astin b

Upload: ceciliapistol

Post on 02-Jun-2018

222 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/10/2019 Astin B

    1/9

    Astin B, a cyclic pentapeptide from Aster tataricus, induces apoptosis

    and autophagy in human hepatic L-02 cells

    Li Wang, Ming-Dan Li, Pei-Pei Cao, Chao-Feng Zhang , Fang Huang, Xiang-Hong Xu,Bao-Lin Liu, Mian Zhang

    Research Department of Pharmacognosy, China Pharmaceutical University, Longmian Road 639, Nanjing 211198, PR China

    a r t i c l e i n f o

    Article history:

    Received 15 February 2014

    Received in revised form 21 August 2014

    Accepted 4 September 2014

    Available online 16 September 2014

    Keywords:

    Astin B

    Liver injury

    Apoptosis

    Autophagy

    Aster tataricus

    a b s t r a c t

    Astins (including astin B) are a class of halogenated cyclic pentapeptides isolated from the medicinal herb

    ofAster tataricus. However, our previous works showed that the herbal medicine was hepatotoxic in vivo,

    and a toxicity-guided isolation method led to the identification of a cyclopeptide astin B. Astin B is struc-

    turally similar to cyclochlorotine, a well-known hepatotoxic mycotoxin. Thus, the aim of this study was

    to determine the potential cytotoxic effects and the underlying mechanism of astin B on human normal

    liver L-02 cells. We found that astin B has hepatotoxic effects in vitro and in vivoand that hepatic injury

    was primarily mediated by apoptosis in a mitochondria/caspase-dependent manner. Astin B provoked

    oxidative stress-associated inflammation in hepatocytes as evidenced by increased levels of reactive oxy-

    gen species (ROS), reduced contents of intracellular glutathione (GSH), and enhanced phosphorylation of

    c-Jun N-terminal kinase (JNK). Furthermore, the mitochondria-dependent apoptosis was evidenced by

    the depolarization of the mitochondrial membrane potential, the release of cytochrome c into cytosol,

    the increased ratio of Bax/Bcl-2, and the increased activities of caspases-9 and -3. Interestingly, astin B

    treatment also induces autophagy in L-02 cells, characterized by acidic-vesicle fluorescence, increased

    LC3-II and decreased p62 expression. Autophagy is a protective mechanism that is used to protect cells

    from apoptosis. The presence of autophagy is further supported by the increased cytotoxicity and the

    enhanced cleaved caspase-3 after co-treatment of cells with an autophagy inhibitor, also by increasedLC3-II and decreased p62 after co-treatment with a caspase inhibitor. Taken together, astin B, most likely

    together with other members of astins, is the substance that is primarily responsible for the hepatotox-

    icity ofA. tataricus.

    2014 Elsevier Ireland Ltd. All rights reserved.

    1. Introduction

    The hepatocyte is especially vulnerable to injury due to its cen-

    tral role in xenobiotic metabolism including the metabolism of

    drugs. Therefore, drug-induced liver injury is the most frequent

    reason for post-marketing warnings and withdrawal[1]. The death

    of hepatocytes and other types of hepatic cells is a characteristic

    feature of drug-induced injury[2]. Based on morphological appear-

    ance, cell death has been classified into several modes such as

    apoptosis, necrosis, necroptosis, autophagy, and cornification [3].

    Of these, apoptosis is considered to be a common pathway for

    execution of hepatocytes upon liver injury. In response to xenobi-

    otic metabolism, hepatocytes often generate excess reactive

    oxygen species (ROS), which evoke oxidative stress-associated

    inflammation, leading to mitochondrial dysfunction [4,5].

    Mitochondria play a key role in the regulation of redox homeosta-

    sis and apoptosis in cells [6]. A loss of mitochondrial function

    allows the release of a number of proapoptotic factors, such as

    cytochrome c, which induces caspase activation to trigger

    mitochondria-dependent apoptotic cell death [7]. Accumulating

    evidence demonstrates that hepatocyte apoptosis is tightly associ-

    ated with drug-induced liver injury [6,8]. However, autophagy is

    understood to be a mechanism of protection against various forms

    of human diseases, including drug-induced liver injury, with an

    extremely complex interplay[9,10].

