B-complex and C). A gradient solvent system with (A) 0.01% TFA in water (pH 3.9)
167
and (B) methanol was applied. The gradient profile started with 95:5 (A:B) and remained
168
constant for the first 4 min then went to 2:98 (A:B) for the next 6 min. It was constant for the
169
next 3 min and then 0:100 (A:B) for the next 17 min (Klejdus et al., 2004). The flow rate was
170
maintained at 0.7 ml/min and the column temperature was kept at 30°C. The analytical
171
wavelength was set at 280 nm and MS/MS analysis was done in both positive and negative
172
mode. Ionisation method: Atmospheric pressure ionization and the mass range was 50-1000 m/z.
173
The results were analyzed using the mass bank database (https://massbank.eu/MassBank) by
174
checking the base peaks and other major fragment ion peaks with the existing mass spectra data
175
of standard vitamins.
176
2.5 Phytochemical extraction and analysis
177
Soxhlet extraction of crude metabolites was performed using dried rhizome powder in
178
solvents with increasing polarity (hexane
179
The extracts were concentrated under low pressure using a rotary evaporator (Buchi R-205,
180
Fisher Scientific, Reinach, Switzerland). Stock solutions (mg/ml) of RE were prepared by
181
dissolving the dried samples in a common solvent, dimethyl sulphoxide (DMSO), whereas the
182
water extract was dissolved in water. Presence of alkaloids, terpenoids, flavonoids, tannins,
183
phlobatanins, saponins, steroids and cardiac glycosides in rhizome was analyzed following the
184
methods described earlier (Mujeeb et al., 2014) with little modification (Supplementary Table 1),
185
and the results were expressed as present (+) and absent (-) for tests.
186
2.6 Total phenol and flavonoids
187
Total phenolic contents in different extracts were quantified using Folin-Ciocalteu (FC)
188
reagent. Briefly, 250 µl sample solution was added to 1 ml diluted (1:9) FC reagent. After 5 min
8
189
incubation, 750 µl 1% Na2CO3 solution was added. Then the sample was incubated at 30℃ for 2
190
h and absorbance was measured at 760 nm (Slinkard & Singleton, 1977). The total phenolic
191
contents of solvent extracts were expressed as equivalents of gallic acid (µg GAE/mg crude
192
extract).
193
Total flavonoid content was quantified by mixing sample solution with 2% AlCl3 in
194
methanol in 1:1 ratio. After 15 min incubation at 30℃, the sample and blank absorbance were
195
read at 415 nm. The absorbance of the blank was subtracted from that of the sample. The total
196
flavonoid content was expressed as equivalents of quercetin (µg QCTE/mg crude extract) (Berk
197
et al., 2011).
198
2.7 Total antioxidant activity (phosphomolybdenum method)
199
RE were mixed with reagent solution (0.6 M sulfuric acid, 28 mM sodium phosphate and
200
4 mM ammonium molybdate) at a 1:10 ratio, and incubated for 90 min at 95°C. The absorbance
201
of the mixture was measured at 695 nm. The total antioxidant capacity of RE was expressed as
202
equivalents of ascorbic acid (µg AE/mg crude extract) (Berk et al., 2011).
203
2.8 Radical scavenging activity
204
2.8.1 DPPH radical scavenging
205
RE (10 µl) were mixed with 250 µl DPPH solutions (0.2 mM in methanol) in a 96 well
206
microtiter plate (WMP). After incubating the samples for 15 min at 30℃ in the dark, the
207
absorbance was read at 517 nm (Blois, 1958). DPPH radical scavenging capability of RE was
208
calculated and expressed as percent inhibition using the following equation: % =
!" #
− !" $ !" #
× 100
209
The IC50 values of crude extracts were compared qualitative with the standards (GA, BHA and
210
BHT). 9
211
2.8.2. ABTS cation radical scavenging
212
ABTS solution was prepared by mixing 2.45 mM potassium persulfate and ABTS (7
213
mM) in water and allowing the mixture to stand for 15-16 h in the dark at 30℃). The ABTS
214
solution was diluted 10 times with methanol before use. RE (10 µl) was added to 200 µl of
215
ABTS solution in a 96 WMP. After incubating the samples for 30 min at 30℃, the absorbance
216
was read at 734 nm (Re et al., 1999). ABTS cation radical scavenging activity of RE was
217
compared with the standards (GA, BHA and BHT) and represented as IC50.
218
2.8.3 Superoxide anion (O2−) radical scavenging
219
To the 175 µl reagent mixture (10 µl 0.1 mg/ml riboflavin, 10 µl 12 mM EDTA
220
ethylenediaminetetraacetic acid, 5 µl 1 mg/ml nitroblue tetrazolium (NBT), 100 µl 50 mM
221
potassium phosphate buffer (KPB, pH 7.8) and 50 µl methionine), 25 µl RE were added into a 96
222
WMP. The reaction mixture was incubated at 30℃ for 10 min using a 20 W fluorescence lamp
223
(DLF Phase-3, Philips, Gurugram, Haryana, India) at 0.5 meter distance. The absorbance of the
224
sample was read at 560 nm and superoxide anion (O2−) radical scavenging activity of RE was
225
expressed as IC50 and compared with the standard (BHA) (Dasgupta & De, 2004; Martinez et al.,
226
2001).
227
2.9 Reducing Power
228
2.9.1 Cupric ion reducing antioxidant capacity (CUPRAC) assay
229
To 150 µl of premixed reaction mixture containing 50 µl 10 mM CuCl2, 50 µl 7.5 mM
230
neocuproine in 95% ethanol and 50 µl 1 M ammonium acetate buffer (pH 7.0) and 25 µl RE
231
were added into a 96 WMP. Similarly, the blank was without CuCl2. The mixture was incubated
232
for 30 min at 30℃, and the absorbance of the sample, BHA as a positive control and blank were
233
read at 450 nm (Apak et al., 2006). The results were expressed as µg BHAE/mg of RE.
10
234
2.9.2 Ferric ion reducing antioxidant power (FRAP) assay
235
RE (10 µl) were added to 240 µl of FRAP reagent containing 20 mM TPTZ ligand in 80
236
mM HCl, 20 mM FeCl3 and 0.3 M acetate buffer (pH 3.6) at a 1:1:10 ratio in a 96 WMP and
237
incubated for 30 min at 37℃. The sample absorbance was read at 593 nm and compared with the
238
standard, which was prepared by taking a different concentration of FeSO4 with 20 mM TPTZ
239
and 0.3 M acetate buffer (pH 3.6) at a 1:1:10 ratio (Oyaizu, 1986). The results were expressed as
240
µg GAE/mg of RE.
241
2.10 Metal chelating activity
242
RE (200 µl) were mixed with 10 µl 2 mM FeCl2 solution into a 96 WMP and incubated
243
for 5 min. Ferrozine (20 µl, 5 mM) was added to initiate the reaction and a similar reaction
244
without ferrozine served as a control. The sample and blank absorbance were read at 562 nm
245
after 10 min incubation at 30°C (Aktumsek et al., 2013). The metal chelating activity was
246
expressed as µg EDTAE/mg of RE.
247
2.11 Biomolecular protection assay
248
2.11.1 DNA protection assay
249
The protective effect of RE against hydroxyl radical-induced metal-assisted DNA
250
degradation was studied using CT DNA. RE (10 µl) were added to 10 µl TE buffer (pH 8) (10
251
mM Tris-HCl and 1 mM EDTA) containing 280 ng of DNA and incubated for 5 min. To the
252
mixture, Fenton’s reagent (8 µl) was added and incubated for 2.5 h at 30℃ (Karas et al., 2013).
253
The integrity of DNA was analyzed using electrophoresis on 1% agarose gel (Agarose
254
Electrophoresis system, Thermo Fisher Scientific, Mumbai, India) followed by ethidium bromide
255
staining (0.5 µg/ml) for 15 min. The DNA degradation pattern was observed with UV
11
256
illumination (UVstar, Biometra GmbH, Gottingen, Germany). The DNA sample without any
257
extract and Fenton's reagent was used as a control.
258
2.11.2 Anti-protein oxidation assay
259
BSA (0.5 mg) was dissolved in phosphate buffer (pH 7.3) and incubated with peroxyl
260
radicals generating AAPH (2,2-azobis (2-methylpropio-namidine) dihydrochloride) (50 mM)
261
with or without different concentrations of RE. After 2 h incubation at 30℃, the integrity of
262
protein samples was analyzed using SDS-PAGE. The proteins bands were visualized by staining
263
the gel with 0.2% Coomassie Brilliant Blue R-250 in methanol:water:glacial acetic acid (5:5:1)
264
overnight and then destaining (same as staining solution but without the Coomassie Brilliant
265
Blue R-250 dye) until clear bands of protein were observed (Mayo et al., 2003).
266
2.11.3 β-carotene linoleic acid model
267
β-carotene (2 ml, 0.5 mg/ml in chloroform) was mixed with 40 mg linoleic acid and 400
268
mg Tween 40. The solution was dried under low pressure in a rotary evaporator at 40°C.
