Available online at www.sciencedirect.com

ScienceDirect

Food Science and Human Wellness 6 (2017) 155–166

Comparison of chicoric acid, and its metabolites and caftaric

acid: In vitro protection of biological macromolecules and inflammatory

responses in BV2 microglial cells

Qian Liu, Fan Liu, Liling Zhang, Yajie Niu, Zhigang Liu, Xuebo Liu

Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, China

Received 11 October 2016; received in revised form 11 August 2017; accepted 11 September 2017

Available online 23 September 2017

Abstract

Chicoric acid (CA), a natural , has been used as a nutraceutical food ingredient due to its powerful antioxidant, anti-HIV and

anti-diabetic bioactivities. CA could be partly metabolized into caffeic acid (CFA) and (CTA) on cytochrome P450s in rat liver

microsomes. To compare the protective effects of CA and its metabolites on biomolecules and inflammatory responses, oxidative damage induced

by free radicals in vitro and microglial inflammation triggered by lipopolysaccharide in BV2 cells were constructed. Results showed that CA,

CTA and CFA all significantly inhibited protein degradation and carbonylation induced by hydroxyl radicals and alcoxyl radicals, and suppressed

hemin/nitrite/H2O2 triggered-nitration. Moreover, CA, CTA and CFA all exerted remarkable inhibition capacities on linoleic acid and soybean

lecithin liposomes peroxidation in a dose-dependent manner, and restrained the oxidation of herring sperm DNA, as well as the breakage of pBR322

plasmid DNA. Furthermore, CA and its metabolites suppressed lipopolysaccharide-induced decline of BV2 cell viability and the production of NO

and ROS. However, bioactivities of CA were significantly stronger than those of its metabolites within a certain concentration range. This study

provides scientific basis for the application of CA and its metabolites as nutrition and natural antioxidants.

© 2017 Beijing Academy of Food Sciences. Production and hosting by Elsevier B.V. This is an open access article under the CC BY-NC-ND

license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Keywords: Chicoric acid and its metabolites; Oxidative damage; Biomolecules; Microglia; Inflammation

1. Introduction sive ROS/RNS will attack biological macromolecules such as

proteins, lipids and DNA, disrupt cell structure and function,

Oxidative stress plays a pivotal role in the pathophysiology and also cause inflammation responses [3,4]. Nicotinamide

of diseases including neurodegenerative diseases, aging cancer, adenine dinucleotide phosphate oxidase is a major source of

obesity and cardiovascular diseases [1,2]. Once being in a state of ROS production in many non-phagocytic cells. Microglial cells

oxidative stress or disease, generation of reactive oxygen species produce inflammatory cytokines such as TNF- induced by acti-

(ROS)/reactive nitrogen species (RNS) would increase. Exces- vated nicotinamide adenine dinucleotide phosphate oxidase [5].

ROS/RNS can also act as a second messenger to activate NF␬B

signaling pathway in microglia cells, causing gene expression

related to inflammation and lead to the production of inflamma-

Abbreviations: CA, chicoric acid; CTA, caftaric acid; CFA, caffeic acid;



␣ β

ROS, reactive oxygen species; RNS, reactive nitrogen species; AAPH, 2,2 - tory mediators, including TNF- , IL-1 , NO and PGE2 [6].

Azobis (2-methylpropionamidine) dihydrochlorid; BSA, bovine serum albumin; antioxidants have been widely used due to

TBARS, thiobarbituric acid reactive substances; hsDNA, herring sperm DNA;

  the higher natural and secure properties compared with syn-

DCFH-DA, 2 ,7 -dichlorofluorescin diacetate.

∗ thetic antioxidants. A large number of studies have confirmed

Corresponding author.

that , especially plant phenols could prevent or

E-mail address: [email protected] (X. Liu).

Peer review under responsibility of Beijing Academy of Food Sciences. alleviate oxidative stress. It has been reported that polyphenolic

extract of Geoffroea decorticans (chanar)˜ exhibited antioxidant

+• •

activities by scavenging ABTS , OH, hydrogen peroxide,

and protecting protein and lipid against oxidative damage [7].

http://dx.doi.org/10.1016/j.fshw.2017.09.001

2213-4530/© 2017 Beijing Academy of Food Sciences. Production and hosting by Elsevier B.V. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

156 Q. Liu et al. / Food Science and Human Wellness 6 (2017) 155–166

2+

It has been proved that caffeic acid could improve cognitive Cu /H2O2 were determined according to the previous method

function through inhibition of lipid peroxidation and NO pro- with some modifications. CA, CFA, and CTA (10, 50, 100, 500,

duction [8]. Another study demonstrated that isoorientin could 1000 ␮mol/L) were mixed with 0.8 mg/mL BSA solution. H2O2

inhibit inflammatory responses induced by lipopolysaccharide (25 mmol/L) and CuSO4 (0.1 mmol/L) were added to induce

through ROS-related MAPK/NF B signaling pathway in BV2 protein oxidative degradation followed by the pre-incubation at

microglial cells [9]. 37 C for 30 min. And the control group used pH 7.4 phosphate

Chicoric acid (CA), isolated from chicory [10], is widely buffer instead of CuSO4 and H2O2. After incubation at 37 C

distributed in the nature, which was also found in iceberg let- for 90 min, the protein content were measured by commasie bril-

tuce, dandelion, ocimum basilicum, cymodoceanodosa and other liant blue staining, and the optical densities were analyzed by

plants [11,12]. As a natural phenolic acid, CA has intense effects Quantity One 4.6.2.

in anti-oxidation, anti-obesity, antiviral, and hpyerglycemic Assays for the inhibition of CA and its metabolites on BSA

activities [13–15]. Our previous report demonstrated that CA oxidative degradation induced by AAPH were measured by the

was metabolized to caffeic acid (CFA) and caftaric acid (CTA) on method described by Mayo et al. [18]. The treatments were the

2+ ◦

cytochrome P450s in rat liver microsomes. CA and its metabo- similar with Cu /H2O2 reaction system. Incubation at 37 C for

• • +•

lites all could significantly scavenge DPPH , OH, ABTS 2 min made AAPH (50 mmol/L) thermal decomposition, pro-

free radicals and had strong ferric reducing capacities in a dose- ducing alcoxyl radicals. After incubation at 37 C for 4 h, the

dependent manner, among which, CA has the highest functional protein content were measured by commasie brilliant blue stain-

activities [16]. However, comparisons on the protection abilities ing, and the optical densities were analyzed by Quantity One

of biological macromolecules and anti-inflammatory activities 4.6.2.

among CA and its metabolites have rarely been reported.

