Accepted Manuscript Title: “In vitro antioxidant and antidiabetic activities of biomodified lignin from Acacia nilotica wood” Author: Anand Barapatre Keshaw Ram Aadil Bhupendra Nath Tiwary Harit Jha PII: DOI: Reference:

S0141-8130(15)00015-X http://dx.doi.org/doi:10.1016/j.ijbiomac.2015.01.012 BIOMAC 4819

To appear in:

International Journal of Biological Macromolecules

Received date: Revised date: Accepted date:

5-6-2014 26-11-2014 8-1-2015

Please cite this article as: A. Barapatre, K.R. Aadil, B.N. Tiwary, H. Jha, “In vitro antioxidant and antidiabetic activities of biomodified lignin from Acacia nilotica wood”, International Journal of Biological Macromolecules (2015), http://dx.doi.org/10.1016/j.ijbiomac.2015.01.012 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. 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.

23

Title:

lignin from Acacia nilotica wood”

25 26

Authors:

Department of Biotechnology, Guru Ghasidas Vishwavidyalaya

cr

Affiliation/Institute:

(A Central University), Bilaspur, Chhattisgarh, India. 495009

29

Address for correspondence: Department of Biotechnology, Guru Ghasidas

us

30

Anand Barapatre, Keshaw Ram Aadil, Bhupendra Nath Tiwary and Harit Jha *

27 28

“In vitro antioxidant and antidiabetic activities of biomodified

ip t

24

Vishwavidyalaya (A Central University), Bilaspur,

32

Chhattisgarh, India. 495009

an

31

Telephone No. :

+91-9826630805

34

Email ID:

[email protected]

Ac ce p

te

d

M

33

2 Page 1 of 36

35 36

Abstract The antioxidant and antidiabetic activity of biomodified alkali lignin extracted

38

from a deciduous plant Acacia nilotica, was evaluated in vitro. The extracted alkali

39

lignin was subjected to microbial biotransformation by ligninolytic fungus Aspergillus

40

flavus and Emericella nidulans. These modifications were done under varying

41

concentration of carbon to nitrogen sources. The structural feature of the lignin

42

samples were compared by FTIR, functional group analysis and 13C solid state NMR.

43

All lignin samples were tested for antioxidant efficiency, reducing power and H2O2

44

scavenging power. Modifications in all lignin samples showed correlation with their

45

antioxidant scavenging activity and reducing power. Antidiabetic properties were

46

evaluated in terms of in vitro glucose movement inhibition and α-amylase inhibition

47

assay. Modified samples exhibited increased glucose binding efficiency as

48

demonstrated by the decreased glucose diffusion (55.5–76.3%) and 1.16-1.18 fold

49

enhanced α-amylase inhibition in comparison to their control samples. The results

50

obtained demonstrate that the structure and functional modifications in lignin

52 53 54

cr

us

an

M

d

te

Ac ce p

51

ip t

37

significantly affects its bioefficacy in term of antioxidant and antidiabetic activities.

Key Word: Acacia lignin, biotransformation, antioxidant, antidiabetic, α-amylase, In vitro glucose movement.

3 Page 2 of 36

55 56 57

1. Introduction Plant phenolics and polyphenols have been increasingly enticing the attraction due to their beneficial therapeutic values including antioxidant,

59

inflammatory, cardio-protective, anticancerous, chemo-preventive and neuro-protective

60

properties. Polyphenols have been considered, a health food supplement and are claimed

61

to possess health promoting or disease-preventing properties [1]. Lignin, the second

62

most abundant natural macromolecule (polyphenolic in nature and 10–35% of dry wt. of

63

lignocellulosic biomass), is a natural polymerized product of optically active p-

64

hydroxycinnamyl alcohol monomers and related monolignols (p-coumaryl, coniferyl,

65

and sinapyl) formed by oxidative reactions. This polymer is the result of various inter

66

unit linkages in the monomer and monolignols (e.g. β-O-4, β-5, β−β, biphenyl (5-5), 4-

67

O-5) [2]. The precise chemical structure of lignin is not known because of its complex

68

polymeric nature and due to many random coupling. Alkali lignin is currently the largest

69

produced among all lignin classes and a less valuable co-product of biofuel and paper

70

pulp industries, which is separated from fibres by a chemical pulping (mainly soda and

72 73 74 75

cr

us

an

M

d

te

Ac ce p

71

antimicrobial, anti-

ip t

58

sulphite) process [3].

Acacia nilotica, locally known as “Babul”, is a multipurpose deciduous tree of

Mimosaseae family predominantly found in central India. This plant contains a variety of bioactive components such as ellagic acid, isoquercitin, leucocyanadin, kaempferol-7diglucoside, derivatives of (+)-catechin, apigenin derivatives etc. [4]. Traditionally in the

76

central region of India leaves, pod, bark and root of A. nilotica is used for the treatment

77

of various diseases related to oral, bone and skin, like cold, bronchitis, diarrhoea,

78

dysentery, biliousness, bleeding piles and Leucoderma [5].