269
Distilled water (50 ml) was added and the flask was vigorously shaken. The reaction was
270
initiated by adding 3.5 ml of the above solution to 500 µl acetone extract (100 and 200 ppm of
271
GA equivalent phenol) in a test tube. After the addition, the samples were measured 470 nm at 0
272
min. The reaction mixtures were incubated in a water bath at 50°C. The samples was measured
273
every 15 min for 120 min. BHA was used as the standard for comparative analysis. A control
274
without RE or standard was also used. Blank samples were prepared by mixing all the samples
275
and reagents except β-carotene. Blank absorbance was subtracted from that of the sample to get
276
the corrected absorbance. The inhibition % was determined using the following equation (Oh &
277
Shahidi, 2018).
278
Antioxidant activity (%) = [1− (S0−St)/(C0−Ct)] × 100
12
279
(S0: Absorbance of the test sample at 0 min, St: Absorbance of the test sample at every 15 min,
280
C0: Absorbance of control at 0 min and Ct: Absorbance of control at every 15 min)
281
2.11.4 Meat model system (TBARS value)
282
The boneless pork meat was purchased from a local market in New Delhi, India. Pork
283
meat (40 g) was pasted in a pre-chilled mortar and pestle, added to 10 ml Millipore water (Elix 3
284
UV Water Purification System (120 V/60 Hz), Millipore, Merck Life Science, Mumbai, India)
285
and the test sample (10 ml acetone extract containing 150 and 300 ppm of GA equivalent phenol)
286
or the positive control (10 ml, 300 ppm BHT). A blank was also prepared without any
287
antioxidants. The samples were mixed and heated at 80°C in a water bath for 40 min with
288
frequent stirring. The mixture was allowed to cool to room temperature and the contents were
289
again pasted. The contents were stored in plastic bags for 7 days at 4°C (Oh & Shahidi, 2018)
290
and analyzed at 532 nm for their oxidative state using the TBARS test (Shahidi & Hong, 1991)
291
on 0, 3, 5, and 7 days.
292
Trichloroacetic acid (10%, w/v, 3 ml) was added to meat samples (1 g) in centrifuge
293
tubes and Vortexed (Remi Cyclo, CM-101 Plus, Mumbai, India) for 2 min. TBA reagent (0.02
294
M, 3 ml) was added and Vortexed again for 30 s. After centrifugation at 3000g for 10 min, the
295
supernatants were filtered through a Whatman No. 3 filter paper. The samples were kept in a
296
water bath at 95°C for 45 min, then cooled to room temperature. The absorbance of pink TBA-
297
MDA adducts was read at 532 nm. TBARS values were calculated using a standard curve,
298
plotted using 1,1,3,3-tetramethoxypropane (a precursor of MDA). TBARS values were
299
calculated for RE and the results were expressed as mg MDA eq/kg of sample.
300
2.12 Enzyme inhibitory assays
301
2.12.1 α-Amylase inhibition
13
302
RE (200 µl) were mixed with 200 µl α-amylase (2 U/ml) prepared in phosphate buffer
303
(pH 6.9) and incubated for 10 min. Further, 200 µl 1% starch (potato) solution was added and the
304
mixture was incubated for 30 min at 30°C. A blank was maintained with all the reagents except
305
RE to check 100% enzyme activity. The reaction was terminated by the addition of 200 µl DNS
306
reagent (12 g sodium potassium tartrate tetrahydrate in 8.0 ml 2 M NaOH and 20 ml 95 mM 3,5-
307
dinitrosalicylic acid solution) and was boiled for 10 min in a water bath at 85–90 °C. The
308
mixture was cooled to room temperature and was diluted with 5 ml distilled water, and the
309
absorbance was read at 540 nm. The α-amylase inhibitory activity of RE was expressed in
310
percentage (Zengin et al., 2014).
311
2.12.2 α-Glucosidase inhibition
312
RE (50 µl) were mixed with 50 µl 1 U/ml α-glucosidase in phosphate buffer (pH 6.8) and
313
incubated for 10 min in a 96 WMP. Then 50 µl 3 mM glutathione (reduced) and 50 µl 10 mM
314
PNPG were added and incubated for 20 min at 35°C. One control was prepared by mixing all
315
reagents except RE to check the maximum released product. Another blank was prepared by RE
316
and adding all other reagents excluding enzyme. The reaction was terminated by adding 50 µl
317
0.2 M sodium carbonate solution. The sample and blank absorbance were read at 400 nm. The
318
inhibitory activity was expressed in percentage (Zengin et al., 2014).
319
2.12.3 Anticholinesterase activity assay
320
Acetylcholinesterase
(AChE)
inhibitory
activity
of
RE
were
carried
out
321
spectrophotometrically (Ellman et al., 1961). To 150 µl 0.1 M PB, 20 µl RE and 20 µl 1 U/ml
322
enzyme solution were added and mixed in a 96 WMP and incubated for 10 min at 30℃ followed
323
by the addition of 15 µl 10 mM DTNB. Then 15 µl 14 mM ATCI was added to initiate the
14
324
reaction and further incubated for 20 min at 30°C. The absorbance of the reaction mixture was
325
read at 412 nm and the AChE inhibitory activity was expressed in percentage.
326
2.13 HR-LCMS analysis of rhizomes extracts
327
HR-LCMS analysis of ethyl acetate, acetone, methanol and water extracts were done
328
using a 6200 series Q-TOF (Q-Exactive Plus Biopharma-High Resolution MS, Thermo Fischer
329
Scientific, Waltham, MA, USA) mass spectrometer coupled to HPLC equipped with a UV–Vis
330
detector. A 0.2 ml/min flow rate was used with injection volume 5 µl; ESI parameters: both
331
negative and positive ion mode; mass range 100–1200 m/z. The solvent system: (A) formic acid
332
(0.1%, v/v) and 10 mM ammonium formate and (B) acetonitrile + 0.1% formic acid. Gradient
333
mobile phase (solvent A:B): (i) 65:35, from 0 to 0.5 min, (ii) 45:55, from 10 min (iii) 5:95, from
334
25 to 33 min (iv) 65:35, at 35–40 min of total run time.
335
2.14 Statistical analysis
336
The results from the experiments were statistically analyzed using the one way analysis
337
of variance (ANOVA) with the Statistical Package for the Social Science v. 16 software (SPSS
338
Inc., Chicago, IL., USA). The significant difference between the means was compared using the
339
highest significant difference (HSD) as obtained using Tukey's test at the p≤0.05 level.
340 341
3. Results and discussion
342
3.1 Proximate and quantitative analysis
343
The results of biomass characterization and nutrient analysis of CI rhizome is shown in
344
Table 2. The rhizome of CI was found to be a source of energy-dense food which had ~423
345
kcal/100 g dw energy, which ranks it ahead of cassava, wheat, rice, corn and sorghum
346
(Montagnac et al., 2009). The presence of a desirable amount of fibers, starch, protein, lipid,
15
347
minerals and vitamins leads to the benefit of consuming the rhizome. Dietary fiber consists of
348
remnants of plant cells resistant to hydrolysis (digestion) by the alimentary enzymes of humans.
349
The rhizome contains 25% crude fiber, which was less than early reports of 33.1% in CI
350
(Okonwu & Ariaga, 2016), close to the white type ginger (21.9%) (Ajayi et al., 2013) and higher
351
than cassava and potato. Canna spp. are known to produce round to oval-shaped starch granules
352
as large as 100 µm (average 30 - 70 µm), which is larger than cassava starch (12 - 15 µm) and is
353
mainly used to prepare noodles or as ingredient in wheat noodles (Wandee et al., 2015). The
354
study rhizome of CI had 28% of starch which is significantly lower in comparison to C. edulis
355
(48.9%) (Piyachomkwan et al., 2002). The lipid content in rhizome was found to be 5.75% on a
356
dw basis (Table 2). This is relatively higher than a report of Okonwu and Ariaga (2016), where
357
they reported 4.35% in CI rhizome, whereas it was significantly higher than cassava roots
358
(0.28%) (Montagnac et al., 2009). The crude protein content was found to be 4.7% on a dw basis
359
which is higher than C. edulis (3.26%) and cassava (1.15%) (Piyachomkwan et al., 2002). On the
360
other hand, the roots of the ginger were reported to have about 12.1% protein (Ajayi et al., 2013).
361
The higher ash content (11.5%) was consistent with the minerals determined separately, which is
362
comparable to different varieties of C. edulis (5.11 – 5.56%) (Piyachomkwan et al., 2002) and
363
ginger rhizome (4.95 – 7.45%). ICP-MS analysis was used to measure the potential of CI
364
rhizome as a source of minerals (Table 2). Among all tested elements, K was found to be most
365
abundant followed by Na, Mg, and P, and a considerable amount of Ca, Mn, Fe, Cu and Zn were
366
also found in the rhizome. RE were also found to have a number of secondary metabolites such
367
as alkaloids, saponin tannin, flavonoids and phenolics, which were only determined qualitatively
368
(Supplementary Table 2).