    Astins, mainly including astins AI, are a class of natural haloge-

    nated cyclic pentapeptides isolated from the root ofAster tataricus

    L. f. (RAT, Compositae) that has been used for over 2000 years in

    traditional Chinese medicine for the relief of coughs and the

    removal of phlegm. This class of compounds exhibits antitumor

    and immunosuppressive activities[1113]. However, our previous

    studies discovered the potent hepatotoxicity of RAT in mice, and a

    http://dx.doi.org/10.1016/j.cbi.2014.09.003

    0009-2797/ 2014 Elsevier Ireland Ltd. All rights reserved.

    Corresponding authors. Tel./fax: +86 25 86185137.

    E-mail addresses: [email protected] (C.-F. Zhang), [email protected]

    (M. Zhang).

    Chemico-Biological Interactions 223 (2014) 19

    Contents lists available at ScienceDirect

    Chemico-Biological Interactions

    j o u r n a l h o m e p a g e : w w w . e l s e v i e r . c o m / l o c a t e / c h e m b i o i n t

    http://dx.doi.org/10.1016/j.cbi.2014.09.003mailto:[email protected]:[email protected]://dx.doi.org/10.1016/j.cbi.2014.09.003http://www.sciencedirect.com/science/journal/00092797http://www.elsevier.com/locate/chembiointhttp://www.elsevier.com/locate/chembiointhttp://www.sciencedirect.com/science/journal/00092797http://dx.doi.org/10.1016/j.cbi.2014.09.003mailto:[email protected]:[email protected]://dx.doi.org/10.1016/j.cbi.2014.09.003http://-/?-http://-/?-http://-/?-http://-/?-http://crossmark.crossref.org/dialog/?doi=10.1016/j.cbi.2014.09.003&domain=pdfhttp://-/?-
  • 8/10/2019 Astin B

    2/9

    toxicity-guided isolation method led to the identification of an

    astin-rich fraction (71.2% of the relative content), from which a

    halogenated cyclic pentapeptide astin B (Fig. 1A) was given [14].

    Astin B is structurally similar to cyclochlorotine, a well-known

    hepatotoxic mycotoxin isolated from Penicillium islandicum [15],

    but the hepatotoxic effects and the underlying mechanisms wereunknown. In this study, we evaluated the effects of astin B on

    the modulation of cell death, apoptosis and autophagy in human

    normal liver L-02 cells. Our experiments indicate that astin B has

    marked toxic effects in vitro and in vivo and induces hepatic cell

    death mainly by apoptosis through a mitochondria/caspase-

    dependent pathway. Astin B also induces autophagy in L-02 cells,

    which appears to protect cells from apoptosis to some extent.

    2. Materials and methods

    2.1. Drugs and chemicals

    Astin B (Fig. 1A) was obtained from previous experiments [14]

    with a purity of more than 98%. Chlorogenic acid (CGA) wasobtained from Nanjing Zelang Medical Technology Company with

    a purity of 98% (Nanjing, China). For experiments in cells, solutions

    of astin B and CGA were prepared in DMSO and diluted to the

    desired concentrations in FBS-free medium. The DMSO concentra-

    tions in the experiments never exceeded 0.1%.

    Fetal bovine serum (FBS) was obtained from Hyclone (Thermo,

    South America). RPMI-1640 medium was obtained from Gibco

    BRL (NY, USA.). Earles balanced salts solution (EBSS), 3-methylad-

    enine (3-MA) and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetra-

    zolium bromide (MTT) were obtained from SigmaAldrich (St.

    Louis, USA.). Antibodies for c-Jun N-terminal kinase (JNK), phos-

    phorylated JNK (pJNK), Bax, Bcl-2, and caspase-3 were purchased

    from Cell Signal Technology Inc. (Massachusetts, USA). Anti-LC3

    antibody was obtained from MBL (Nagano, Japan). Anti-b-actinantibody and secondary anti-mouse and anti-rabbit antibodies

    were obtained from Lianke Biotechnology (Hangzhou, China). All

    of the other reagents were purchased from Amresco (Ohio, USA).

    2.2. Cell culture

    L-02 cells, a normal human liver cell line from the Cell Bank ofthe Chinese Academy of Sciences (Shanghai, China), were grown in

    RPMI-1640 medium supplemented with 10% FBS, 100 U/mL peni-

    cillin and 100 lg/mL streptomycin and incubated at 37C in ahumidified atmosphere (5% CO2). The medium was renewed every

    2 days until the cells were grown to confluence.