Therefore, this study is aimed to compare the inhibition of

2.3. BSA carbonylation

CA and its metabolites on the protein oxidation, carbonylation

and nitration, lipid peroxidation and DNA oxidation break-

DNPH method was used to detect the degree of BSA car-

age induced by hydroxyl radicals or alcoxyl radicals. Besides,

bonylation [19]. 400 ␮L samples were added into 4 mL DNPH

in order to evaluate the effects of CA and its metabolites on

solution, and placed the solution in the dark at room tempera-

lipopolysaccharide induced inflammation in BV2 microglial

ture for 1 h. Then mixed with 4 mL 10% icy trichloroacetic acid,

cells, cell viability and the production of NO, ROS were

and centrifugated at 4000 rmp at 4 C for 10 min, followed by

detected. This study will improve our understanding of the

washing precipitate three times with ethanol and ethyl acetate in

antioxidant and anti-inflammatory activities of CA and its

1:1 proportions. The absorbance of the precipitate dissolved in

metabolites, and provide a scientific basis for the application

4 mL guanidine hydrochloride was measured at 370 nm using a

of CA and its metabolites as nutrition and natural antioxidants.

UV–vis spectrophotometer. The carbonyl contents were calcu-

lated using formula (1):

2. Materials and methods

carbonyl contents (μmol/mg)

2.1. Chemicals and reagents

=

OD370/(22.0mmol/L · cm) × 1000 (1)

Chicoric acid, caffeic acid, and caftaric acid (purity ≥ 98%)

were purchased from Weikeqi Biological Technology Co.,

Ltd. (Sichuan, China). Bovine serum albumin (BSA), 2,2-

2.4. BSA nitration

azobis (2-amidinopropane) dihydrochloride (AAPH), herring

sperm DNA (hsDNA, D3159), linoleic acid (LA), Anti-DNP

Hemin/NaNO /H O reaction system was used to induce

antibody, lipopolysaccharide, and 3-(4,5-dimethyl-2-thiazolyl)- 2 2 2

protein nitration [20]. CA, CFA, and CTA (10, 50, 100, 500,

2,5-diphenyl-2-H-tetrazolium bromide (MTT) were purchased

1000 ␮mol/L) were mixed with 1.0 mg/mL BSA solution and

from Sigma Chemical Co. (St. Louis, MO, USA). Soybean

preincubating for 5 min. And then hemin (100 ␮mol/L), NaNO

lecithin was obtained from Huamaike Biotechnology., Ltd. (Bei- 2

(10 mmol/L) and H O (1 mmol/L) were added. After incuba-

2 2

jing, China). PBR322 plasmid was purchased from Takara ◦

tion at 37 C for 30 min, the levels of nitration were measured

Shuzo Co., Ltd. (Kyoto, Japan). Hydrogen peroxide (H2O2)

by western blotting, and the optical densities were quantified by

was obtained from Sinopharm Group Chemical Reagent Co.,

Quantity One 4.6.2.

Ltd. (Shanghai, China). The BCA protein assay kit was obtained

from Thermo Scientific (Rockford, IL, USA). All other reagents

were made in China and were of HPLC-grade or the highest 2.5. Western blotting

commercially available grade.

After the reaction, add 25 ␮L 5 × SDS loading buffer to

2.2. BSA oxidative degradation 100 ␮L reaction solution, and then immediately heated at 95 C

for 10 min. The proteins were separated by SDS-PAGE and

2+

Hydroxyl free radicals generated by Cu react with H2O2 transferred onto PVDF membranes (0.45 ␮m, Millipore). Block-

will cause protein oxidation [17]. The inhibition of CA and ing was carried out for 1.5 h in 5% nonfat dry milk in TBST

its metabolites on BSA oxidative degradation induced by buffer (20 mmol/L Tris, 166 mmol/L NaCl, and 0.05% Tween

Q. Liu et al. / Food Science and Human Wellness 6 (2017) 155–166 157

20, pH 7.5). Then, the membrane was washed 3 times each for 2.10. pBR322 plasmid DNA oxidative cleavage

5 min using TBST buffer. The primary antibody 3-NT (sc-32731,

Santa Cruz, 1:300) was added overnight at 4C. Then, after three By adopting DNA oxidative cleavage method, the inhibitions

washes in TBST, secondary antibodies (sc-2005, Santa Cruz) of CA and its metabolites were studied [24]. Mix 100 ng pBR322

were added for 1.5 h. After another three washes with TBST, plasmid DNA with CA, CFA, and CTA (10, 50, 100, 500,

the immunoreactive bands were visualized with an enhanced 1000 ␮mol/L), 37 C preincubate for 10 min, and add 10 mmol/L

chemiluminescence reagent (Thermo Fisher, China). AAPH to starting reactions. After 37 C for 4 h, the levels

of DNA oxidative cleavage were tested by agarose gel elec-

trophoresis, and the optical densities were quantified by Quantity

2.6. Lipidosome peroxidation

One 4.6.2.

Lipidosome peroxidation was determined by previous meth-

2.11. Cell culture and treatment

ods [21]. Mix CA, CFA, and CTA (10, 50, 100, 500,

1000 ␮mol/L) with 20 ␮L lipidosome (20 mg lecithin dissolved

Mouse microglial BV2 cells were obtained from the China

in 5 mL 0.05 mol/L pH 7.4 phosphate buffer filled with nitrogen)

Center for Type Culture Collection (Wuhan, China). BV2 cells

and preincubate for 10 min. Then 0.8 mmol/L FeSO4 was added

◦ were grown in RPMI 1640 medium with 10% fetal bovine serum,

to induce peroxidation. After 37 C for 40 min, the reaction fin- ␮ ◦

100 g/mL of streptomycin, and 100 U/mL of penicillin at 37 C

ished and the amount of thiobarbituric acid reactive substances

under a 5% CO2 atmosphere. The medium was replaced every

(TBARS) induced by lipid peroxidation was observed by col-

2 days. BV2 cells were pretreated with different concentrations

orimetry.

of CA, CFA, and CTA dissolved in dimethyl sulfoxide for 4 h,

and then treated with lipopolysaccharide (1 ␮g/mL) for 12 h.