16
369
Vitamins are the chemical compounds not synthesized in humans and animals. Hence, a
370
plant diet is considered as a primary source of vitamins. Because of their redox chemistry, their
371
role as an enzyme cofactor or their antioxidant activity, they are beneficial for humans. The
372
occurrence of these vitamins in the rhizome of CI has not generally been reported. Ong and
373
Siemonsama (1996) reported the presence of 0.1 mg vitamin B1 and 10 mg vitamin C in 100 g
374
fw (fresh weight) of CI rhizome. The results of this study are shown in Table 2. Vitamin A
375
(retinoic acid) was probably present in both the cis and trans-retinoic acid forms (Supplementary
376
Fig 1). The peak at Rt 14.39 min shows a fragmentation pattern where the peaks of m/z 301.2
377
(100%), 283, 205, 131 matches with that of retinoic acid in LC-MS/MS analysis (Kane et al.,
378
2008). Although HPLC chromatogram supported the presence of vitamin E (Supplementary Fig
379
2), but it was not detected with LC-MS/MS analysis. Several base peaks were observed in the
380
negative mode LC-MS/MS chromatogram (Supplementary Fig 3), which may be attributed to the
381
metabolic products of α-tocopherol such as γ-CMHHC, γ-CDMOHC, carboxyl δ-tocopherol, δ-
382
CDMOHC and α-CMHHC
383
(Supplementary Fig 4) and quantified to be 11.0 µg/g fw. The HR-LCMS of ethyl acetate and
384
acetone extracts showed the presence of pantothenic acid (vitamin B5) (Table 7). The above
385
results showed the possible utilization of CI rhizome as a vitamin source. Also, the results
386
obtained are consistence with reports of 5-35 µg/g wt vitamin A in cassava roots and, 0.107 and
387
0.296 mg/100 g pantothenic acid in cassava and potato, respectively (Bradbury, 1988; Woot-
388
Tsuen, & Jardin, 1968). Whereas among these three, vitamin C was the highest in cassava (20.6
389
mg/100 g) (Montagnac et al., 2009). Vitamin D was not detected in the rhizome of CI, and other
390
vitamins such in the B-complex were not analyzed.
391
3.2 Antioxidant and biomolecule protection activity
(Zhao et al., 2010). Vitamin C was found with HPLC
17
The antioxidant activity measured with crude extracts of CI rhizome is shown in Table 3.
392 393
The IC
394
ABTS and O2.− radical scavenging activity compared to other solvent extracts tested. In the case
395
of DPPH and ABTS assay, the standard, GA showed the lowest IC50. Whereas, the IC50 values of
396
BHA was comparable to acetone extract in the O2.− radical scavenging assay (Table 3).
50
values measured using acetone extract was significantly (p≤0.05) lower for DPPH,
397
In both the CUPRAC and FRAP assays, acetone extracts showed significant (p<0.05)
398
higher reducing power (Table 3). In CUPRAC assay, the reducing power of acetone extracts
399
were not significantly different from methanol extracts but higher than other extracts. Similar
400
activities were shown by solvent extracts for the FRAP assay. Acetone extracts showed
401
significantly (p≤0.05) higher reducing power in comparison to other extracts (Table 3).
402
The metal ion chelating power of different extracts against ferrous ion are shown in Table
403
3 as an equivalent of EDTA/mg of extract. Methanol extracts shown the highest chelating
404
activity followed by acetone and ethyl acetate. Results in Table 3 show that among all solvent
405
extracts, acetone extracts have the highest total antioxidant capacity followed by methanol and
406
ethyl acetate extracts. Water extracts showed the least activity.
407
The etiology of numerous human and animal diseases is linked to the free radicals
408
generated with stress causing damage to cellular constituents. Even though all living organisms
409
have multiple defense mechanisms to tackle the adverse effect of reactive oxygen species (ROS)
410
generated in the body, but the reduction/failure of these mechanisms leads to diseases conditions.
411
To prevent this condition, it is recommended to consume a diet rich in antioxidants (Prior & Cao,
412
2000). Usually, plants have an array of antioxidant systems because they have to cope with a
413
wide range of biotic and abiotic stresses throughout their life. The antioxidant capacity of plants
414
is related to their total phenolic or total flavonoid contents (Kähkönen et al., 2001; Miliauskas et
18
415
al., 2004; Singleton et al., 1999). Prior and Cao (2000) established a linear correlation between
416
the total phenolic content of wines and their antioxidant properties. Also, the disease protection
417
properties of wines were linked to their free radical scavenging and the transition metal chelating
418
capabilities of flavonoids fractions. Consistent with the previous reports, acetone extracts which
419
showed the highest antioxidant activity, also showed the highest total phenolics and flavonoids,
420
in comparison with other extracts (Table 3).
421
Mishra et al. (2012) found that diethyl ether, ethyl acetate and acetone extracts of C.
422
edulis alone or in combination showed the highest DPPH radical scavenging activity. Further
423
higher concentrations of flavonol and total proanthocyanidins were studied in these fractions.
424
Extracts of CI aerial plant parts had antioxidant properties (Joshi et al., 2009) and
425
hepatoprotective effects (Kaldhone, 2009). In similar studies, the authors correlated this
426
observation with the antioxidant properties of the extracts. Further, these correlations were
427
extended to macromolecular protection (DNA, protein and lipid) from these plant extracts. LC-
428
MS analysis of CI RE obtained a wide range of metabolites which were previously reported to be
429
antioxidant (Table 7).
430
The capability of RE to protect DNA or protein is shown in Fig 1(a). Acetone extracts
431
were found effective in protecting DNA damage at lower concentrations. Methanol was next
432
followed by ethyl acetate and water (little protection). Acetone extracts which showed higher
433
antioxidant and metal chelating activity, also showed higher DNA protection ability and are
434
correlated with metal ion chelation and toxic radical scavenging activity. Similar results were
435
seen using the protein protection assay where acetone extracts protected BSA from damage
436
caused by the AAPH radical at lower concentrations (Fig 1(b)) followed by methanol. Extracts
437
of ethyl acetate and water showed partial protection with the highest concentration used.
19
438
The β-carotene bleaching assay is based on the degradation of β-carotene entrapped in
439
linoleic acid micelles due to the effect of hydroperoxides and free radicals generated with the
440
heat-induced oxidation of linoleic acid. Plant extracts with potential antioxidants have been
441
reported to prevent the damage of β-carotene by scavenging the free radicals (Oh & Shahidi,
442
2018). Acetone extracts of CI rhizome were found to protect the β-carotene from the damaging
443
effect of hydroperoxides and free radicals up to 82% in comparison to control (11%) and BHA
444
(88%) over 120 min incubation period. Also, the β-carotene protection was correlated with the
445
phenolic content of RE (Table 4).
446
Ground pork meat without antioxidant amendment had the highest MDA. Whereas
447
acetone extracts of CI rhizome and BHT decreased the level of MDA significantly (Table 5).
448
3.3 Enzyme inhibitory activity
449
Diabetes Mellitus (DM) is a metabolic disorder of glucose metabolism of multiple
450
etiologies. The condition is characterized by chronic hyperglycemia resulting from decreased
451
insulin secretion or altered response of cells to insulin action, or both. One of the effective
452
measures adopted is to manage the excess postprandial rise of blood glucose level. It is mainly
453
achieved through the intake of proper diet or by using α-amylase and α-glucosidase inhibitors
454
(Tadera et al., 2006). However, long-term use of drugs (Acarbose, Miglitol, Metformin, Sulfonyl
455
Urea, etc.) is often reported to adversely affect human health (Fujisawa et al., 2005; Kelble,
456
2005; Lebovitz, 1997). Alternatively, several natural molecules of plant origin which are
457
consumed by humans reversibly inhibit the activity of α-amylase and α-glucosidase, thereby
458
reducing the rate of free sugar release. Polyphenolic fractions from plants such as catechin
459
gallates, quercetin, isoquercetin and rutin have been studied for their α-amylase and α-
460
glucosidase inhibiting properties (Li et al., 2009). A significant α-glucosidase inhibitory activity
20
461
was shown by CI water extracts and acetone extracts (Table 6). The enzyme inhibition plots for
462
acetone and water extracts (Fig. 2 (a) and (b)) indicated that the rhizome is rich in secondary
463
metabolite(s) as natural α-glucosidase inhibitors. It was also observed that acetone and ethyl
464
acetate extracts inhibit the enzyme activity of α-amylase up to 21 and 18%, respectively, at 166
465
µg/ml and beyond, which was also about the concentration where the solubility of the extracts
466
was decreased. Other extracts were shown to have no inhibitory activity against the enzyme even
467
at the highest concentration used. This suggested the presence of bioactive compounds that can
468
control the activity of enzymes related to the release of free sugar molecule from oligomer or
469
polymers. On the other hand, Purintrapiban (2006) observed that the aqueous extracts of CI roots
470
which had higher total phenolics compounds (catechin) induced 2-deoxy-(3H) glucose uptake in
471
cultured L8 muscle cells and was correlated with increased glucose transporter isoforms 1 and 4
472
on the cell surface.
473
Tripathi (2004) reported the inhibition of AChE activity by extracts of Punica granatum
474
bark or CI root, along with other plant-derived molluscicides in the nervous tissue of Lymnaea
475
acuminate. However, with in vitro conditions, a significant reduction in AChE was observed. In
476
this study, none of the RE showed AChE inhibition.