4 Page 3 of 36

Oxidative damages creates by free radicals, play a substantial role in the evolution of

80

human diseases. Toxicity of free radicals contributes to proteins and DNA damage,

81

inflammation, tissue injury and subsequent cellular apoptosis, which finally leads to

82

cancer, emphysema, cirrhosis, arteriosclerosis and arthritis [6]. Oxidative stress is

83

created in the body due to a disruption in the equilibrium between the production of

84

reactive oxygen/nitrogen species (ROS/RNS) and the removal via the antioxidant

85

defence system [7]. Antioxidants can interfere with the oxidative processes by reacting

86

with free radicals, chelating catalytic metals and also by acting as oxygen scavengers

87

thus helping the human body to reduce oxidative damage. From various epidemiological

88

studies, it is proved that polyphenolic compounds possess an excellent antioxidant

89

properties [6, 8]. Previous studies also reported that the polyphenols found in Acacia sp.

90

plants having good antioxidant power [4, 5, 9, 10]. As a complex phenolic polymer,

91

lignin also possess a respectable medicinal properties [2, 3, 11].

d

M

an

us

cr

ip t

79

Over the last century changes in human behaviour and life style have resulted in a

93

dramatic increase in the incidence of diabetes world over. Presently, it is estimated that

94

more than 220 million suffer from diabetes in which 90% is from type 2 diabetes Type 2

96 97 98 99

Ac ce p

95

te

92

diabetes is the results of ineffectiveness of insulin and the primary cause of complications linked to cardiovascular disease, renal failure, blindness, neurological complications, and so on [12]. Currently available conventional therapies for the treatment of diabetes include insulin and oral antidiabetic agents such as sulfonylurea, biguanides, and alpha-glucosidase inhibitors. Traditionally, many active compounds of

100

plant origin including several Acacia sp. plants, have been also employed in the

101

treatment of diabetes, mostly the secondary metabolites including alkaloids, flavonoids,

102

phenolics, steroids, carbohydrates, glycopeptides, terpenoids etc. [13]. The inhibition of

5 Page 4 of 36

103

enzyme like α-amylase as well as the delay in glucose absorption to be an important

104

strategy in the management of blood glucose level in type 2 diabetic [14]. Based on the fact that structural heterogeneity will affect the chemical properties of

106

lignin, we report the bioefficacy of biologically modified lignin in terms of the

107

antioxidant, antiradical and hydrogen peroxidase scavenging property and also evaluate

108

the effectiveness of modified lignin as an inhibitor of α-amylase activity and in vitro

109

glucose movement.

110

2. Material and methods

111

2.1. Chemical and reagents

an

us

cr

ip t

105

Gallic acid, D-glucose, α-amylase (EC 3.2.1.1), 1, 1-diphenyl-2-picrylhydrazyl

113

(DPPH), neocuproine (Nc), catalase from bovine liver (966 U mg-1) were purchased

114

from Sigma–Aldrich Inc. (Mumbai, India). All other chemicals and reagents used were

115

of high purity analytical grade and purchased from Merck Pvt. Ltd. (India). Ultrapure

116

water (Elix, Merck Milipore, India) was used throughout the experiment. Wood dust (18

117

mesh size) of A. nilotica hardwood was procured locally from the saw mill of Bilaspur,

118

Chhattisgarh, India.

120 121 122 123

d

te

Ac ce p

119

M

112

2.2. Extraction and characterization of alkali lignin Alkali extraction of lignin was achieved by treatment of wood dust with an aqueous

solution of NaOH (1.2% w/v) in a 1 L glass flask for 1 h at 120 ºC, using a solid/liquid ratio of 1:10 (g/mL). The solution was filtered through Whatman filter paper No. 4 to remove wood dust. The filtrate (black liquor) was concentrated by slow heating at 60 ºC

124

in an oven and acidified up to pH 5.5 with 6 M HCl. The water-soluble hemicellulosic

125

fraction was removed by precipitation, after adding two volumes of 95% ethanol (v/v).

126

The precipitated hemicellulosic were removed by gravity filtration. The remaining

127

filtrate was concentrated to 20–30 mL, and the pH was adjusted to 1.5–2.0 with 6 M

6 Page 5 of 36

HCl. The alkali lignin was precipitated and sediments by centrifugation at 10,000 rpm

129

(Remi R-24, India) for 10 min. The pellet was dried and stored at room temperature for

130

further study [15].