2.7. Linoleic acid peroxidation

2.12. Measurement of cell viability

2+

Fe /Vc reaction system was adopted to induce linoleic acid

peroxidation [22]. Linoleic acid (1 mmol/L) was mixed with The cell vitality was determined using MTT assay [25]. BV2

6

×

CA, CFA, and CTA (10, 50, 100, 500, 1000 ␮mol/L). Reaction cells were seeded in 96-well plates at a density of 1 10

solution was vortexed with 50 ␮mol/L FeSO4 and 1 mmol/L Vc, cells/well and incubated over night at 37 C with 5% (v/v) CO2.

follow by 37 C water bath for 24 h away from light. The degree After various treatments, the medium was removed carefully and

of lipid peroxidation was assessed by TBARS. the cells were incubated with MTT (0.5 mg/mL) for 4 h at 37 C.

Formazan crystals formed by live cells were solubilized with

100 ␮L DMSO and absorbance at 490 nm was measured with a

2.8. TBARS method microplate reader (Bio-Rad Laboratories Ltd., China). Cell via-

bility was expressed as a percentage of the control (untreated

TBA method was used to measure the lipid peroxidation level cells). The relative cell viability was calculated according to

␮ [23]. Reacting 10% trichloroacetic acid 100 L and 0.8% thio- formula (3):

barbituric acid 100 L with the samples in 100 C for 15 min.

= A /A ×

The reaction mixture was centrifuged for 10 min at 5000 r/min Relative cell viability (%) 1 0 100 (3)

and the absorbance of the supernatant was measured at 532 nm

where A1 is the absorbance of the experimental group, and A0

using a UV–vis spectrophotometer. The inhibition of CA and its

is the absorbance of the control group.

metabolites on lipid peroxidation induced by free radicals were

calculated according to formula (2):

2.13. Measurement of NO production

inhibition rate(%) = (1 − (A1 − A2) /A 0) × 100 (2)

The production of NO was determined with Griess reagent

[26]. 50 ␮L samples or standards were added into 96-well plates,

where A1 is the absorbance of the experimental group, A2 is the

which were incubated with the same volume of the Griess

absorbance of the induced group (using phosphate buffer instead

reagent [0.1% (w/v) N-(1-naphathyl)-ethylenediamine and 1%

of FeSO4 solution), and A0 is the absorbance of the control group ◦

(w/v) sulfanilamide in 5% (v/v) phosphoric acid] at 37 C. Ten

(using phosphate buffer instead of the sample).

minutes later, the absorbance at 540 nm was measured using a

microplate reader. The amount of nitrite was calculated by using

2.9. hsDNA oxidative damage a sodium nitrite standard curve.

CA, CFA, and CTA (10, 50, 100, 500, 1000 mol/L) were 2.14. Measurement of ROS

mixed with 2.0 mg/mL hsDNA. 40 mmol/L AAPH was added

to induce DNA oxidative damage. After incubation at 37 C for DCFH-DA fluorescence dye itself has no fluorescence, and

12 h, the degree of DNA oxidative damage was measured by can go through the cell membrane freely. However, it can be oxi-

TBA method. dized into DCF which has fluorescence by intracellular reactive

158 Q. Liu et al. / Food Science and Human Wellness 6 (2017) 155–166

Fig. 1. Effects of CA and its metabolites on protein oxidative damage. (a–d) Effects of CA, CTA and CFA on protein oxidative degradation induced by hydroxyl

2+

radicals. BSA was incubated with Cu /H2O2 in the presence or absence of CA, CTA and CFA, as described in the Materials and Methods section; (e–h) Effects

of CA, CTA and CFA on protein oxidative degradation induced by alcoxyl radicals. BSA was incubated with AAPH in the presence or absence of CA, CTA and

CFA, as described in the Materials and Methods section. The protein contents were measured by commasie brilliant blue staining; (i–l) Effects of CA, CTA and

2+

CFA on protein carbonylation induced by hydroxyl radicals. BSA was incubated with Cu /H2O2 in the presence or absence of CA, CTA and CFA, as described

in the Materials and Methods section; (m–p) Effects of CA, CTA and CFA on protein carbonylation induced by alcoxyl radicals. BSA was incubated with AAPH

in the presence or absence of CA, CTA and CFA, as described in the Materials and Methods section. The carbonyl content was determined using a colorimetric

method. (q–t) Effects of CA and its metabolites on protein nitration. BSA was incubated with hemin/nitrite/H2O2 in the presence or absence of CA, CTA and CFA,

##

as described in the Materials and Methods section; The levels of nitration were measured by western blotting. Each value represents the mean ± SD (n ≥ 3),

**

represents significant differences compared to the control group (p < 0.01); * represents significant differences compared to the induced group (p < 0.05); represents

 

significant differences compared to the induced group (p < 0.01); represents significant differences compared to the same concentration of CA (p < 0.05);

represents significant differences compared to the same concentration of CA (p < 0.01).

oxygen species. Therefore, the fluorescence intensity can reflect 3. Results

the level of ROS [27].

BV2 microglial cells were washed with PBS 2–3 times after 3.1. Effects of CA, CTA and CFA on protein oxidative

treated, then DCFH-DA dyes diluted by serum free medium degradation

were added, after 37 C incubation for 30 min, inverted fluores-

cence microscope was used to observe the generation of ROS Excessive generation of ROS will induce protein oxidative

and Multiscan Spectrum to make quantitative determinations of damage. And degradation and carbonylation are the main char-

2+

intracellular ROS. acteristics of protein oxidation [28]. Cu /H2O2 and AAPH

reaction systems were adopted to detect the effects of CA and

its metabolites on BSA oxidative damage induced by these two

different kinds of free radicals.

2.15. Statistical analysis

2+

Fig. 1(a–d) showed that after Cu /H2O2 treatment, protein

bands were notably weakened compared to the control group,

All the experiments were performed with at least three repli-

2+

indicating hydroxyl free radicals produced by Cu /H O sys-

cations, and all the data were presented as the mean ± standard 2 2

tem substantially destroyed the structure of BSA. Pretreatment

errors (SD). Significant differences were analyzed using the

of CA and CTA increased the content of residual BSA in a con-

one-way analysis of variance (ANOVA) followed by Tukey’s

centration dependent manner, which demonstrated that CA and

multiple-range test with the SPSS 18.0 system.