477
3.4 HR-LC MS/MS analysis of RE
478
Although the crude extracts of different plant parts of Canna were reported to have some
479
biological activity, any effort to correlate the activity with the phenolic metabolites has been
480
minimal. Previously, Sook Yun (2004) reported the presence of 4 phenylpropanoids such as
481
caffeic acid, rosmarinic acid, caffeoyl-1-4-hydroxyphenyllactic acid and salvianolic acid B from
482
the rhizome of Canna edulis. HR-LC MS/MS analysis of different RE of CI showed a wide range
483
of metabolites with reported diverse biological activities (Table 7). The antioxidant,
21
484
macromolecular protection and enzyme inhibitory activities shown by these extracts may be
485
because of the synergistic effect of these metabolites. The presence of relatively higher
486
concentration of metabolites such as usnic acid, ellagic acid, p-coumaric acid, rosmarinic acid,
487
psoromic acid, phenylacetic acid and swietenine like compound in the rhizome indicated its
488
potential use as an antioxidant, anti-inflammatory or anticancerous and to cure/manage diseases
489
related to heart, dementia, etc. The involvement of some unidentified metabolites in the above-
490
studied activities cannot be ignored. In most cases, swietenine was reported in the seeds of
491
Swietenia sp. which were studied for their antidiabetic properties (Dewanjee et al., 2009). The
492
presence of swietenine or swietenine like compounds in canna rhizome has a potential benefit as
493
DM medication. Many naturally occurring phenolic compounds and flavonoids such as
494
quercetin, taxifolin, luteolin, curcumin, p-coumaric acid, caffeic acid, resveratrol, and many
495
other polyphenolics have been studied for their significant α-glucosidase inhibitory activities
496
(Jiang et al., 2017; Proença et al., 2017; Rasouli et al., 2017). Thus the phenolic compounds
497
(Table 7) in acetone extracts of CI such as psoromic acid, usnic acid and rosmarinic acid could
498
be potential inhibitors of α-glucosidase. Further study on individual compounds may show the
499
enzyme-ligand interaction and the importance of CI rhizome as a valuable source of drug
500
ingredients for DM.
501 502
4. Conclusions
503
The rhizomes of CI are good source of nutrients, i.e., starch, dietary fibers, vitamins and
504
minerals, and are comparable to C. edulis and different varieties of ginger. Antioxidant activity
505
of rhizome extracts indicated their potential use in preventing oxidative damage in food and
22
506
biological systems. Further, α-glucosidase inhibitory properties of RE and other metabolites that
507
may have health benefits suggested its potential use as a functional food for diabetic patients.
508 509
Acknowledgments
510
We are grateful to the Council of Scientific and Industrial Research (CSIR), New Delhi
511
for financial support and our premier research institute, the Indian Institute of Technology Delhi
512
(IITD) for providing research space and necessary facilities. We are thankful to the Nanoscale
513
Research Facility (NRF), IITD, Sophisticated Analytical Instrument Facility (SAIF), IIT
514
Bombay and SAIF, the Central Drug Research Institute (CDRI) Lucknow for providing HPLC
515
and LC-MS/MS facilities.
516 517
Conflict of interest
518
The authors declare that there is no conflict of interest
519 520
Human and animal rights
521
This article does not report any studies with human or animal subjects.
522 523
References
524
Ajayi, O. B., Akomolafe, S. F., & Akinyemi, F. T. (2013). Food value of two varieties of ginger
525
(Zingiber officinale) commonly consumed in Nigeria. ISRN Nutrition, 2013, 1–5.
526
https://doi.org/10.5402/2013/359727.
527
Aktumsek, A., Zengin, G., Guler, G. O., Cakmak, Y. S., & Duran, A. (2013). Antioxidant
528
potentials and anticholinesterase activities of methanolic and aqueous extracts of three
23
529
endemic Centaurea L. species. Food and Chemical Toxicology, 55, 290–296.
530
https://doi.org/10.1016/j.fct.2013.01.018.
531
Akula, R., & Ravishankar, G. A. (2011). Influence of abiotic stress signals on secondary
532
metabolites
533
https://doi.org/10.4161/psb.6.11.17613.
534 535
536 537
in
plants.
Plant
Signaling
&
Behavior,
6(11),
1720–1731.
AOAC. (1990). Official Methods of Analysis (15th ed.). Washington, District of Columbia, USA: Association of Official Analytical Chemists. AOAC. (2000). Official Methods of Analysis (17th ed.). Arlington, Virginia, USA: Association of Official Analytical Chemists.
538
Apak, R., Güçlü, K., Özyürek, M., Karademir, S. E., & Erçağ, E. (2006). The cupric ion reducing
539
antioxidant capacity and polyphenolic content of some herbal teas. International Journal
540
of
541
https://doi.org/10.1080/09637480600798132.
542 543
Food
Sciences
and
Nutrition,
57(5–6),
292–304.
Bayer, R., & Mannhold, R. (1987). Fendiline: A review of its basic pharmacological and clinical properties. Pharmatherapeutica, 5(2), 103–136.
544
Bean, W. B., & Hodges, R. E. (1954). Pantothenic acid deficiency induced in human subjects.
545
Proceedings of the Society for Experimental Biology and Medicine, 86(4), 693–698.
546
https://doi.org/10.3181/00379727-86-21204.
547
Behera, B. C., Mahadik, N., & Morey, M. (2012). Antioxidative and cardiovascular-protective
548
activities of metabolite usnic acid and psoromic acid produced by lichen species Usnea
24
549
complanata under submerged fermentation. Pharmaceutical Biology, 50(8), 968–979.
550
https://doi.org/10.3109/13880209.2012.654396.
551
Berk, M., Dean, O., Drexhage, H., McNeil, J. J., Moylan, S., O’Neil, A., Davey, C. G., Sanna,
552
L., & Maes, M. (2013). Aspirin: A review of its neurobiological properties and therapeutic
553
potential for mental illness. BMC Medicine, 11(1), 74. https://doi.org/10.1186/1741-7015-
554
11-74.
555
Berk, S., Tepe, B., Arslan, S., & Sarikurkcu, C. (2011). Screening of the antioxidant,
556
antimicrobial and DNA damage protection potentials of the aqueous extract of Asplenium
557
ceterach DC. African Journal of Biotechnology, 10(44), 8902-8908.
558 559
Blois, M. S. (1958). Antioxidant determinations by the use of a stable free radical. Nature, 181(4617), 1199–1200. https://doi.org/10.1038/1811199a0.
560
Bradbury, J. H., & Holloway, W. D. (1988). Chemistry of tropical root crops: Significance for
561
nutrition and agriculture in the Pacific. Chemistry of Tropical Root Crops: Significance for
562
Nutrition and Agriculture in the Pacific. Canberra, Australia: Australian Center for
563
International
564
https://www.cabdirect.org/cabdirect/abstract/19890724438.
Agricultural
Research.
565
Cai, Y., Luo, Q., Sun, M., & Corke, H. (2004). Antioxidant activity and phenolic compounds of
566
112 traditional Chinese medicinal plants associated with anticancer. Life Sciences, 74(17),
567
2157–2184. https://doi.org/10.1016/j.lfs.2003.09.047.
568
Campos, F. M., Ribeiro, S. M. R., Della Lucia, C. M., Pinheiro-Sant’Ana, H. M., & Stringheta,
569
P. C. (2009). Optimization of methodology to analyze ascorbic and dehydroascorbic acid
25
570
in
vegetables.
Química
571
40422009000100017.
Nova,
32(1),
87–91.
https://doi.org/10.1590/S0100-
572
Cato, A., Sutton, L., Kaplan, A. S., & Manning, G. N. (2001). Activity of a triamcinolone
573
acetonide/laurocapram formulation: Double-blind comparisons with triamcinolone
574
acetonide, placebo vehicle, and mid-strength (class IV) to potent (class II) corticosteroids.
575
Current
576
393X(01)80029-1.
Therapeutic
Research,
62(3),
180–186.
https://doi.org/10.1016/S0011-
577
Cevc, G., & Blume, G. (2003). Biological activity and characteristics of triamcinolone-acetonide
578
formulated with the self-regulating drug carriers, Transfersomes®. Biochimica et
579
Biophysica
580
https://doi.org/10.1016/S0005-2736(03)00172-X.
Acta
(BBA)
-
Biomembranes,
1614(2),
156–164.
581
Çırak, C., Radušienė, J., & Çamas, N. (2008). Pseudohypericin and hyperforin in two Turkish
582
Hypericum species: Variation among plant parts and phenological stages. Biochemical
583
Systematics and Ecology, 36(5), 377–382. https://doi.org/10.1016/j.bse.2007.12.009.
584
Cosmosil
technical
note:
Vitamin
analysis
by
HPLC.
Internet:
585
https://www.nacalai.co.jp/global/cosmosil/pdf/Vitamine_Analysis.pdf. Nacalai Tesque,
586
Inc., Kyoto, Japan. (accessed January 2019).
587
Daniels, C. W., Rautenbach, F., Mabusela, W. T., Valentine, A. J., & Marnewick, J. L. (2011).
588
Comparative antioxidant-capacity and -content of leaves, bulbs, roots, flowers and fruit of
589
Gethyllis multifolia L. Bolus and G. villosa Thunb. species. South African Journal of
590
Botany, 77(3), 711–717. https://doi.org/10.1016/j.sajb.2011.03.005.
26
591 592
Dasgupta, N., & De, B. (2004). Antioxidant activity of Piper betle L. leaf extract in vitro. Food Chemistry, 88(2), 219–224. https://doi.org/10.1016/j.foodchem.2004.01.036.
593
Deraeve, C., Guo, Z., Bon, R. S., Blankenfeldt, W., DiLucrezia, R., Wolf, A., Menninger, S.,
594
Stigter, E.A., Wetzel, S., Choidas, A. & Alexandrov, K. (2012). Psoromic acid is a
595
selective and covalent Rab-Prenylation inhibitor targeting autoinhibited RabGGTase.