    2.3. Cytotoxicity assay

    L-02 cells were seeded at an initial density of 1 105 cells/mL

    in 96-well plates for 24 h and were incubated with fresh medium

    containing different concentrations of astin B for 12, 24, and

    48 h. After incubation, MTT was added to each well to reach a final

    concentration of 0.5 mg/mL. The insoluble formazan was collected

    and dissolved in DMSO and then measured using a microplatereader (Thermo, Finland) at a wavelength of 490 nm. For the assay

    of lactate dehydrogenase (LDH), the cells were incubated with

    astin B for 24 h. The supernatant was collected, and LDH activity

    was determined with a commercial kit (Jiancheng, Nanjing, China)

    in accordance with the manufacturers instructions.

    2.4. Measurement of intracellular ROS and GSH

    The generation of intracellular ROS was assessed using the

    ROS-specific fluorescent dye 2,7-dichlorofluorescein diacetate

    (DCFH-DA; Beyotime, Haimen, China). L-02 cells were seeded at

    an initial density of 1 105 cells/mL in 96-well plates. After 12 h

    exposure to astin B, the cells were washed with PBS, loaded with

    10 lmol/L DCFH-DA at 37 C for 30 min away from light, rinsedthree times with serum-free culture media, and measured at an

    0.3

    0.4

    0.5

    0.6

    0.7

    0.8

    0.9

    1

    1.1

    0 15 30 45 60

    Relativeviability

    Astin B (M)

    *

    *

    **

    **

    *

    **

    *

    B

    0

    20

    40

    60

    80

    100

    120

    140

    160

    180

    0 15 30 45 60

    LDHrelease(%)

    Astin B (M)

    **

    C

    NH

    NH

    NHN

    HN

    O

    O Cl

    ClO

    O

    OH

    O

    OH

    A

    Fig. 1. Effects of astin B on cell viability and lactate dehydrogenase (LDH) activity in L-02 cells. (A) Chemical structure of astin B. (B) Cells were treated with astin B for 12, 24,

    and 48 h, and cell viability was determined by the MTT assay. (C) Cells were treated with astin B for 24 h, and LDH leakage was measured using commercially available kits.

    Data in B and C are expressed as the mean SD from three independent experiments with n= 6 for MTT and n= 4 for LDH. p< 0.05 compared with the control group (0 lM).

    2 L. Wang et al. / Chemico-Biological Interactions 223 (2014) 19

  • 8/10/2019 Astin B

    3/9

    excitation wavelength of 488 nm and an emission wavelength of

    530 nm using a microplate fluorescence reader (MD, spectramax

    M3, USA). For GSH assay, the cells were cultured with astin B for

    24 h and then harvested by centrifuging. The intracellular GSH

    level was determined with commercially available kits (Jiancheng,

    Nanjing, China) in accordance with the manufacturers

    instructions.

    2.5. Analysis of mitochondrial membrane potential (DWm)

    The membrane potential assay is based on the JC-1 dye: JC-1

    emits green fluorescence when the DWm is relatively low. How-

    ever, JC-1 aggregates and emits a red fluorescence when the

    DWm is high [16]. The assay was performed with a JC-1 kit in

    accordance with the manufacturers instructions (Beyotime).

    Briefly, L-02 cells (1 105 cells/mL) in 6-well plates were sepa-

    rately treated with or without astin B for 24 h and were incubated

    with JC-1 staining solution (5 lg/mL) for 20 min at 37C in thedark. The cells were rinsed twice with JC-1 staining buffer and

    were then observed and photographed using an Olympus

    fluorescence microscope.

    2.6. Measurement of cytochrome c (cyt c) content

    After treatment with astin B for 24 h, the cells were lysed in

    preparation buffer (250 mM sucrose, 20 mM HEPES-KOH (pH

    7.4), 10 mM NaCl, 1.5 mM MgCl2, 1 mM EDTA, 1 mM EGTA,

    1 mM dithiothreitol, 2 lg/mL aprotinin and 1 mM phenylmethyl-sulfonyl fluoride) for 10 min on ice followed by centrifugation at

    10,000 rpm for 20 min at 4 C to separate the cytosolic and mito-

    chondrial fractions [17]. Protein concentrations were measured

    with a bicinchoninic acid protein assay kit (Beyotime). The con-

    tents of cyt c in the cytosol and mitochondria were determined

    by ELISA kits (R&D Systems, Minnesota, USA) in accordance with

    the manufacturers instructions.