Q. Liu et al. / Food Science and Human Wellness 6 (2017) 155–166 159

Fig. 2. Effects of CA and its metabolites on lipid peroxidation. (a-d) Effects of CA, CTA and CFA on liposomes peroxidation induced by hydroxyl radicals. Lecithin

2+

liposomes were incubated with Fe in the absence or presence of CA, CTA and CFA, as described in the Materials and Methods section; (e–h) Effects of CA, CTA

2+

and CFA on linoleic acid peroxidation induced by hydroxyl radicals. Linoleic acid was incubated with Fe in the absence or presence of CA, CTA and CFA, as

##

described in the Materials and Methods section. The amount of TBARS formed was determined by colorimetry. Each value represents the mean ± SD (n ≥ 3),

**

represents significant differences compared to the control group (p < 0.01); * represents significant differences compared to the induced group (p < 0.05); represents



significant differences compared to the induced group (p < 0.01); represents significant differences compared to the same concentration of CA (p < 0.01).

160 Q. Liu et al. / Food Science and Human Wellness 6 (2017) 155–166

CTA had strong abilities to protect BSA protein from hydroxyl dation concentration dependently. The ability of 100 ␮mol/L CA

radicals induced degradation. However, the optical density of to inhibit liposome peroxidation was 55.60%, which was 2.81

protein bands in CFA groups decreased. The degradation inhi- times than that of CFA. However, it had no significant difference

bition rate of 1 mmol/L CA was 60.95%, which was 1.78 times compared with CTA (p < 0.05).

than that of CTA. Moreover, Fig. 2(e–h) exhibited a concentration-dependent

AAPH system producing alcoxyl radicals was also employed inhibition in linoleic acid peroxidation when treated with CA

to investigate the effects of CA and its metabolites on BSA and its metabolites, which were consistent with liposome per-

oxidative degradation (Fig. 1(e–h)). CA and its metabolites sup- oxidation system. The inhibition rates of CA at 1 mmol/L was

pressed BSA degradation in concentration-dependent manners. 1.54 and 0.78 times than those of CTA and CFA, respec-

The degradation inhibition rates of CA, CTA and CFA were tively.

79.74%, 55.00% and 66.88% at 100 ␮mol/L, respectively (Sup-

plementary Fig. 1). 3.5. Effects of CA, CTA and CFA on DNA oxidative

damages

3.2. Effects of CA, CTA and CFA on protein carbonylation

DNA is the main attack target of free radicals. Oxidative

2+

As illustrated in Fig. 1(i–l), Cu /H2O2 dramatically stress leads to DNA damages such as DNA base modifica-

increased BSA carbonylation. Within the concentration range tions, double chain rupture, and base mutation. AAPH thermal

of 10–1000 ␮mol/L, CA and its metabolites all exerted remark- decomposition generates alcoxyl radicals, which attack DNA

able inhibition capacities. Besides, the effects were strengthened and the degraded product of DNA (reactive carbonyl com-

along with the increase of the concentrations of CA and CTA. pounds) will react with thiobarbituric acid under acidic condition

However, CFA promoted carbonylation in 10–1000 ␮mol/L, to produce TBARS, which has a significant absorption peak at

which is consistent with the protein degradation at same con- 532 nm. As demonstrated in Fig. 3(a–d), the absorbance of DNA

centration. The carbonylation suppression rate of 1 mmol/L CA reaction solution at 532 nm significantly (p < 0.01) increased

was 37.36% higher than CFA, which had statistically significant after treatment with 40 mmol/L AAPH for 12 h at 37 C. The

difference (p < 0.01). absorbance of CA and its metabolites treatment group were

Fig. 1(m–p) showed that the level of BSA carbonylation obvi- significantly (p < 0.01) lower than AAPH-induced group, indi-

ously increased when treated with AAPH, while CA, CTA and cating that CA and its metabolites all had protective effects on

CFA all could reverse this trend. The results were consistent DNA. The degradation suppression rate of 100 ␮mol/L CA was

with those obtained in BSA degradation system. Carbonylation 72.45%, which was 22.32% and 16.50% statistically signifi-

inhibition rates of CA at 100 ␮mol/L was 51.46%, dramatically cant (p < 0.01) higher than those of CTA and CFA, respectively.

stronger than its metabolites. These results showed that the inhibiting effect of CA on her-

ring sperm DNA induced by alcoxyl radicals was significantly

3.3. Effects of CA, CTA and CFA on protein nitration (p < 0.01) stronger than its metabolites at same experimental

concentrations.

As shown in Fig. 1(q–t), treatment with hemin/nitrite/H2O2 Fig. 3(e–h) showed that supercoiled form of DNA unwound

reaction system for 60 min significantly induced the formation of and gradually disappeared after the treatment of 10 mmol/L

3-nitrotyrosine in BSA, which suggested obvious BSA protein AAPH, while control group was almost all consisted of

nitration. In a certain concentration range (10–1000 ␮mol/L), suprahelical helixes. Pretreatment with 5–100 ␮mol/L CA and

CA and its metabolites had significant dose-dependent inhibition its metabolites increased superhelical structure concentration

abilities on nitration induced by hemin/nitrite/H2O2. In addi- dependently, implying that CA and its metabolites all remark-

tion, 500 ␮mol/L CA and its metabolites pretreatment almost ably inhibited DNA oxidative cleavage caused by alcoxyl

completely restrained the formation of 3-nitrotyrosine. The sup- radicals. The content of supercoiled DNA was 82.73% when

pression rates of CA, CTA and CFA at 100 ␮mol/L were 83.51%, treated with 100 ␮mol/L CA, which was significantly (p < 0.01)

36.09% and 43.99%, respectively (Supplementary Fig. 2). higher than its metabolites CTA and CFA. The result demon-

Results indicated the stronger inhibiting capacity of CA on strated that the inhibiting effect of CA on pBR322 plasmid DNA

hemin/nitrite/H2O2-induced BSA nitration (p < 0.01) (Supple- breakage induced by alcoxyl radicals was significantly (p < 0.01)

mentary Fig. 2). stronger than its metabolites.