596
Journal
597
https://doi.org/10.1021/ja211305j.
of
the
American
Chemical
Society,
134(17),
7384–7391.
598
Dewanjee, S., Maiti, A., Das, A. K., Mandal, S. C., & Dey, S. P. (2009). Swietenine: A potential
599
oral hypoglycemic from Swietenia macrophylla seed. Fitoterapia, 80(4), 249–251.
600
https://doi.org/10.1016/j.fitote.2009.02.004.
601 602
Duke, J. A., & Ayensu, E. S. (1985). Medicinal Plants of China. 2. Algonac, MI: Reference Publications.
603
Ellman, G. L., Courtney, K. D., Andres, V., & Featherstone, R. M. (1961). A new and rapid
604
colorimetric determination of acetylcholinesterase activity. Biochemical Pharmacology,
605
7(2), 88–95. https://doi.org/10.1016/0006-2952(61)90145-9.
606
Fischer, L. M., da Costa, K.-A., Kwock, L., Galanko, J., & Zeisel, S. H. (2010). Dietary choline
607
requirements of women: Effects of estrogen and genetic variation. The American Journal
608
of Clinical Nutrition, 92(5), 1113–1119. https://doi.org/10.3945/ajcn.2010.30064.
609
Food and Drug Administration (2004). Apomorphine hydrochloride injection. Internet:
610
https://www.accessdata.fda.gov/drugsatfda_docs/label/2004/21264_apokyn_lbl.pdf
611
(accessed December 2018).
27
612
Fugh-Berman, A., & Ernst, E. (2001). Herb-drug interactions: Review and assessment of report
613
reliability: Short report. British Journal of Clinical Pharmacology, 52(5), 587–595.
614
https://doi.org/10.1046/j.0306-5251.2001.01469.x.
615
Fujisawa, T., Ikegami, H., Inoue, K., Kawabata, Y., & Ogihara, T. (2005). Effect of two α-
616
glucosidase inhibitors, voglibose and acarbose, on postprandial hyperglycemia correlates
617
with
618
https://doi.org/10.1016/j.metabol.2004.10.004.
619 620
subjective
abdominal
symptoms.
Metabolism,
54(3),
387–390.
Gade, D. W. (1966). Achira, the edible canna, its cultivation and use in the Peruvian Andes. Economic Botany, 20(4), 407–415. https://doi.org/10.1007/BF02904063.
621
Gimeno, E., Castellote, A. L., Lamuela-Raventós, R. M., de la Torre, M. C., & López-Sabater,
622
M. C. (2000). Rapid determination of vitamin E in vegetable oils by reversed-phase high-
623
performance liquid chromatography. Journal of Chromatography A, 881(1–2), 251–254.
624
https://doi.org/10.1016/S0021-9673(00)00219-3.
625
Gonçalves, C., Rodriguez-Jasso, R. M., Gomes, N., Teixeira, J. A., & Belo, I. (2010). Adaptation
626
of dinitrosalicylic acid method to microtiter plates. Analytical Methods, 2(12), 2046.
627
http://doi:10.1039/c0ay00525h.
628 629
630 631
Harborne, A. J. (1998). Phytochemical Methods: A Guide to Modern Techniques of Plant Analysis. Heidelberg, Germany: Springer Netherlands. ISBN: 978-0-412-57270-8. Hollenbeck, C. B. (2012). An introduction to the nutrition and metabolism of choline. Central Nervous System Agents in Medicinal Chemistry, 12(2), 100–113.
28
632
Huang, W. Y., Cai, Y. Z., & Zhang, Y. (2009). Natural phenolic compounds from medicinal
633
herbs and dietary plants: Potential use for cancer prevention. Nutrition and Cancer, 62(1),
634
1–20. https://doi.org/10.1080/01635580903191585.
635 636
Ingólfsdóttir,
K.
(2002).
Usnic
acid.
Phytochemistry,
61(7),
729–736.
https://doi.org/10.1016/S0031-9422(02)00383-7.
637
Jeong, W. H., Harada, K., Yamada, T., Abe, J., & Kitamura, K. (2010). Establishment of new
638
method for analysis of starch contents and varietal differences in soybean seeds. Breeding
639
Science, 60(2), 160–163. https://doi.org/10.1270/jsbbs.60.160R.
640
Jiang, P., Xiong, J., Wang, F., Grace, M. H., Lila, M. A., & Xu, R. (2017). α-Amylase and α-
641
glucosidase inhibitory activities of phenolic extracts from Eucalyptus grandis × E.
642
urophylla bark. Journal of Chemistry, 2017 https://doi.org/10.1155/2017/8516964.
643
Joshi, Y. M., Kadam, V. J., & Kaldhone, P. R. (2009). In vitro antioxidant activity of methanolic
644
extract of aerial parts of Canna indica L. Journal of Pharmacy Research, 2(11), 1712–
645
1715.
646
Kähkönen, M. P., Hopia, A. I., & Heinonen, M. (2001). Berry phenolics and their antioxidant
647
activity.
648
https://doi.org/10.1021/jf010152t.
649 650
Journal
of
Agricultural
and
Food
Chemistry,
49(8),
4076–4082.
Kaldhone, P. R. (2009). Investigation of hepatoprotective activity of aerial parts of Canna indica L. on carbon tetrachloride treated rats. Journal of Pharmacy Research, (12), 4.
29
651
Kane, M. A., Folias, A. E., Wang, C., & Napoli, J. L. (2008). Quantitative profiling of
652
endogenous retinoic acid in vivo and in vitro by tandem mass spectrometry. Analytical
653
Chemistry, 80(5), 1702–1708. https://doi.org/10.1021/ac702030f.
654
Karas, V. O., Westerlaken, I., & Meyer, A. S. (2013). Application of an in vitro DNA protection
655
assay to visualize stress mediation properties of the Dps protein. Journal of Visualized
656
Experiments, (75). https://doi.org/10.3791/50390.
657 658
Kelble, A. (2005). Spices and type 2 diabetes. Nutrition & Food Science, 35(2), 81–87. https://doi.org/10.1108/00346650510585868.
659
Klejdus, B., Petrlová, J., Potěšil, D., Adam, V., Mikelová, R., Vacek, J., Kizek, R., & Kubáň, V.
660
(2004). Simultaneous determination of water- and fat-soluble vitamins in pharmaceutical
661
preparations by high-performance liquid chromatography coupled with diode array
662
detection.
663
https://doi.org/10.1016/j.aca.2004.02.027.
Analytica
Chimica
Acta,
520(1–2),
57–67.
664
Landete, J. M. (2011). Ellagitannins, ellagic acid and their derived metabolites: A review about
665
source, metabolism, functions and health. Food Research International, 44(5), 1150–
666
1160. https://doi.org/10.1016/j.foodres.2011.04.027.
667 668
Lebovitz, H. E. (1997). Alpha-glucosidase inhibitors. Endocrinology and Metabolism Clinics of North America, 26(3), 539–551. https://doi.org/10.1016/S0889-8529(05)70266-8.
669
Lee, J. E., Giovannucci, E., Fuchs, C. S., Willett, W. C., Zeisel, S. H., & Cho, E. (2010). Choline
670
and betaine intake and the risk of colorectal cancer in men. Cancer Epidemiology
671
Biomarkers & Prevention, 19(3), 884–887. https://doi.org/10.1158/1055-9965.EPI-09-
672
1295. 30
673 674
Lee, J., & Scagel, C. F. (2009). Chicoric acid found in basil (Ocimum basilicum L.) leaves. Food Chemistry, 115(2), 650–656. https://doi.org/10.1016/j.foodchem.2008.12.075.
675
Li, Y. Q., Zhou, F. C., Gao, F., Bian, J. S., & Shan, F. (2009). Comparative evaluation of
676
quercetin, isoquercetin and rutin as inhibitors of α-glucosidase. Journal of Agricultural
677
and Food Chemistry, 57(24), 11463-11468.
678
Lipmann, F., Kaplan, N. O., Novelli, G. D., Tuttle, L. C., & Guirard, B. M. (1947). Coenzyme
679
for acetylation, a pantothenic acid derivative. Journal of Biological Chemistry, 167, 869–
680
870.
681
Lou, Z., Wang, H., Rao, S., Sun, J., Ma, C., & Li, J. (2012). p-Coumaric acid kills bacteria
682
through
dual
damage
mechanisms.
683
https://doi.org/10.1016/j.foodcont.2011.11.022.
Food
Control,
25(2),
550–554.
684
Martinez, C. A., Loureiro, M. E., Oliva, M. A., & Maestri, M. (2001). Differential responses of
685
superoxide dismutase in freezing resistant Solanum curtilobum and freezing sensitive
686
Solanum tuberosum subjected to oxidative and water stress. Plant Science, 160(3), 505–
687
515. https://doi.org/10.1016/S0168-9452(00)00418-0.
688
Mayo, J. C., Tan, D. X., Sainz, R. M., Natarajan, M., Lopez-Burillo, S., & Reiter, R. J. (2003).
689
Protection against oxidative protein damage induced by metal-catalyzed reaction or
690
alkylperoxyl radicals: comparative effects of melatonin and other antioxidants. Biochimica
691
et
692
https://doi.org/10.1016/S0304-4165(02)00527-5.