131

2.3. Biodegradation and characterization of alkali lignin

132

2.3.1. Microorganism (fungus)

ip t

128

The two potent ligninolytic fungus A. flavus and E. nidulans were used for the

134

biotransformation of alkali lignin. The strains were isolated from soil samples collected

135

from Guru Ghasidas Vishwavidyalaya campus, Bilaspur (C.G.) and near the effluent

136

discharge site of the Orient paper mill situated in Amalai (M.P.), India. Two potentially

137

ligninolytic strains were characterized and identified based on morphological

138

characterization and partial gene sequencing of Internal Transcribed Spacer (ITS)

139

regions as A. flavus (F10, NCBI accession no. KC91163) and E. nidulans (APF4, NCBI

140

accession no. KC911632) respectively. The strains were maintained on malt agar slants

141

for further use.

142

2.3.2. Basal culture medium

144 145 146 147 148

us

an

M

d te

Biodegradation of lignin was performed under different carbon to nitrogen ratio i.e.

Ac ce p

143

cr

133

low and high in Basal Salt Medium (BSM). The BSM contained gL-1 of KH2PO4, 0.2 g; MgSO4.7H2O, 0.05 g; CaC12, 0.01 g supplemented with a 1 mL mineral solution. The mineral solution contained (in gL−1) nitrilotriacetate, 1.5; MgSO4 .7H2O, 3.0; MnSO4.H2O, 0.5; NaCl, 1.0; FeSO4.7H2O, 0.1; CoSO4, 0.1; CaC12, 0.082; ZnSO4, 0.1; CuSO4.5H2O, 0.01; AlK(SO4)2, 0.01; H3BO3, 0.010; NaMoO4, 0.01. The high carbon

149

low nitrogen (HCLN) medium contained 56 mM D-glucose as a carbon source and 2.4

150

mM nitrogen (0.6 mM NH4NO3 and 0.6 mM L-asparagine) whereas the low carbon high

151

nitrogen (HNLC) medium contained 8.8 mM D-glucose was and 24 mM nitrogen

7 Page 6 of 36

152

(NH4NO3 and L-asparagine, 6 mM each).The pH of BSM was maintained at 5.6-5.8 for

153

biomodification of alkali lignin [16].

154

2.3.3. Biomodification and characterization of alkali lignin Two hundred mililiter of HCLN and HNLC BSM medium maintained at pH 5.6-5.8

156

were transferred to 500 mL erlenmeyer flasks. The autoclaved media were aseptically

157

inoculated with 3 bores (6 mm diameter) of 7 days old culture of the strains F10 and

158

APF4 and incubated at 28 °C for 21 days under static condition. The fungal mat was

159

separated from the medium by filtration through sterilized Whatman filter paper No 4.

160

The modified lignin was recovered from filtrate by the same method as described in

161

section 2.2. Uninoculated media were used as negative control.

an

us

cr

ip t

155

Biotransformed alkali lignin was characterized by FTIR. A small fraction of the

163

sample was ground properly with equal amount of KBr. The FTIR spectrum was

164

obtained in the range of 400–4000 cm-1 using FTIR spectrophotometer Affinity A1,

165

(Shimadzu, Japan).

168 169 170 171 172

d te

167

A high-resolution 1D solid-state

13

C NMR spectra were obtained with the Cross-

Polarization Magic-Angle Spinning (CPMAS) technique on a Bruker spectrometer

Ac ce p

166

M

162

(Indian Institute of Science, Bangalore, India) operating at 100.525 MHz frequency for the 13C carbon nuclei. A total of 300 mg solid sample was used for the NMR spectra at 294 K. The 2000-6000 scan was performed to obtain a 1D spectra with a 29.10 ms acquisition time and 5 s relaxation delay. 2.4. Total polyphenol content (TPC)

173

Total polyphenol content (TPC) was determined by reaction with Folin-Denis

174

reagent [17]. One mL of each lignin sample (50 μg/mL) was added to 0.5 mL of Folin-

175

Denis reagent. After 30 sec, 1 mL of 20% (w/v) sodium carbonate was added and the

176

volume was made up to 5 mL with distilled water. The mixture was allowed to stand at

8 Page 7 of 36

room temperature for 10 min. The absorbance of resulting blue complex was measured

178

at 765 nm against blank using UV-Visible double beam spectrophotometer (Shimatzu

179

UV-1800, Japan). A calibration curve of gallic acid was prepared, and phenolic contents

180

were determined from the linear regression equation of this curve. The results were

181

expressed as μg gallic acid equivalents (GAE) per milligram of dry material.