3.4. Effects of CA, CTA and CFA on lipid peroxidation 3.6. Effects of CA, CTA and CFA on BV2 cell viability

induced by lipopolysaccharide

Lipid peroxidation product such as malondialdehyde can

react with thiobarbituric acid to generate TBARS, which is an Chronic inflammation plays a pivotal role in pathogenesis of

important index in measuring the degree of lipid peroxidation. neurodegenerative diseases such as Alzheimer’s disease, Parkin-

Lipid had a slow process of auto-oxidation in the absence of son’s disease and multiple sclerosis. As illustrated in Fig. 4, CA

2+

Fe , and the content of TBARS in induced group increased sig- and its metabolites themselves had no influence on cell viability.

nificantly (p < 0.01) compared with control group (Fig. 2(a–d)). After treated with lipopolysaccharide (1 ␮g/mL) for 12 h, via-

CA and its metabolites all inhibited the lecithin liposome peroxi- bility of BV2 cells significantly decreased to 81.58% compared

Q. Liu et al. / Food Science and Human Wellness 6 (2017) 155–166 161

Fig. 3. Effects of CA and its metabolites on DNA oxidative damage. (a–d) Effects of CA, CTA and CFA on hsDNA oxidation induced by alcoxyl radicals. hsDNA

was incubated with AAPH in the presence or absence of CA, CTA and CFA, as described in the Materials and Methods section. The degree of DNA oxidative damage

was measured by TBA method; (e–h) Effects of CA, CTA and CFA on pBR322 plasmid DNA oxidative cleavage induced by alcoxyl radicals. pBR322 plasmid

DNA was incubated with AAPH in the presence or absence of CA, CTA and CFA, as described in the Materials and Methods section. The levels of DNA oxidative

##

cleavage were tested by agarose gel electrophoresis, and the optical densities were quantified by Quantity One 4.6.2. Each value represents the mean ± SD (n ≥ 3),

**

represents significant differences compared to the control group (p < 0.01); * represents significant differences compared to the induced group (p < 0.05); represents



significant differences compared to the induced group (p < 0.01); represents significant differences compared to the same concentration of CA (p < 0.01).

162 Q. Liu et al. / Food Science and Human Wellness 6 (2017) 155–166

Fig. 4. Effect of CA and its metabolites on lipopolysaccharide −induced BV2 cell viability. BV2 cells were treated with lipopolysaccharide (1 ␮g/mL) 12 h with

##

or without of pretreatment of CA, CTA and CFA for 4 h. Cell viability was measured by MTT assay. Each value represents the mean ± SD (n ≥ 8), represents

**

significant differences compared to the control group (p < 0.01); * represents significant differences compared to the induced group (p < 0.05); represents significant

differences compared to the induced group (p < 0.01).

to control group. However, pretreatment of various concentra- 4. Discussion

tion of CA and its metabolites improved cell viability compared

to lipopolysaccharide treatment group. After pretreatment with Various studies have confirmed that phytochemicals, espe-

80 mol/L CA, CTA and CFA, cell viability elevated back to cially plant phenols could prevent or alleviate oxidative stress

88.84%, 83.64%, and 84.24%, respectively. in diseases [29]. In this study, antioxidant effects of CA and

its metabolites on the protection of biological macromolecules

in vitro and inflammatory responses in microglial cells were

3.7. Effects of CA, CTA and CFA on the production of NO investigated. Excessive ROS/RNS will attack biological macro-

induced by lipopolysaccharide molecules such as proteins, lipids and DNA, disrupt cell

structure and functions.

NO, an inflammatory product, results in biomacromolecule Recent studies have found that many diseases such as

damage and aggravates inflammatory process. Fig. 5 showed that Alzheimer’s, diabetes, cardiovascular disease are associated

CA and its metabolites had no effects on the production of NO with elevated levels of protein carbonylation [30–32]. Protein

in BV2 cells. However, 20–80 ␮mol/L CA and its metabolites tyrosine nitration has been identified as an important selective

remarkably reduced the increased production of NO induced post-translational modification, and is vital in the initiation and

by lipopolysaccharide in concentration-depended manners. The aggravation in cardiovascular diseases, ischemia/reperfusion

NO inhibition rates of CA, CTA and CFA at 80 ␮mol/L were injury, inflammation, and neurodegenerative disorder. CA, CFA

11.14%, 10.25% and 9.60%, respectively. and CTA all significantly inhibited protein degradation and car-

bonylation induced by hydroxyl radicals and alcoxyl radicals,

suppressed the RNS-induced nitration in the experimental con-

3.8. Effects of CA, CTA and CFA on the production of ROS centrations (10–1000 mol/L). And the protective effects of

induced by lipopolysaccharide CA were markedly stronger than its metabolites. It should be

noted that CFA promoted oxidation in high concentration range

3+

ROS is mainly produced by mitochondria and NADPH oxi- induced by hydroxyl radicals, may due to the reduction of Fe to

2+ 2+

dase located on the cell membrane. Excessive ROS can trigger a Fe , and the reaction between Fe and H2O2 forming hydroxyl

variety of signaling pathways such as NF␬B and MAPK, lead- radicals [33].

ing to cell proliferation, differentiate, or death. As demonstrated Lipid peroxidation readily occurs in mitochondria, cell mem-

in Fig. 6, lipopolysaccharide treatment induced elevated ROS brane, endoplasmic reticulum, lysosomes, perioxisomes and

levels in BV2 cells. As expected, CA significantly (p < 0.01) other membrane structures that contain rich polyunsaturated

reduced the ROS levels induced by lipopolysaccharide, which fatty acids. In the process of lipid peroxidation, a large num-

was 7.93 and 2.25 times than the effects of CTA and CFA. ber of toxicities including malondialdehyde, acraldehyde and

Q. Liu et al. / Food Science and Human Wellness 6 (2017) 155–166 163

Fig. 5. Effect of CA and its metabolites on lipopolysaccharide-induced NO production. BV2 cells were treated with lipopolysaccharide (1 ␮g/mL) 12 h with or

without of pretreatment of CA, CTA and CFA for 4 h. The accumulation of nitrite was measured to assess the NO production in BV2 cells. Each value represents the

##

mean ± SD (n ≥ 8), represents significant differences compared to the control group (p < 0.01); * represents significant differences compared to the induced group

**

(p < 0.05); represents significant differences compared to the induced group (p < 0.01).