    2.7. Analysis of caspase activity

    The activities of caspase-3 and caspase-9 were evaluated using

    caspase-3 and caspase-9 activity assay kits (Beyotime). After a

    24-h exposure, L-02 cells from 6-well plates were collected and

    rinsed with cold PBS and later lysed using lysis buffer for 1 h on

    ice. The cell lysates were centrifuged at 18,000 rpm for 10 min at

    4C. The assays were performed in 96-well plates by incubating

    10 lL of the cell lysate supernatant in 80lL reaction buffercontaining 10lL Ac-DEVD-pNA (for caspase-3) and 10lLAc-LEHD-pNA (for caspase-9). After further incubation at 37C

    for 4 h, the absorbance was measured using a microplate reader

    (Thermo, Finland) at a wavelength of 405 nm.

    2.8. Flow cytometric analysis of apoptotic cells

    2.8.1. Sub-G1 peak

    The apoptosis induced by astin B was determined using a cell

    cycle and apoptosis analysis kit (Beyotime) in accordance with

    the manufacturers instructions. In brief, approximately 1 105

    L-02 cells were cultured in 6-well plates and separately treated

    with or without astin B for 24 h. After treatment, the cells were col-

    lected and fixed with 70% ethanol at 4 C overnight, centrifuged at

    800 rpm for 5 min and stained with 500 lL of buffer, 25 lL of pro-pidium iodide (PI), and 10 lL of RNase A in the dark for 30 min at37C. Next, the cells were analyzed using a flow cytometer (Becton

    Dickinson, New Jersey, USA). The apoptotic cells with hypodiploid

    DNA content were measured by quantifying the sub-G1 peak in the

    cell cycle pattern. For each experiment, 10,000 events wererecorded per sample.

    2.8.2. Annexin-V/PI staining

    After 24 h treatment, the L-02 cells were gently trypsinized and

    washed once with PBS, centrifuged at 800 rpm for 5 min, and

    resuspended in 200 lL of binding buffer. After gentle pipetting,the cells were stained by 3 lL of Annexin-V-FITC (AV) and 3 lLof propidium iodide (PI) (BD Pharmingen, California, USA) for

    15 min at room temperature in the dark and analyzed using a flow

    cytometer (Becton Dickinson, New Jersey, USA). For each experi-ment, 10,000 events were recorded per sample.

    2.9. Acridine orange (AO) staining of autophagic cells

    Autophagic cells were detected by AO staining in accordance

    with published procedures [18]. In brief, approximately 1 105

    cells were seeded in 24-well plates and incubated with astin B

    for 12 h, washed with PBS, and colored with AO (1lg/mL)(Generay, Shanghai, China) at 37C for 1 min in the dark. The

    cells were then observed and photographed by fluorescence

    microscopy.

    2.10. Western blot analysis

    The L-02 cells (approximately 6 105 in number) were

    extracted with 100lL of lysate buffer, which consists of 1 MTrisHCl, 50% glycerine, 1.0% bromophenol blue, 10% SDS, andb-mercaptoethanol. After boiling for 10 min, the extracted samples

    were loaded at 20 lL/per lane, fractionated on 1215% trisglycineprecast gels, and then transferred to a PVDF membrane (Millipore,

    Billerica, USA). The proteins were probed with primary antibodies

    and HRP-labeled secondary antibodies and visualized using super

    ECL detection reagent (Beyotime).

    2.11. Animal experiments

    2.11.1. Animals

    Male ICR mice (68 weeks of age) were obtained from theLaboratory Animal Center of Nanjing Qinglongshan (Nanjing,

    China). They were maintained with free access to pellet food and

    water in plastic cages at 21 2 C and kept on a 12-h light/dark

    cycle. The care and treatment of these mice were in accordance

    with the Provisions and General Recommendations of the Chinese

    Experimental Animals Administration Legislation. This study was

    approved by the Animal Ethics Committee of the School of Chinese

    Materia Medica, China Pharmaceutical University.