182

2.5. Functional group analysis

183

2.5.1. Phenolic hydroxyl groups by ultraviolet-spectroscopy

cr

ip t

177

The content of hydroxyl phenolic units in lignin fractions was determined by UV

185

spectroscopy as described Aadil et al. [9]. This method is based on the difference in

186

absorption of lignin samples at pH 6 (495 mL of 0.2 N potassium dihydrogen phosphate

187

solution mixed with 113 mL of 0.1 N NaOH and diluted to 2 L with distilled water) and

188

at alkaline buffer solution pH 12 (0.1 N of boric acid in 0.1 N NaOH solution). The

189

difference in spectra was obtained by taking the absorbance of the alkaline solution

190

relative to that of the neutralized solution in the range of 200–400 nm. The phenolic

191

hydroxyl group content of lignin samples was calculated using ∆amax.

193 194 195 196 197 198

an

M

d

te

% phenolic hydroxyl = ∆amax X 17/41

Ac ce p

192

us

184

2.5.2. Carboxyl groups determination by aqueous titration method An amount of 25 mg of recovered lignin samples was suspended in 25 mL of an

alkaline 0.1 N NaOH solution and stirring for 3 h. The pH was adjusted to 12 with 0.1 N NaOH. Followed by potentiometric titration with 0.1 N HCl, as described Aadil et al. [9].

2.6. Total reducing power assay

199

Reducing the power of all samples was determined according to the method

200

of Oyaizu [18]. Briefly, different concentrations of lignin samples were prepared in

201

sodium phosphate buffer (200 mM, pH 6.6) and 2.5 mL of each sample was separately

9 Page 8 of 36

mixed with 2.5 mL of 1% (w/v) potassium ferricyanide. The mixture was incubated at

203

50 °C for 20 min. A mixture containing all the reaction reagents except the test material

204

serves as the control. The reaction was stopped by adding 2.5 mL of 10% (w/v) TCA and

205

the mixture was centrifuged at 1,750 rpm for 10 min. The upper layer (2 mL) was mixed

206

with 1 mL of 0.1% (w/v) of ferric chloride and made up to 8 mL with deionised water.

207

The perl’s prussian blue colour formed due to reduction in Fe3+ was measured at A700.

208

The EC50 of extracts were calculated from the graph of A700 versus extracts

209

concentration.

210

2.7. DPPH free radical scavenging activity

an

us

cr

ip t

202

The antiradical activity of lignin samples was measured based on their reaction with

212

stable free radical DPPH* and subsequent reduction in λmax of DPPH* [19]. In this

213

reaction 1.5 mL sample, (100 µg/mL, prepared in 50 mM phosphate buffer, pH 7.5) was

214

allowed to react with 1.5 mL of 100 µM methanolic solution of DPPH* for 30 min in

215

darkness at room temperature. The decrease in absorbance was measured at A515.

216

DPPH* radical scavenging capacity was calculated using the following equation

218 219 220 221 222

d

te

DPPH* scavenging (%) = [(A0 – A1) / A0 x 100]

Ac ce p

217

M

211

where A0 and A1 are absorbance of DPPH* radical at 515 nm in the absence and presence of the samples.

2.8. Hydrogen peroxide scavenging (HPS) assay HPS capacity of lignin samples was estimated by cupric reducing antioxidant

capacity method according to Ozyurek et al. [20]. In this method, hydrogen peroxide

223

incubation solution and scavenger solutions were prepared. The hydrogen peroxide

224

incubation solution (used as a reference) contained 0.7 mL of phosphate buffer (0.2 M,

225

pH 7.4), 0.4 mL of 1 mM H2O2, 0.4 mL of 0.1 mM CuCl2.2H2O, whereas scavenger

226

solutions (I and II) were prepared in two test tubes containing 0.2 mL of test samples,

10 Page 9 of 36

0.5 mL of phosphate buffer (0.2 M, pH 7.4), 0.4 mL of 1 mM H2O2 and 0.4 mL of 0.1

228

mM CuCl2.2H2O (identical up to this step). The mixtures were incubated for 30 min at

229

37 ºC in water bath. After incubation 0.4 mL of H2O was added to the reference and

230

scavenger solution-I and 0.4 mL of catalase solution (268 U mL-1) in scavenger solution-

231

II. The mixture was vortexes for 30 sec. From the above incubated mixtures, 1 mL

232

solution was mixed with 1 mL of Nc (0.0075 M, freshly prepared in ethanol), 1 mL of

233

0.1 mM CuCl2.2H2O and 2 mL of ammonium acetate buffer (1 M, pH 7). After 30 min,

234

the absorbance of the final solution was taken at 450 nm against the reagent blank. The

235

HPS activity (%) of samples was calculated using the following formula:

an

us

cr

ip t

227

HPS (%) = [A0 – {A1 – A2}] / A0 X 100

236

where A0 is the absorbance of reference hydrogen peroxide incubation solution, A1 and

238

A2 are the absorbance of scavenger solution-I and –II, respectively.