4-hydroxynonenal are produced [34]. Linoleic acid, an essential nervous system, can secrete cytokines such as NO, TNF-␣,

fatty acid containing two easily oxidized double bonds, is an and IL-1β [40]. Lipopolysaccharide is an efficient activator

ideal material for studying lipid peroxidation [35]. The polyun- of microglial cells and it can increase the production of ROS

saturated fatty acids of very low density lipoprotein and low and pro-inflammatory cytokines, which damage the surrounding

density lipoprotein located in the C-2 of lecithin produce perox- nerve cells [41]. Lipopolysaccharide activates receptor ser-

2+

idation under the catalysis of Fe . So lecithin is usually used as a ine/threonine kinases to trigger the phosphorylation of a series

cell model in vitro. This study indicated that the inhibition of CA protein kinase through specific receptor CD14 and TLR4 on the

2+

on Fe -induced linoleic acid and soybean lecithin liposomes cell membrane, and it also activates p38MAPK and NF␬B sig-

peroxidation were remarkably stronger than its metabolites. It naling pathways to participate in the process of iNOS expression,

is also worthy of noting that CFA had a better protective effect which promotes the production of NO [42]. NO, as an intra-

of linoleic acid in comparison with CA. cellular messenger molecule, takes part in vascular regulation,

DNA oxidative damage caused by radicals attack is closely neurotransmitter, immune responses and other physiological

related to a variety of diseases such as inflammation, tumor, processes [43]. However, excess NO will react with superox-

•− −

cardiovascular disease and aging. In this work, CA and its ide anion (O2 ) to generate peroxynitrite (ONOO ), which

metabolites showed a concentration-dependent inhibition on the decomposes to generate a potent oxidant similar to HO in

oxidation of hsDNA and protection for pBR322 plasmid DNA reactivity, and it may cross cell membranes through anion

from breakage induced by alcoxyl radicals. Protective effects of channels [44]. ONOO induces protein nitration, lipid peroxida-

CA were statistically significant better than its metabolites. It has tion, and DNA breakage [45]. Increased formation of ONOO

been demonstrated that in mango wine could pro- has been linked to Alzheimer’s disease, rheumatoid arthritis,

tect DNA against UV + H2O2 and ␥-irradiation induced DNA atherosclerosis, lung injury, amyotrophic lateral sclerosis, and

damage [36]. (+)-catechin hydrate, (−)-epigallocatechin gallate other diseases [46,47]. This study demonstrated that both CA

hydrate, morin hydrate, quercetin hydrate and resveratrol have and its metabolites significantly increased BV2 microglial cell

potential for protecting both the viability of cells and ability viability and inhibited the production of NO and ROS induced by

to proliferate from damage caused by UV-A-irradiation [37]. lipopolysaccharide. As noted, the inhibition of CA on the ROS

Green tea (Camellia sinensis) strongly reversed apoptotic hep- levels induced by lipopolysaccharide was stronger than CTA and

atic degeneration and prevented micronecrosis and mutagenic CFA, which was consistent with the scavenging of free radicals

• • +•

DNA damage induced by arsenic [38]. including DPPH , OH, ABTS and biological macromolecu-

Chronic inflammation in the brain is one of the most impor- lar protection effects in vitro. Whereas CA, CTA and CFA had

tant pathological features of neural degenerative diseases such no significant difference in NO levels and cell viability. Since

as Alzheimer’s, Parkinson’s disease and multiple sclerosis [39]. DCFH-DA assay was used to detect overall intracellular redox,

Microglia, the most representative immune cells in the central cells incubation with compounds with more hydroxyl groups is

164 Q. Liu et al. / Food Science and Human Wellness 6 (2017) 155–166

Fig. 6. Effect of CA and its metabolites on lipopolysaccharide −induced ROS production. BV2 cells were treated with lipopolysaccharide (1 ␮g/mL) 12 h with or

without of pretreatment of CA, CTA and CFA for 4 h. The production of ROS was monitored with DCFH-DA fluorescence dye. (a) using inverted fluorescence

microscope to observe the generation of ROS; (b) using multiscan spectrum to make quantitative determinations of intracellular ROS. Each value represents the

## **

mean ± SD (n ≥ 8), represents significant differences compared to the control group (p < 0.01); represents significant differences compared to the induced group



(p < 0.01); represents significant differences compared to the same concentration of CA (p < 0.01).

likely to have a lower DCF fluorescence, which may partly con- ings will provide scientific bases for the application of CA and

tributed to the more powerful inhibitory effects of CA. There its metabolites as nutrition and natural antioxidants.

are structure-activity relationships of polyphenols to inhibit-

ing lipopolysaccharide-induced inflammation [48]. However, Conflict of interest

the molecular mechanism remains to be further investigated.

The authors declare that there are no conflicts of interest.

5. Conclusion

Acknowledgement

In this study, effects of CA and its metabolites on in vitro

This work was supported by the National Key Research and

protection of biological macromolecules and inflammatory

Development Program of China (No. 2016YFD0400601) and

responses in microglial cell were investigated. Results demon-

the Science and Technology Coordination Project of Innovation

strated that CA and its metabolites all exerted remarkable

in Shaanxi province (NO. 2014KTCL02-07).

inhibition capacities to biological macromolecules damages

including protein, lipid, and DNA induced by different kinds

Appendix A. Supplementary data

of free radicals. In addition, CA and its metabolites suppressed

the decline of BV2 microglia cell viability and the production

of NO and ROS induced by lipopolysaccharide. Furthermore, Supplementary data associated with this article can be

found, bioactivities of CA were significantly stronger than those of its in the online version, at http://dx.doi.org/10.1016/

j.fshw.2017.09.001 metabolites within a certain concentration range. These find- .

Q. Liu et al. / Food Science and Human Wellness 6 (2017) 155–166 165

References [20] N.H. Lu, M.Y. Zhang, H.L. Li, Z.H. Gao, Completely different effects

of desferrioxamine on hemin/nitrite/H2O2-induced bovine serum albumin

nitration and oxidation, Chem. Res. Toxicol. 21 (2008) 1229–1234.