Biophysica
Acta
(BBA)
-
General
Subjects,
1620(1–3),
139–150.
31
693
McDonald, M., & Santucci, R. A. (2004). Successful management of stuttering priapism using
694
home self-injections of the alpha-agonist metaraminol. International Brazilian Journal of
695
Urology, 30(2), 121-122.
696
Meruelo, D., Lavie, G., & Lavie, D. (1988). Therapeutic agents with dramatic antiretroviral
697
activity and little toxicity at effective doses: Aromatic polycyclic diones hypericin and
698
pseudohypericin. Proceedings of the National Academy of Sciences, 85(14), 5230–5234.
699
https://doi.org/10.1073/pnas.85.14.5230.
700
Metaraminol. Internet: https://www.drugbank.ca/drugs/DB00610 (accessed January 2019).
701
Miliauskas, G., Venskutonis, P. R., & van Beek, T. A. (2004). Screening of radical scavenging
702
activity of some medicinal and aromatic plant extracts. Food Chemistry, 85(2), 231–237.
703
https://doi.org/10.1016/j.foodchem.2003.05.007.
704 705
Miller, G. L. (1959). Use of dinitrosalicylic acid reagent for determination of reducing sugar. Analytical Chemistry, 31(3), 426-428. http://doi:10.1021/ac60147a030.
706
Mishra, T., Das, A. P., & Sen, A. (2012). Phytochemical screening and in-vitro antioxidant
707
profiling of solvent fractions of Canna edulis Ker Gawler. Free Radicals and
708
Antioxidants, 2(1), 13–20. https://doi.org/10.5530/ax.2012.2.5.
709
Mishra, T., Goyal, A. K., Middha, S. K., & Sen, A. (2011). Antioxidative properties of Canna
710
edulis Ker-Gawl. Indian Journal of Natural Product Research, 2, 315‑321.
711
https://nopr.niscair.res.in/handle/123456789/12735.
712
Montagnac, J. A., Davis, C. R., & Tanumihardjo, S. A. (2009). Nutritional value of cassava for
713
use as a staple food and recent advances for improvement. Comprehensive Reviews in
32
714
Food Science and
715
4337.2009.00077.x.
716 717
Food Safety, 8(3), 181–194. https://doi.org/10.1111/j.1541-
Moudi, M., Go, R., Yien, C.Y.S., & Nazre, M., (2013). Vinca alkaloids. International Journal of Preventive Medicine, 4(11), p.1231.
718
Mujeeb, F., Bajpai, P., & Pathak, N. (2014). Phytochemical evaluation, antimicrobial activity,
719
and determination of bioactive components from leaves of Aegle marmelos. BioMed
720
Research International, 2014. https://doi.org/10.1155/2014/497606.
721
Nirmal, S. A., Shelke, S. M., Gagare, P. B., Jadhav, P. R., & Dethe, P. M. (2007).
722
Antinociceptive and anthelmintic activity of Canna indica. Natural Product Research,
723
21(12), 1042–1047. https://doi.org/10.1080/14786410701526016.
724
Odugbemi, T., Akinsulire, O., Aibinu, I., & Fabeku, P. (2008). Medicinal plants useful for
725
malaria therapy in Okeigbo, Ondo State, Southwest Nigeria. African Journal of
726
Traditional,
727
https://doi.org/10.4314/ajtcam.v4i2.31207.
728
Complementary
and
Medicines,
4(2).
Oh, W. Y., & Shahidi, F. (2018). Antioxidant activity of resveratrol ester derivatives in food and
729
biological
730
https://doi.org/10.1016/j.foodchem.2018.03.085.
731
Alternative
model
systems.
Food
Chemistry,
261,
267–273.
Okonwu, K., & Ariaga, C. (2016). Nutritional evaluation of various parts of Canna indica L.
732
Annual
Research
&
Review
733
https://doi.org/10.9734/ARRB/2016/31029.
in
Biology,
11(4),
1–5.
33
734
Ong, H. C., & Siemonsma, J. S. (1996). Canna indica L. In Plant Resources of South-East Asia
735
9. Plants Yielding Non-Seed Carbohydrates. Bogor, Indonesia: Prosea Foundation. p 63-
736
66.
737 738
Oyaizu, M. (1986). Antioxidative activities of browning reaction prepared from glucosamine. Japanese Journal of Nutrition, 44, 307–315.
739
Pei, K., Ou, J., Huang, J., & Ou, S. (2016). p-Coumaric acid and its conjugates: Dietary sources,
740
pharmacokinetic properties and biological activities. Journal of the Science of Food and
741
Agriculture, 96(9), 2952–2962. https://doi.org/10.1002/jsfa.7578.
742 743
744
Petersen,
M.
(2003).
Rosmarinic
acid.
Phytochemistry,
62(2),
121–125.
https://doi.org/10.1016/S0031-9422(02)00513-7. Pierpoint, W. S. (1994). Salicylic acid and its derivatives in plants: medicines, metabolites and
745
messenger
molecules.
In
Advances
in
746
https://doi.org/10.1016/S0065-2296(08)60217-7.
Botanical
Research,
20,
163–235.
747
Piyachomkwan, K., Chotineeranat, S., Kijkhunasatian, C., Tonwitowat, R., Prammanee, S.,
748
Oates, C. G., & Sriroth, K. (2002). Edible canna (Canna edulis) as a complementary
749
starch source to cassava for the starch industry. Industrial Crops and Products, 16(1), 11–
750
21. https://doi.org/10.1016/S0926-6690(02)00003-1.
751 752
Prior, R. L., & Cao, G. (2000). Antioxidant phytochemicals in fruits and vegetables: Diet and health implications. HortScience, 35(4), 588-592.
753
Proença, C., Freitas, M., Ribeiro, D., Oliveira, E. F. T., Sousa, J. L. C., Tomé, S. M., Ramos, M.
754
J., Silva, A. M. S., & Fernandes, E. (2017). α-Glucosidase inhibition by flavonoids: An in
34
755
vitro and in silico structure-activity relationship study. Journal of Enzyme Inhibition and
756
Medicinal Chemistry, 32(1), 1216–1228. https://doi.org/10.1080/14756366.2017.1368503.
757
Purintrapiban, J., Suttajit, M., & Forsberg, N. E. (2006). Differential activation of glucose
758
transport in cultured muscle cells by polyphenolic compounds from Canna indica L. root.
759
Biological
760
https://doi.org/10.1248/bpb.29.1995.
&
Pharmaceutical
Bulletin,
29(10),
1995–1998.
761
Rasouli, H., Hosseini-Ghazvini, S. M. B., Adibi, H., & Khodarahmi, R. (2017). Differential α-
762
amylase/α-glucosidase inhibitory activities of plant-derived phenolic compounds: A
763
virtual screening perspective for the treatment of obesity and diabetes. Food & Function,
764
8(5), 1942–1954. https://doi.org/10.1039/C7FO00220C.
765
Re, R., Pellegrini, N., Proteggente, A., Pannala, A., Yang, M., & Rice-Evans, C. (1999).
766
Antioxidant activity applying an improved ABTS radical cation decolorization assay. Free
767
Radical Biology and Medicine, 26(9–10), 1231–1237. https://doi.org/10.1016/S0891-
768
5849(98)00315-3.
769
Schempp, C. M., Simon-Haarhaus, B., & Simon, J. C. (2002). Phototoxic and apoptosis-inducing
770
capacity of pseudohypericin. Planta Medica, 68(2), 171–173. https://doi.org/10.1055/s-
771
2002-20264.
772
Shahidi, F., & Hong, C. (1991). Evaluation of malonaldehyde as a marker of oxidative rancidity
773
in
meat
products.
Journal
of
Food
774
https://doi.org/10.1111/j.1745-4514.1991.tb00147.x.
Biochemistry,
15(2),
97–105.
775
Shen, W., Hao, J., Feng, Z., Tian, C., Chen, W., Packer, L., Liu, J. (2011). Lipoamide or lipoic
776
acid stimulates mitochondrial biogenesis in 3T3-L1 adipocytes via the endothelial NO 35
777
synthase-cGMP-protein kinase G signalling pathway: Lipoamide, lipoic acid and
778
mitochondrial biogenesis. British Journal of Pharmacology, 162(5), 1213–1224.
779
https://doi.org/10.1111/j.1476-5381.2010.01134.x.
780
Singleton, V. L., Orthofer, R., & Lamuela-Raventós, R. M. (1999). [14] Analysis of total phenols
781
and other oxidation substrates and antioxidants by means of Folin-Ciocalteu reagent.
782
Methods in Enzymology, 299, 152–178. https://doi.org/10.1016/S0076-6879(99)99017-1.
783 784
785 786
787
Slinkard, K., & Singleton, V. L. (1977). Total phenol analysis: Automation and comparison with manual methods. American Journal of Enology and Viticulture, 28(1), 49–55. Sneader W (1996). Drug Prototypes and their Exploitation (p 414-415). Chichester, England: Wiley. Sook Yun, Y., Satake, M., Katsuki, S., & Kunugi, A. (2004). Phenylpropanoid derivatives from
788
edible
canna,
Canna
edulis.
Phytochemistry,
789
https://doi.org/10.1016/j.phytochem.2004.06.003.