239

2.9. Antidiabetic assay by in vitro glucose movement

d

M

237

To evaluate the effects of biotransformed lignin on glucose movement an in vitro

241

model system was used according to Büyükbalci and Nehir [21] with slight

242

modification. The dialysis tube (6 cm X 14.3 mm) (HiMedia, Mumbai, India; pore size

244 245 246 247

Ac ce p

243

te

240

2.4nm) was filled with a total volume of 6 mL test sample mixtures containing 1 mg/mL biotransformed lignin and 1.65 mM D-glucose (prepared in 0.15 M NaCl) in the ratio of 2:1 (v/v). The dialysis tube was sealed at both ends and placed in a flask containing 45 mL 0.15 M NaCl. Dialysis experiment was performed on an orbital shaker water bath (100 rpm) at 37 °C for 3 h to induce the movement of glucose into the external solution.

248

Concentration of glucose outside the dialysis tubing was measured by DNS

249

(dinitrosaliacylic acid) reagent. The control experiment was conducted in the absence of

250

the test sample.

251

2.10. In vitro α-amylase inhibition assay

11 Page 10 of 36

The α-amylase inhibitory activity was determined by the method of Quesille-

253

Villalobos et al. [22]. A total of 500 µL of different concentrations of each sample (0.1,

254

0.5, 1 and 5 mg/mL) and 500 µL of 0.02 M sodium phosphate buffer (pH 6.9 with 0.006

255

M NaCl) containing α-amylase (0.5 mg/mL) was incubated for 10 min at 25°C. After

256

preincubation, 500 µL of 1% (w/v) starch solution in 0.02 M sodium phosphate buffer

257

(pH 6.9 with 0.006 M NaCl) was added to each of the pre-incubated tubes. The reaction

258

mixtures were then incubated at 25 °C for 10 min and stopped with 1 mL of DNS

259

reagent. The test tubes were further incubated in a boiling water bath for 10 min and

260

cooled to room temperature. The reaction mixture was diluted with 10 mL distilled water

261

and the absorbance was measured at 540 nm. The absorbance of blank samples (buffer

262

instead of enzyme solution) and a control (buffer in place of the sample extract) was also

263

recorded for comparison. The final activity of α-amylase was calculated by subtracting

264

the final A540 of sample with its corresponding A540 blank.

265

2.11. Statistical analysis

te

d

M

an

us

cr

ip t

252

Unless otherwise stated experiments were performed in triplicate and statistical

267

analysis was done in term of mean ± standard deviation (SD). Significance levels were

268 269 270 271 272

Ac ce p

266

calculated using Graph Pad Prism 5.0 by one way analysis of variance (ANOVA) followed by Tukey’s multiple comparison test. 3. Results and discussion

The biotransformation and biodegradation of lignin occur in a multistep process

involving Lignin Peroxidase (LiP), Manganese–dependent Peroxidase (MnP), Laccase,

273

Versatile Peroxidases (VPs) and dioxygenases enzymes, working in association with

274

small molecules and radicals [23]. The production of these enzymes is associated with

275

nutrient stress conditions. White rot basidiomycetes and brown rot ligninolytic fungi

276

display a broad diversity in response to carbon and nitrogen source and their C/N ratio.

12 Page 11 of 36

In most of the ligninolytic fungi LiP, MnP and laccase production is primarily regulated

278

by the nitrogen concentration [24, 25]. Biodegradation of lignin starts with

279

depolymerizing activity of LiP and MnP followed by demethylation activity of laccase.

280

It is reported that the side chain and aromatic rings of lignin model compounds and

281

synthetic lignin (DHPs) were cleaved via aryl cation radical and phenoxy radical

282

intermediates which is mediated by LiP/H2O2 and laccase/O2/mediator system. Whereas

283

MnP catalyse demethylation, Cα-Cβ cleavage, alkyl-aryl cleavage and Cα oxidation of

284

phenolic syringyl type β-1 and β-O-4 lignin structures [26].

us

cr

ip t

277

In present study bio-modified lignin was obtained by biomodification of lignin using

286

two ligninolytic fungi F10 and APF4. During microbial transformation of lignin by F10

287

and APF4, the onset of MnP and LiP activity in HCLN medium starts from 3rd day and

288

continuously increase up to the 21st day, whereas in HNLC medium it starts from 3rd in

289

F10 and 12th day and APF4. The activity of both enzymes is minimal in HNLC as

290

compared to HCLN medium. On the other hand, the laccase activity was high in HCLN

291

medium, whereas in HNLC medium it was very low (unpublished data). After

292

biomodification recovered biomodified lignin samples were evaluated for their TPC,

294 295 296 297

M

d

te

Ac ce p

293

an

285

functional group analysis, reducing power, hydrogen peroxide scavenging capacity, DPPH free-radical scavenging capacity, α-amylase and in vitro glucose movement inhibition activity.