[1] I. Afanas’ev, Mechanisms of superoxide signaling in epigenetic processes:

[21] A. Mavi, G.D. Lawrence, S¸. Kordali, A.L.I. Yildirim, Inhibition of iron-

relation to aging and cancer, Aging Dis. 6 (2015) 216–227.

fructose-phosphate-Induced lipid peroxidation in lecithin liposome and

[2] F. Santilli, M. Guagnano, N. Vazzana, S. La Barba, G. Davi, Oxidative

linoleic acid emulsion systems by some edible plants, J. Food Biochem. 35

stress drivers and modulators in obesity and cardiovascular disease: from

(2011) 833–844.

biomarkers to therapeutic approach, Curr. Med. Chem. 22 (2015) 582–595.

[22] I.C. Vlachogianni, E. Fragopoulou, I.K. Kostakis, S. Antonopoulou, In vitro

[3] H. Bartsch, J. Nair, Chronic inflammation and oxidative stress in the genesis

assessment of antioxidant activity of tyrosol, resveratrol and their acetylated

and perpetuation of cancer: role of lipid peroxidation, DNA damage, and

derivatives, Food Chem. 177 (2015) 165–173.

repair, Langenbecks Arch. Surg. 391 (2006) 499–510.

[23] S.H. Helmer, A. Kerbaol, P. Aras, C. Jumarie, M. Boily, Effects of realistic

[4] Y. Zhang, Y. Du, W. Le, K. Wang, N. Kieffer, J. Zhang, Redox control

doses of atrazine metolachlor, and glyphosate on lipid peroxidation and

of the survival of healthy and diseased cells, Antioxid. Redox Signal. 15

diet-derived antioxidants in caged honey bees (Apis mellifera), Environ.

(2011) 2867–2908.

Sci. Pollut. Res. Int. 22 (2015) 8010–8021.

[5] N. Chaudhari, P. Talwar, A. Parimisetty, C. Lefebvre d’Hellencourt, P.

[24] K. Sevgi, B. Tepe, C. Sarikurkcu, Antioxidant and DNA damage protection

Ravanan, A molecular web: endoplasmic reticulum stress, inflammation,

potentials of selected phenolic acids, Food Chem. Toxicol. 77 (2015) 12–21.

and oxidative stress, Front. Cell. Neurosci. 8 (2014) 213.

[25] W. Wu, Y. Li, Y. Wu, Y. Zhang, Z. Wang, X. Liu, Lutein suppresses inflam-

[6] B. Fubini, A. Hubbard, Reactive oxygen species (ROS) and reactive nitro-

matory responses through Nrf2 activation and NF-kappaB inactivation in

gen species (RNS) generation by silica in inflammation and fibrosis, Free

lipopolysaccharide-stimulated BV-2 microglia, Mol. Nutr. Food Res. 59

Rad. Biol. Med. 34 (2003) 1507–1516.

(2015) 1663–1673.

[7] M.S. Costamagna, I.C. Zampini, M.R. Alberto, S. Cuello, S. Torres,

[26] I. Guevara, J. Iwanejko, A. Dembinska-Kiec, J. Pankiewicz, A. Wanat, P.

J. Perez, C. Quispe, G. Schmeda-Hirschmann, M.I. Isla, Polyphenols

Anna, I. Golabek, S. Bartus, M. Malczewska-Malec, A. Szczudlik, Deter-

rich fraction from Geoffroea decorticans fruits flour affects key enzymes

mination of nitrite/nitrate in human biological material by the simple Griess

involved in metabolic syndrome, oxidative stress and inflammatory process,

reaction, Clin. Chim. Acta 274 (1998) 177–188.

Food Chem. 190 (2016) 392–402.

[27] Q.S. Wang, Y.M. Zheng, L. Dong, Y.S. Ho, Z.M. Guo, Y.X. Wang, Role

[8] J.H. Kim, Q. Wang, J.M. Choi, S. Lee, E.J. Cho, Protective role of caffeic

of mitochondrial reactive oxygen species in hypoxia-dependent increase

acid in an Abeta25-35-induced Alzheimer’s disease model, Nutr. Res. Pract.

in intracellular calcium in pulmonary artery myocytes, Free Radic. Biol.

9 (2015) 480–488.

Med. 42 (2007) 642–653.

[9] L. Yuan, Y. Wu, X. Ren, Q. Liu, J. Wang, X. Liu, Isoorientin attenuates

[28] I. Dalle-Donne, R. Rossi, D. Giustarini, A. Milzani, R. Colombo, Protein

lipopolysaccharide-induced pro-inflammatory responses through down-

carbonyl groups as biomarkers of oxidative stress, Clin. Chim. Acta 329

regulation of ROS-related MAPK/NF-kappaB signaling pathway in BV-2

(2003) 23–38.

microglia, Mol. Cell. Biochem. 386 (2014) 153–165.

[29] H.Y. Aboul-Enein, P. Berczynsk, I. Kruk, Phenolic compounds: the role of

[10] E.M. Saad, A. Madbouly, N. Ayoub, R.M. El Nashar, Preparation and appli-

redox regulation in neurodegenerative disease and cancer, Mini Rev. Med.

cation of molecularly imprinted polymer for isolation of chicoric acid from

Chem. 13 (2013) 385–398.

Chicorium intybus L. medicinal plant, Anal. Chim. Acta 877 (2014) 80–89.

[30] M.A. Baraibar, R. Ladouce, B. Friguet, Proteomic quantification and iden-

[11] P.M. Flanigan, E.D. Niemeyer, Effect of cultivar on phenolic levels, antho-

tification of carbonylated proteins upon oxidative stress and during cellular

cyanin composition, and antioxidant properties in purple basil (Ocimum

aging, J. Proteom. 92 (2013) 63–70.

basilicum L.), Food Chem. 164 (2014) 518–526.

[31] R.L. Levine, Carbonyl modified proteins in cellular regulation aging, and

[12] F. Mai, M.A. Glomb, Isolation of phenolic compounds from iceberg let-

disease, Free Radic. Biol. Med. 32 (2002) 790–796.

tuce and impact on enzymatic browning, J. Agric. Food Chem. 61 (2013)

2868–2874. [32] K. Mukherjee, T.I. Chio, D.L. Sackett, S.L. Bane, Detection of oxidative

stress-induced carbonylation in live mammalian cells, Free Radic. Biol.