    2.11.2. Hepatotoxicity in mice

    All of the mice were acclimatized for 3 days prior to experimen-

    tal procedures. Two groups of mice (10 per group) were adminis-

    tered daily with astin B (10 mg/kg, suspended in distilled water)

    or distilled water by gavage for 7 days. One hour after the lastadministration, the mice were killed by cervical dislocation after

    isoflurane inhalation, blood was collected from the orbital sinus

    and centrifuged at 12,000 rpm for 5 min at 4C to obtain serum

    for the test of alanine aminotransferase (ALT) and aspartate amino-

    transferase (AST) activities by commercial kits (Jiancheng, Nanjing,

    China). Liver tissue was excised and fixed in 10% phosphate-

    buffered formalin and embedded in paraffin. Sections were

    prepared and stained with hematoxylin and eosin (H&E) for histo-

    pathological analysis.

    2.12. Statistical analysis

    The data are expressed as the means SD (standard deviation)

    from at least three independent experiments. Statistical analyseswere performed using one-way analysis of variance (ANOVA)

    L. Wang et al./ Chemico-Biological Interactions 223 (2014) 19 3

    http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-
  • 8/10/2019 Astin B

    4/9

    followed by Students two-tailed t-test. Values of p< 0.05 were

    considered to be statistically significant.

    3. Results

    3.1. Astin B inhabits proliferation of L-02 cells

    The potential cytotoxic effect of astin B was investigated byincubating L-02 cells with astin B at different concentrations for

    12, 24, and 48 h. The results of the MTT assay showed that astin

    B decreased cell viability in a concentration-dependent and time-

    dependent manner (Fig. 1B). After 48 h treatment, the cell viability

    decreased to 39.22% at 60 lM. Leakage of LDH from cells is an indi-cator of cell necrosis. As shown inFig. 1C, there was no significant

    leakage of LDH in cells after treatment with astin B at 15 and

    30 lM for 24 h. Increased LDH leakage was observed only in cellsexposed to high concentrations of 45 and 60 lM.

    3.2. Astin B induces oxidative stress

    To determine whether astin B induces oxidative stress, we

    observed the effects of astin B on ROS production and GSH levels.Compared with untreated cells, the intracellular ROS levels

    increased significantly when the cells were exposed to astin B for

    12 h (Fig. 2A). In contrast, the GSH levels in the cells were reduced

    in a concentration-dependent manner after treatment with astin B

    for 24 h (Fig. 2B). Astin B promotes ROS production and decreases

    GSH levels. Because JNK activity and oxidative stress are tightly

    associated, we further observed the effects of astin B on JNK phos-

    phorylation in cells treated with astin B at 30 lM for 0, 3, 6, 12, 24,and 48 h. As shown inFig. 2C, the phosphorylation of JNK was obvi-

    ously enhanced after treatment with astin B for 3, 6, and 12 h.

    Chlorogenic acid (CGA) is a well-known antioxidant and potential

    hepatoprotective [19]. After pretreatment with CGA for 12 h, the

    cell viability of astin B-treated (15, 30, and 60lM) groupsincreased by 7.76% (p< 0.001), 9.24% (p< 0.01), and 6.03%

    (p< 0.05), compared with the corresponding groups without pre-

    treated by CGA (Fig. 2D). This may be evidenced from the reverse

    side that astin B can cause oxidative stress and liver injury.

    3.3. Astin B induces mitochondrial dysfunction

    To assess whether astin B affects the functioning of mitochon-

    dria, we determined the changes in the mitochondrial membrane

    potential (DWm). The results showed that astin B induced concen-

    tration-dependent DWm collapse in L-02 cells after incubation

    with astin B for 24 h (Fig. 3A). Furthermore, the release of cyt c

    from the mitochondria into the cytosol in the L-02 cells was also

    detected. Exposure of cells to astin B for 24 h significantly

    increased the cyt c content of the cytosol fraction (Fig. 3B).