3.1. Characterization of modified lignin 3.1.1. FTIR of lignin samples

298

The FTIR spectrum of the control and biomodified lining samples is presented in Fig

299

1a and 1b. All modified lignin samples (B, C, E and F) showed a broad absorption band

300

at 3410–3460 cm−1, attributed to the O-H groups stretching in phenolic and aliphatic

301

structures and oscillation of the hydroxyl group. The relative intensity of this band

13 Page 12 of 36

stretching was more in the treated sample (both HCLN F10 and HCLN APF4) as

303

compared to control sample, which indicated the phenolic ring modification by addition

304

of a hydroxyl group. Whereas in modified HNLC samples (HNLC F10 and HNLC

305

APF4), the intensity of O-H groups stretching was almost same in comparison to

306

modified HCLN samples. A strong bands at 2847 cm−1 arising from C-H stretching in

307

the aliphatic methylene group was less intense in treated sample (B, C, E and F) as

308

compared to their control samples (A and D). Bands centred on 2938 and 2842 cm−1,

309

predominantly arising from C-H stretching in aromatic methoxyl groups and in methyl

310

and methylene groups of side chains. The intensity of both peaks is reduced in treated

311

samples as compared to control samples. The increase in the intensity of -OH group and

312

decrease in C-H stretching also an outcome of laccase activity as suggested in the

313

literature [27,28].

M

an

us

cr

ip t

302

All samples displayed weak bands in the carbonyl/carboxyl region 1705–1720 cm−1,

315

assigned for unconjugated ketone or unconjugated carbonyl stretching. The intensity of

316

aromatic skeleton vibrations at 1600, 1515 and 1426 cm−1 characteristic of the aromatic

317

ring in alkali lignin showed significant decrease in treated samples as compared to

319 320 321 322

te

Ac ce p

318

d

314

controls, indicating the cleavage of the aromatic ring structure. The C–H deformation combined with aromatic ring vibration at 1462 cm−1 was observed in the untreated control samples, however, these peaks were absent in treated samples. These results suggested that the structural and functional groups of alkali lignin were altered by both the fungal strains [29].

323 324

Fig. 1a.

325

Fig. 1b.

326

3.1.2. 13C NMR 14 Page 13 of 36

327

A comparison of the

13

C NMR spectra of an untreated control lignin samples with

biodegraded lignin sample under two different nutritional conditions is presented in

329

Figure 2. In all three NMR spectra of lignin, there was an absence of signals between 90

330

and 102 ppm which indicates that the samples were free of carbohydrate contamination.

331

The signals for unconjugated carboxylic acids –COOH (178.0-167.5 ppm) were high in

332

HNLC sample in comparison to control while low in HCLN sample. The relative

333

increase in this signal could be attributed to the formation of aldehydes, acids and

334

aroxiacetic structures probably because of the oxidation of the side-chains. These results

335

suggest oxidative attack on the lignin by the microbial enzymatic system of fungus [30].

336

The signal for the aliphatic (171–168.5 ppm) and phenolic (168.5–167 ppm) hydroxyl

337

groups were observed in the lignin samples treated with fungus (HCLN and HNLC)

338

whereas they are absent in control lignin. The three aromatic region signals for

339

protonated aromatic specially unsubstituted aromatic carbons ortho or para to the

340

substituted carbon (125–103 ppm), the condensed aromatic mainly C-substituted

341

aromatic carbon (141–125 ppm), and the oxygenated aromatic mainly O-substituted

342

aromatic carbon of guaiacol (160–141 ppm) were higher in control lignin as compared to

344 345 346 347

cr

us

an

M

d

te

Ac ce p

343

ip t

328

treated lignin [31].

The region between 162 to 103 ppm belongs to aromatic carbon, a syringal units

produce strong signal at 153-151 ppm (C-3 esterified) whereas the guaicyl unit signals at 119 ppm (C-6), 115 ppm (C-5) and 111 ppm (C-2) were also observed in control and treated samples. The changes in the aromatic-C region was observed, notably the

348

decrease in the syringyl and guaiacyl amount (signals at 153 ppm and 148 ppm

349

respectively) after the fungal degradation of the alkali-lignin. The relative decrease in

350

syringyl units reflects an easier accessibility of the microorganisms to the less condensed

351

syringyl units of the polymer [30, 31].