[13] H. Xiao, J. Wang, L. Yuan, C. Xiao, Y. Wang, X. Liu, Chicoric acid induces

Med. 84 (2015) 11–21.

apoptosis in 3T3-L1 preadipocytes through ROS-mediated PI3K/Akt and

[33] O.I. Aruoma, A. Murcia, J. Butler, B. Halliwell, Evaluation of the antioxi-

MAPK signaling pathways, J. Agric. Food Chem. 61 (2013) 1509–1520.

dant and prooxidant actions of gallic acid and its derivatives, J. Agric. Food

[14] H. Xiao, G. Xie, J. Wang, X. Hou, X. Wang, W. Wu, X. Liu, Chicoric

Chem. 41 (1993) 1880–1885.

acid prevents obesity by attenuating hepatic steatosis, inflammation and

[34] Y. Yoshida, A. Umeno, Y. Akazawa, M. Shichiri, K. Murotomi, M. Horie,

oxidative stress in high-fat diet-fed mice, Food Res. Int. 54 (1) (2013)

345–353. Chemistry of lipid peroxidation products and their use as biomarkers in

early detection of diseases, J. Oleo. Sci. 64 (2015) 347–356.

[15] H.L. Zhang, L.H. Dai, Y.H. Wu, X.P. Yu, Y.Y. Zhang, R.F. Guan, T. Liu, J.

[35] A. Catala, A synopsis of the process of lipid peroxidation since the dis-

Zhao, Evaluation of hepatocyteprotective and anti-hepatitis B virus prop-

covery of the essential fatty acids, Biochem. Biophys. Res. Commun. 399

erties of cichoric acid from cichorium intybus leaves in cell culture, Biol.

(2010) 318–323.

Pharm. Bull. 37 (2014) 1214–1220.

[36] N. Kondapalli, V. Sadineni, P.S. Variyar, A. Sharma, V.S.R. Obulam,

[16] Q. Liu, Y. Wang, C. Xiao, W. Wu, X. Liu, Metabolism of chicoric acid by

Impact of ␥-irradiation on antioxidant capacity of mango (Mangifera indica

rat liver microsomes and bioactivity comparisons of chicoric acid and its

L.) wine from eight Indian cultivars and the protection of mango wine

metabolites, Food Funct. 6 (2015) 1928–1935.

against DNA damage caused by irradiation, Process Biochem. 49 (2014)

[17] S. Mohan, K. Thiagarajan, R. Chandrasekaran, J. Arul, In vitro protection

1819–1830.

of biological macromolecules against oxidative stress and in vivo toxicity

[37] A. Shirai, M. Onitsuka, H. Maseda, T. Omasa, Effect of polyphenols on

evaluation of Acacia nilotica (L.) and ethyl gallate in rats, BMC Complem.

reactive oxygen species production and cell growth of human dermal fibrob-

Altern. Med. 14 (2014) 257.

lasts after irradiation with ultraviolet-a light, Biocontrol Sci. 20 (2015)

[18] J.C. Mayo, D.X. Tan, R.M. Sainz, M. Natarajan, S. Lopez-Burillo,

27–33.

R.J. Reiter, Protection against oxidative protein damage induced by

[38] N. Acharyya, S. Chattopadhyay, S. Maiti, Chemoprevention against

metal-catalyzed reaction or alkylperoxyl radicals: comparative effects of

arsenic-Induced mutagenic DNA Breakage and apoptotic liver damage in

melatonin and other antioxidants, Biochim. Biophys. Acta 1620 (2003)

139–150. rat via antioxidant and SOD1 upregulation by green tea (Camellia sinensis)

which recovers broken DNA resulted from arsenic-H2O2 related in vitro

[19] P. Stocker, E. Ricquebourg, N. Vidal, C. Villard, D. Lafitte, L. Sellami,

oxidant stress, J. Environ. Sci. Health C. Environ. Carcinog. Ecotoxicol.

S. Pietri, Fluorimetric screening assay for protein carbonyl evaluation in

Rev. 32 (2014) 338–361.

biological samples, Anal. Biochem. 482 (2015) 55–61.

166 Q. Liu et al. / Food Science and Human Wellness 6 (2017) 155–166

[39] J.H. Woo, J.H. Lee, H. Kim, S.J. Park, E.H. Joe, I. Jou, Control of inflam- [44] W.A. Pryor, G.L. Squadrito, The chemistry of peroxynitrite: a product from

matory responses: a new paradigm for the treatment of chronic neuronal the reaction of nitric oxide with superoxide, Am. J. Physiol. 268 (1995)

diseases, Exp. Neurobiol. 24 (2015) 95–102. L699–722.

[40] M.A. Mena, J. Garcia de Yebenes, Glial cells as players in parkinsonism: the [45] J.S. Beckman, Oxidative damage and tyrosine nitration from peroxynitrite,

good, the “bad, and the mysterious” glia, Neuroscientist (2008) 544–560. Chem. Res. Toxicol. 9 (1996) 836–844.

[41] S. Abbasi Habashi, F. Sabouni, A. Moghimi, S. Ansari Majd, Modulation of [46] G.L. Squadrito, W.A. Pryor, Oxidative chemistry of nitric oxide: the roles

lipopolysaccharide stimulated nuclear factor kappa B mediated iNOS/NO of superoxide, peroxynitrite, and carbon dioxide, Free Radic. Bio. Med. 25

production by bromelain in rat primary microglial cells, Iran. Biomed. J. (1998) 392–403.

20 (2016) 33–40. [47] M.A. Smith, P.L.R. Harris, L.M. Sayre, J.S. Beckman, G. Perry, Widespread

[42] K.W. Zeng, M.B. Zhao, Z.Z. Ma, Y. Jiang, P.F. Tu, Protosappanin peroxynitrite-mediated damage in Alzheimer’s disease, J. Neurosci 17

A inhibits oxidative and nitrative stress via interfering the inter- (1997) 2653–2657.

action of transmembrane protein CD14 with Toll-like receptor-4 in [48] S.Y. Shin, S. Ahn, M.J. Park, H. Yoon, M. Kim, S.Y. Ji, D. Koh, Y.H.

lipopolysaccharide-induced BV-2 microglia, Int. Immunopharmacol. 14 Lee, Y. Lim, Structure-activity relationships of polyphenols inhibiting

(2012) 558–569. lipopolysaccharide-induced NF-kappa B activation, J. Korean Soc. Appl.

[43] B. Olas, Gasomediators (.NO CO, and H(2)S) and their role in hemostasis Biol. 55 (2012) 669–675.

and thrombosis, Clin. Chim. Acta 445 (2015) 115–121.