65(14),
2167–2171.
790
Sudha, G., & Ravishankar, G. A. (2002). Involvement and interaction of various signaling
791
compounds on the plant metabolic events during defense response, resistance to stress
792
factors, formation of secondary metabolites and their molecular aspects. Plant Cell, Tissue
793
and Organ Culture, 71(3), 181–212. https://doi.org/10.1023/A:1020336626361.
794 795
Suhaj, M., (2006). Spice antioxidants isolation and their antiradical activity: A review. Journal of Food Composition and Analysis. 19, 531–537. https://doi.org/10.1016/j.jfca.2004.11.005.
36
796
Tadera, K., Minami, Y., Takamatsu, K., & Matsuoka, T. (2006). Inhibition of α-glucosidase and
797
α-amylase by flavonoids. Journal of Nutritional Science and Vitaminology, 52(2), 149–
798
153. https://doi.org/10.3177/jnsv.52.149.
799
Tang, Y., Wang, M., Le, X., Meng, J., Huang, L., Yu, P., Chen, J., & Wu, P. (2011). Antioxidant
800
and cardioprotective effects of Danshensu (3-(3, 4-dihydroxyphenyl)-2-hydroxy-
801
propanoic acid from Salvia miltiorrhiza) on isoproterenol-induced myocardial
802
hypertrophy
803
https://doi.org/10.1016/j.phymed.2011.05.007.
in
rats.
Phytomedicine,
18(12),
1024–1030.
804
Tonwitowat, R. (1994). Yield and some agronomic characteristics of edible canna (Canna edulis
805
Ker) as affected by varieties, planting dates, fertilizer rates and plant densities. Ph.D.
806
Thesis. Kasetsart University, Bangkok, Thailand.
807
Tucker, C. S., & Debusk, T. A. (1981). Seasonal growth of Eichhornia crassipes (Mart.) solms:
808
Relationship to protein, fiber, and available carbohydrate content. Aquatic Botany, 11,
809
137–141. https://doi.org/10.1016/0304-3770(81)90055-3.
810
Tripathi, S. M., Singh, V. K., Singh, S., & Singh, D. K. (2004). Enzyme inhibition by the
811
molluscicidal agent Punica granatum Linn. bark and Canna indica Linn. root.
812
Phytotherapy Research, 18(7), 501–506. https://doi.org/10.1002/ptr.1168.
813
Van Jaarsveld, L. C., Kriel, W.-M., & Minnaar, A. (2006). First report of Puccinia thaliae on
814
canna lily in South Africa. Plant Disease, 90(1), 113–113. https://doi.org/10.1094/PD-90-
815
0113C.
37
816
Verma N., B. B. C., & Sharma B.O. (2012). Glucosidase inhibitory and radical scavenging
817
properties of lichen metabolites salazinic acid, sekikaic acid and usnic acid. Hacettepe
818
Journal of Biology and Chemistry, 40(1), pp.7-21.
819
Wandee, Y., Uttapap, D., Puncha-arnon, S., Puttanlek, C., Rungsardthong, V., & Wetprasit, N.
820
(2015). Quality assessment of noodles made from blends of rice flour and canna starch.
821
Food Chemistry, 179, 85–93. https://doi.org/10.1016/j.foodchem.2015.01.119.
822
Ward, P., Small, I., Smith, J., Suter, P., & Dutkowski, R. (2005). Oseltamivir (Tamiflu®) and its
823
potential for use in the event of an influenza pandemic. Journal of Antimicrobial
824
Chemotherapy, 55(suppl_1), pp. i5-i21.
825 826
White, N. J. (1997). Assessment of the pharmacodynamic properties of antimalarial drugs in vivo. Antimicrobial Agents and Chemotherapy, 41(7), p.1413.
827
Woot-Tsuen, W. L., Busson F., & Jardin C. (1968). Food composition table for use in Africa. US
828
Department of Health, Education, and Welfare, Public Health Service, Health Services
829
and Mental Health Admin., National Center for Chronic Disease Control Nutrition
830
Program.
831
December 2018).
Internet:
http://www.fao.org/docrep/003/X6877E/X6877E00.htm (accessed
832
Yasunaka, K., Abe, F., Nagayama, A., Okabe, H., Lozada-Pérez, L., López-Villafranco, E.,
833
Muñiz, E. E., Aquilar, A., & Reyes-Chilpa, R. (2005). Antibacterial activity of crude
834
extracts from Mexican medicinal plants and purified coumarins and xanthones. Journal of
835
Ethnopharmacology, 97(2), 293–299. https://doi.org/10.1016/j.jep.2004.11.014.
836
Yue, G. G.-L., Lee, J. K. M., Kwok, H.-F., Cheng, L., Wong, E. C. W., Jiang, L., Yu, H., Leung,
837
H-W., Wong, Y-L., Leung, P-C., Fung, K-P., & Lau, C. B. S. (2015). Novel PI3K/AKT 38
838
targeting anti-angiogenic activities of 4-vinylphenol, a new therapeutic potential of a well-
839
known
840
https://doi.org/10.1038/srep11149.
styrene
metabolite.
Scientific
Reports,
5(1),
11149.
841
Zengin, G., Uysal, A., Gunes, E., & Aktumsek, A. (2014). Survey of phytochemical composition
842
and biological effects of three extracts from a wild plant (Cotoneaster nummularia Fisch.
843
et Mey.): A potential source for functional food ingredients and drug formulations. PLoS
844
ONE, 9(11), e113527. https://doi.org/10.1371/journal.pone.0113527.
845
Zhao, Y., Lee, M.-J., Cheung, C., Ju, J.-H., Chen, Y.-K., Liu, B., Hu, L-Q., & Yang, C. S.
846
(2010). Analysis of multiple metabolites of tocopherols and tocotrienols in mice and
847
humans.
848
https://doi.org/10.1021/jf904464u.
Journal
of
Agricultural
and
Food
Chemistry,
58(8),
4844–4852.
849
Zhong, Y., & Shahidi, F. (2012). Lipophilised epigallocatechin gallate (EGCG) derivatives and
850
their antioxidant potential in food and biological systems. Food Chemistry, 131(1), 22–30.
851
https://doi.org/10.1016/j.foodchem.2011.07.089.
852
Zucker, M. B., & Peterson, J. (1970). Effect of acetylsalicylic acid, other nonsteroidal anti-
853
inflammatory agents, and dipyridamole on human blood platelets. The Journal of
854
Laboratory
855
https://doi.org/10.5555/uri:pii:0022214370900958.
and
Clinical
Medicine,
76(1),
66–75.
39
Table 1. Extraction and HPLC conditions for different vitamins
Sl Vitamin Extraction
A
Dried rhizome in methanol
2
D
Dried rhizome in methanol and hexane*
3
E
1
4
C
Dried rhizome in hexane and petroether*
Fresh rhizome in water
Mobile Phase A: Methanol B: 20 mm PB (pH 2.5) A:B ratio 90:10 Run time: 10 min Flow rate: 1.0 ml/min Methanol Run time: 10 min. Flow rate: 1.0 ml/min A: Methanol B: Water A:B ratio 96:4 Run time: 10 min Flow rate: 1.0 ml/min 1 mM NaH2PO4, 1 mM EDTA, pH adjusted to 3.0 by Phosphoric acid Run time 5 min. Flow rate: 1.0 ml/min
Column Temperat ure
Injection Volume
Wavelength (nm)
Reference
30℃
20 µl
325 and 350
Cosmosil technical note
30℃
15 µl
265
Cosmosil technical note
45℃
50 µl
292
Gimeno et al. (2000)
30℃
25 µl
245
Campos et al. (2009)
*Both solvent extracts were separately analyzed for vitamins
1
Table 2. Proximate, ultimate, vitamin, and mineral composition of Canna indica rhizome Proximate composition (100 g dry weight) Crude Fiber (wt%, extractives-free basis) Starch (%) Total Crude Protein (wt%, extractives-free basis) Total Lipid (%) Calorific values (bomb calorimeter) (kcal/100 g dw) Ultimate analysis (Atomic wt%, dry-ash free basis) Nitrogen Carbon Hydrogen Minerals (mg/100 g dry biomass basis) Na Mg P K Ca Mn Fe Cu Zn Vitamins (µg/g dry biomass basis) Vitamin A (Retinoic acid)** Vitamin D (D3 and D2) # Vitamin E (Tocopherol)$ Vitamin B5 (Pantothenic acid)* Vitamin C (L-Ascorbic acid)∆**
25±2 28±3 4.7±0.6 5.7±1.2 423
0.75 40.4 6.41
30.7 13.2 6.8 199 2.13 0.04 0.53 0.04 0.25
10.3 ---11
∆
µg/g Fresh biomass basis, *Detected using HR-LCMS, **Detected in both HPLC and LCMS, Only its metabolic products were detected in HR-LCMS, #Not detected in either HPLC or LCMS. $
2
Table 3. Total phenolic, flavonoids, and antioxidant activities of different extracts and standards Rhizome extracts and standards Ethyl acetate Acetone Methanol Water GA BHA BHT
Total Phenolics$
Total Flavonoids∆
DPPH*
ABTS*
230±10c
91±4c
48±4e
53±10f
334±10a 250±10b 110±10d ND ND
220±10a 122±4b 0 ND ND ND
21±3c 23±3d d 34±2 42±3e 61±4f 114±5g a 2.8±0.8 4.3±0.9a 7.2±1.8b 12±1b 16±3c 19±1c
Superoxide* CUPRAC#
FRAP$
Metal chelating@
Total Antioxidant activityΨ
NA
230±10b
91±10c
1.3±0.2c
300±10
170±10a 210±10b NA ND 164±10a ND
640±10a 590±10a 50±4c ND ND
180±10a 122±10b 50±2d ND ND
2.9±0.2a 4.1±0.4b NA ND ND ND
520±10a 350±10b 80±10d ND ND ND
*IC50 in µg/ml; #µg BHAE/mg extract; $µg GAE/mg extract; @µg EDTAE/mg extract; ∆µg QCTE/mg extract; Ψµg AE/mg extract; ND-Not determined, NA: No activity showed. Results are mean values of three determinations ± SD. Means sharing the same superscript are not significantly (p≤0.05) different from one another.