15 Page 14 of 36

The signals at 90-57.5 ppm display aliphatic C-O bonds and α, β, γ carbons on the lignin

353

side chain. In which Cα, Cβ, and Cγ in β-O-4 can be identified in the regions 79.0-67.0,

354

90.0-78.0, and 61.5-57.5 ppm, respectively [31]. A significant decrease was observed in

355

Cγ in β-O-4 (61.8 ppm) both treated samples as compare to control, while the other two

356

signals for Cα and Cβ in β-O-4 increased in HNLC sample as compared to control. Some

357

new signal intensity (30.5, 28.98 and 26.08) were also observed in the 46-10 ppm

358

spectral region, mainly in the case of HNLC lignin sample, suggesting accumulation of

359

saturated alkyl structures (Aliphatic CH2, 35.9 ppm) in the biomodified lignin. One of

360

the signal at 35.9 corresponding to Cα in arylpropanol unit was decreased in HCLN

361

sample in comparison to control while it was unaffected in HNLC lignin sample [30].

363

cr

us

an

Fig. 2

M

362

ip t

352

3.2. Total phenolic content and functional group analysis All treated samples exhibited significantly lower TPC as compared to their

365

respective controls (Table 1). It was detected that in F10 and APF4 modified lignin

366

samples, the quantity of TPC was low under HCLN than HNLC condition. In lignin

367

degradation when lignin is exposed to peroxidases, it undergoes decomposition into

369 370 371 372

te

Ac ce p

368

d

364

lower molecular weight fragments containing methoxyl groups. Laccase demethylates these fragments and peroxidases further degrade them into smaller fragments. These smaller fragments reduce into their respective phenols by MnP, LiP and VPs which undergoes ring cleavage and form keto acids. The keto acids enter through kreb’s cycle and are metabolised by the fungus [32]. Variations in TPC were observed possibly due

373

to difference in the activity of ligninolytic enzymes under varying nutrient conditions.

374

The results obtained revealed that the TPC of samples was influenced by lignin

375

degradation under carbon and nitrogen surplus/stress conditions. Under HCLN condition

376

enzymatic activity was high due to which lignin depolymerized into smaller fragments 16 Page 15 of 36

377

and subsequently metabolized by the fungus. Previous results also supported our

378

contention that the ligninolytic enzymes activities of different fungi were low/diminished

379

in nitrogen rich medium as compare to nitrogen deficient medium [16, 23]. Functional group analysis were also carried out by UV spectrophotometric and

381

potentiometric titration methods and results were presented in Table 1. All treated

382

sample have equal and high phenolic hydroxyl group in comparison to their control

383

sample which was also confirmed by FTIR analysis. In our results a correlation was

384

found in the phenolic and carboxyl group amount in control and biomodified samples.

385

Higuchi [26] reported that, laccase oxidizes the phenolic hydroxyl groups on lignin to

386

carboxylic groups, this might be the possible reason of this correlation.

387

3.3. Reducing power assay

M

an

us

cr

ip t

380

A reducing agent contributes to antioxidant activity by donating its electron to free

389

radicals, which result in neutralization of the reactivity of the radical, and the reduced

390

species subsequently acquire a proton from the solution. It was previously reported that

391

the Acacia species contain a number of active phenol and polyphenolic contents which

392

were implied in the reducing reactions [9, 10, 33, 34]. Reducing power of all samples

394 395 396 397

te

Ac ce p

393

d

388

was expressed in the form of EC50 value (Table 1), which ranged from 405.41–1056.34 µg. From the results, it was observed that no significant change occurred in reducing power of lignin samples biotransformed under HCLN condition as compared to control, it was almost equal to the control samples. On the other hand, reducing power of other two lignin samples modified in HNLC condition (HNLC F10 and HNLC APF4) showed

398

a decrease in the reducing power. The reduction in reducing power activity might be due

399

to the partial depolymerisation of lignin polymer into low electropositive products.

400

Nevertheless, the alkali lignin (modified and unmodified) tested in this experiment

401

showed significant reducing power in the range of 0.4-1 mg/mL, whereas Sultana et al.

17 Page 16 of 36

[33] reported them ethanolic extract of A. nilotica bark contains reducing power at 10

403

mg/mL range. Kalaivani and Mathew [5] also reported that the different solvent extracts

404

of A. nilotica leaves showed good reducing ability which increased with the sample

405

concentration.

406

3.4. DPPH free radical scavenging activity

ip t

402

The radical-scavenging activity of the control and bio-modified lignin was tested

408

using a methanolic solution of the ‘‘stable DPPH*”. On accepting hydrogen from a

409

corresponding donor, it loses the characteristic purple colour. The radical scavenging

410

activity of phenolic compounds depends on the rate of abstracting the hydrogen atom

411

from a phenol molecule by a free radical and, also on the stability of the radical formed.

412

This abstracting ability was increased if some additional conjugation with substituents

413

took place [11]. From results (Table 1) it was revealed that all alkali lignin samples

414

(unmodified and modified) exhibited good scavenging activity, ranging from 37.94% to

415

71.44%. Both HCLN samples which were modified by F10 and APF4, displayed

416

increased antioxidant scavenging power i.e. 56.27% and 55.33% and both lignin samples

417

increase 21.2% and 12.6% of their free radical scavenging power with respect to HCLN

419 420 421 422

us

an

M

d

te

Ac ce p

418

cr

407

control sample. Barclay et al. [35] reported that dimeric and tetrameric phenolic lignin model compounds have significantly higher antioxidant properties. Kiliç and Yesiloglu [34] and Rice-Evans et al. [37] also reported that hydroxycinnamtes (the basic units of lignin related compound) like p-coumaric acid, ferulic acids, chlorogenic acid, caffic acid also contain good antioxidant activity. Microbial depolymerisation of lignin in both

423

HCLN modified samples formed a high quantity of small monomeric, dimeric and

424

tetrameric compounds which might have increased the radical scavenging power, which

425

was also confirmed in NMR spectra.

18 Page 17 of 36

Lignin samples which were modified in HNLC condition displayed a decrease in

427

radical scavenging property. It was observed that HNLC F10 and HNLC APF4 lignin

428

brought a decrease of 32.6% and 23.9%, respectively, in their capability to scavenge the

429

free radical as compared to HNLC control. In HNLC condition, the produced

430

depolymerized lignin fractions by the activity of LiP and MnP, was probably less

431

demethylated due to suppression in laccase activity. Heim et al. [38] reported that O-

432

methylation decrease the antioxidant properties in flavonoid related compounds.

433

Dizhbite et al. [11] also reported that the due to extension of conjugation by carbonyl

434

groups in the propanoid side chain, radical scavenging activities diminishes significantly.

435

These above statements are the probable cause of reduction of antioxidant properties of

436

lignin sample which was modified in HNLC condition.

437

3.5. Hydrogen peroxide scavenging assay

M

an

us

cr

ip t

426

Free radicals (especially OH*) are usually unstable, highly reactive, and energized

439

molecules generated from superoxide anion and hydrogen peroxide in the catabolic

440

system [7]. The results presented in Table 1, suggested that both modified HCLN

441

samples had increased hydrogen peroxide scavenging activity against control samples

443 444 445 446

te

Ac ce p

442

d

438

while in lignin samples modified under high nitrogen condition showed decrease in scavenging activity as compared to their control lignin. Sroka and Cisowski [39] studied hydrogen peroxide scavenging activities of some

water soluble phenolic acids and concluded that the hydrogen peroxide scavenging activity is positively correlated with the number of hydroxyl group bounded to the

447

aromatic ring, position of hydroxyl substitution in respect to each other and other

448

substituents (carboxyl and acetyl group) with their position in relation to the hydroxyl

449

group. They also concluded that there is a substantial correlation between antioxidant

450

and hydrogen peroxide scavenging activity of phenolic acids. Our studies also revealed a

19 Page 18 of 36

451

strong correlation (r2 = 0.83, Fig 2) between antioxidant and hydrogen peroxide

452

scavenging activity of modified lignin samples.

454

Fig. 3 3.6. Antidiabetic assay by in vitro glucose movement

ip t

453

A surge in high post-prandial plasma glucose concentrations is associated with an

456

increased risk of developing type 2 diabetes and any reduction in the postprandial

457

glucose rush is potentially advantageous in avoiding diabetes. Gallagher et al. [40]

458

studied glucose diffusion inhibition activity of 10 traditional antidiabetic plants by in

459

vitro diffusion method. They reported agrimony and avocado as the most potent ( >60%

460

inhibition) inhibitor, whereas others inhibit the glucose diffusion in the range of 6–48%.

461

They also reported that the plant extracts exhibited a concentration dependent inhibitory

462

effect on glucose movement. While in another study Büyükbalci and Nehir [21] found

463

that there was no significant effect of herbal teas and infusions (traditionally used in the

464

treatment of diabetes in Turkey) on in vitro glucose diffusion.

te

d

M

an

us

cr

455

In the present study, control and modified lignin samples were tested for inhibitory

466

effects on glucose movementout of dialysis tube. No significant inhibition in glucose

467 468 469 470 471

Ac ce p

465

movement was observed by both control lignin samples as compared to blank (Table 2). But HCLN F10 and HNLC F10 lignin samples appeared to be the most potent inhibitor in glucose movement out of the dialysis tube, decreasing movement upto 76.3% and 68.1% (p

In vitro antioxidant and antidiabetic activities of biomodified lignin from Acacia nilotica wood.

The antioxidant and antidiabetic activity of biomodified alkali lignin extracted from a deciduous plant Acacia nilotica, was evaluated in vitro. The e...
545KB Sizes 7 Downloads 10 Views