3
Table 4. Inhibitory effect of acetone extract of C. indica and standard (BHA) against βcarotene oxidation in a β-carotene-linoleate model system at 50°C for 120 min. Sample Control (no antioxidant) Acetone extract (200 ppm)* Acetone extract (100 ppm)* BHA (200 ppm)
% Inhibition after 120 min 11±1c 82±4a 56±4b 88±3a
Results are mean values of three determinations ± SD. Means sharing the same superscript are not significantly (p≤0.05) different from one another.
4
Table 5. TBARS values (mg MDA eq/Kg) in a meat model system over a 7-day period in the presence of an acetone extract of C. indica and standard (BHT). Sample Control Acetone extract (300 ppm)* Acetone extract (150 ppm)* BHT (300 ppm)
Day 0 1.2±0.0d
Storage Period Day 3 Day 5 6.5±0.2c 6.9±0.2d
Day 7 8.7±0.3c
0.081±0.01a
0.65±0.01a
0.96±0.01a
1.2±0.1a
0.45±0.02c
2.9±0.1b
3.6±0.1c
4.5±0.1b
0.13±0.01b
0.55±0.02a
2.5±0.1b
4.3±0.1b
Results are mean values of three determinations ± SD. Means sharing the same superscript are not significantly (p≤0.05) different from one another. *The ppm values in parenthesis for rhizome extract indicates GA equivalent phenol.
5
Table 6. Enzyme inhibitory activity of different solvent extracts of Canna indica rhizome Rhizome extracts
IC50 for Enzyme Inhibition Experiments α-amylase α-glucosidase Acetylcholinesterase
Ethyl acetate
21% (166)
N
N
Acetone Methanol Water
18% (166) N N
27±3 N 2.4±0.3
N N N
N – no inhibition, Values in the parentheses represents the maximum concentration (µg/ml) of solvent extract used.
6
Table 7. List of major secondary metabolites identified using HR-LCMS/MS in solvent extracts of Canna indica rhizome. (The ethyl acetate, acetone, methanol and water solvent extracts were separately subjected to HR-LCMS/MS.) Sl.
Secondary Metabolites
Rt (min)
1
m-Salicylic acid*
7.48
2
Phenyl acetic acid*
8.03
3
Ellagic acid*
8.94
4
p-Coumaric acid*
9.15
5
4-Hydroxystyrene*
9.16
6
Triamcinolone*
7
Rosmarinic acid*
8
(-)-Usnic acid*
9
Coumarin*
10 Isoeugenitol*
11 Pantothenic acid ** 12
Acetylsalicylic acid ** (aspirin)
9.29
Biological Importance Ethyl acetate extract Analgesic, Antiinflammatory, Anti-pyretic Rab-prenylation inhibitor, Antioxidant Antioxidant’ Antiproliferative, Apoptosis-inducing Antioxidant, Antiinflammatory, Antibacterial, Analgesic Anti-angiogenic, Antiimflammatory Self-regulating drug carriers
Antioxidant, Dioxygenase inhibitor Antioxidative, 10.80 Cardiovascular-protective’ Antibiotic Antibacterial, Antiinflammatory, 11.63 Anticoagulant, Antifungal, antiviral, Antitumor Antioxidant, Antiinflammatory, 12.80 Cardiovascular properties, Anesthetic, Analgesic Vitamin B5, Synthesis of 5.57 Coenzyme A 10.01
10.07 Anti-inflammatory drug
Reference(s)
Pierpoint (1994) Deraeve et al. (2012) Landete (2011) Lou et al. (2012); Pei et al. (2016) Yue et al. (2015) Cato et al. (2001); Cevc & Blume (2003); FughBerman & Ernst (2001) Lee & Scagel (2009); Petersen (2003) Behera, Mahadik, & Morey (2012); Ingólfsdóttir (2002); Verma & Sharma (2012) Cai et al. (2004); Huang, Cai, & Zhang (2009); Yasunaka et al. (2005)
Daniels et al. (2011) Bean & Hodges (1954); Lipmann et al. (1947) Berk et al. (2013); Zucker & Peterson (1970)
Acetone extract 1
m-Hydroxyphenylpyruvic acid*
8.06
2
Psoromic acid*
8.79
3
Ellagic acid*
8.92
Possible antioxidant Rab-prenylation inhibitor, Antioxidant Antioxidant’
-Deraeve et al. (2012) Landete (2011) 7
4
5 6 7
8 9
1 2 3 4 5
p-Coumaric acid*
9.16
Antiproliferative, Apoptosis-inducing Antioxidant, Antiinflammatory, Antibacterial, Analgesic
oHydroxyphenylpyruvic 10.01 Possible antioxidant acid lactone* Antioxidant, Dioxygenase Rosmarinic acid* 10.03 inhibitor Antioxidative, Usnic acid* 10.81 Cardiovascular-protective, Antibiotic Acetylsalicylic acid * * 9.91 Anti-inflammatory (aspirin) Stimulates mitochondrial Lipoamide ** 14.31 biogenesis Methanol extract m-HydroxyphenylPhenolic acid; Possible 8.10 pyruvic acid* antioxidant. Rab-Prenylation Inhibitor, Psoromic acid* 8.83 Antioxidative o-Hydroxyphenyl10.04 Possible antioxidant. pyruvic acid lactone* Antioxidant, Dioxygenase Rosmarinic acid* 10.05 inhibitor Antioxidative, Usnic acid* 10.83 Cardiovascular-protective, Antibiotic
6
Pseudohypericin*
Antiretroviral activity, 11.27 Anticancer property, Apoptosis inducing
7
3-(4-ydroxyphenyl) pyruvic acid **
9.87
1
3,4-Dihydroxyphenylpropionic acid*
6.73
Antioxidant property
2
Choline **
1.64
Essential nutrient
3
Metaraminol **
2.69
Adrenergic receptor agonist
Phenolic acid; Possible antioxidant Water extract
Lou et al. (2012); Pei, Ou, Huang, & Ou (2016)
-Lee & Scagel (2009); Petersen (2003) Behera et al. (2012); Ingólfsdóttir (2002); Verma & Sharma (2012) Berk et al. (2013); Zucker & Peterson (1970) Shen et al. (2011)
Deraeve et al. (2012) -Lee & Scagel (2009); Petersen (2003) Behera et al. (2012); Ingólfsdóttir (2002); Verma & Sharma (2012) Çırak, Radušienė, & Çamas (2008); Meruelo, Lavie, & Lavie (1988); Schempp, Simon-Haarhaus, & Simon (2002) --
Tang et al. (2011) Hollenbeck (2012); Fischer et al. (2010); Lee et al. (2010) McDonald & Santucci (2004) 8
4
Fendiline **
5
Swietenine **
7.16
Antianginal agent Anti-Hyperglycemic 12.82 property
Bayer & Mannhold (1987) Dewanjee et al. (2009)
* HR LCMS in negative mode ** HR LCMS in positive mode
9
Fig 1. Visualization of the damage induced by hydroxyl radicals on genomic DNA (a) and AAPH on protein (BSA) (b) in the presence and absence of acetone extracts from C. indica using agarose gel electrophoresis and SDS PAGE analysis. (a) Lane 1. DNA incubated without Fenton’s reagent; Lane 2. DNA incubated with Fenton’s reagent; Lanes 3-12, DNA incubated with Fenton’s reagent in the presence of 0.04, 0.08, 0.17, 0.35, 0.70, 1.39, 2.79, 5.58, 11.1 and 22.3 µg/ml of acetone extract, respectively (final concentrations). (b) Lane 1: BSA incubated without AAPH. Lane 2: BSA with AAPH, Lane 3 to 9: BSA with AAPH in presence of 2.6, 5.2, 10.4, 20.8, 41.6, 83.2 and 166 µg/ml, of acetone extract, respectively.
Fig 2. α-Glucosidase inhibition plot for acetone extract (a) and water extract (b) of C. indica with increasing concentrations (mean values).
Fig 1. (a) and (b)
Fig 2. (a) and (b)
Highlights Canna indica was found to be a good source of starch, vitamins and minerals. Rhizomes were shown to be a good source of antioxidants, showing significant activity in food and biological model systems. Rhizome extracts were found to contain α-glucosidase inhibitory metabolites. Major compounds such as rosmarinic acid, psoromic acid, usnic acid, ellagic acid, coumaric acid, isoeugenitol and swietenine were identified using HR LC-MS/MS.