    3.4. Astin B regulates Bcl-2/Bax expression and increases caspase-3

    and -9 activity

    As apoptotic proteins, Bcl-2 is a powerful antagonist of apopto-tic death programs, whereas Bax accelerates apoptotic death and

    counters the death repressor activity of Bcl-2. The ratio of Bcl-2

    to Bax determines the survival or death of the cells following an

    apoptotic stimulus[20]. Treatment of L-02 cells with astin B signif-

    icantly induced the expression of pro-apoptotic Bax and decreased

    the expression of anti-apoptotic Bcl-2 in a concentration-

    dependent fashion (Fig. 4A). Thus, astin B significantly increased

    the Bax/Bcl-2 ratio (Fig. 4B) in a concentration-dependent manner,

    leading to a state associated with apoptosis. Caspases are most

    likely the most important effector molecules for the execution of

    apoptosis[21]. When the L-02 cells were treated with astin B for

    0

    50

    100

    150

    200

    250

    300

    0 15 30 45 60

    GSH(nmol/mgprotein)

    Astin B (M)

    *

    **

    *

    B

    0

    1000

    2000

    3000

    4000

    5000

    0 15 30 60

    DCFfluorescenceintensity

    Astin B (M)

    A C* * *

    0.0

    0.2

    0.4

    0.6

    0.8

    1.0

    1.2

    1.4

    0 15 30 60

    Relativeviability

    Astin B (M)

    D

    Fig. 2. Astin B induced oxidative stress in L-02 cells. (A) Cells were treated with astin B for 12 h, and intracellular ROS was assessed by the ROS-specific fluorescent dye DCFH-

    DA. (B) Cells were treated with astin B for 24 h, and the intracellular GSH level was determined using commercially available kits. (C) Cells were treated with astin B (30 lM)at regular intervals, and phosphorylated JNK was determined using Western blot analysis. (D) Cells were pretreated with chlorogenic acid (CGA+, 100 lM) for 12 h, followingtreated with astin B for 24 h, and cell viability was determined by the MTT assay. Data in (A, B, and D) are expressed as the mean SD from three independent experiments

    withn = 46.

    p< 0.05 compared with the control group (0 lM), #p< 0.05 compared with the corresponding CGA group. The results shown in C are representative of threeindependent experiments.

    4 L. Wang et al. / Chemico-Biological Interactions 223 (2014) 19

  • 8/10/2019 Astin B

    5/9

    0

    50

    100

    150

    200

    250

    300

    350

    400

    Cytc(ng/m

    gprotein)

    Astin B (M)

    0 15 30 45 60 0 15 30 45 60

    Cytosol Mitochondria

    *

    *

    * *

    *

    *

    BA

    Fig. 3. Astin B decreased the mitochondrial membrane potential (DWm) and promoted cytochrome c release. (A) L-02 cells were treated with astin B for 24 h and

    subsequently incubated with JC-1 dye for 20 min, then observed and photographed by an Olympus fluorescence microscope. The proportion of green fluorescence emission

    represents the DWm collapse degree. (B) Cells were treated with astin B for 24 h, and the content of cytochrome c was measured with ELISA kits. The results shown in A are

    representative of three independent experiments. Data in (B) are expressed as the mean SD from three independent experiments with n = 4. p< 0.05 compared with the

    control group (0 lM). (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)

    0

    1

    2

    3

    4

    5

    6

    7

    8

    9

    0 15 30 45 60

    Foldofcontrol

    Astin B (M)

    * *

    *

    *D

    Caspase-3

    0

    1

    2

    3

    4

    0 15 30 45 60

    Foldofcontrol

    Astin B (M)

    *

    *

    **

    C Caspase-9

    0

    1

    2

    3

    4

    5

    0 15 30 45 60

    RatioofBax/Bcl-2

    Astin B (M)

    *

    *

    *

    BA

    0

    0.2

    0.4

    0.6

    0.8

    1

    1.2

    1.4

    0 15 30 60

    Relativeviability

    Astin B (M)

    zVAD - zVAD +

    **

    *

    E

    Fig. 4. Astin B regulated Bcl-1/Bax expression and increased activities of caspases-9 and -3 in L-02 cells. Cells were treated with astin B for 24 h: (A) Bax and Bcl-2 expressions

    were determined by Western blot, and (B) the ratio of Bax/Bcl-2 was calculated by densitometry. Enzymatic activities of (C) caspase-9 and (D) caspase-3 were measured using

    commercial kits. (E) Cell viability in the presence of z-VAD-fmk (z-VAD+, 50 lM), a pan-caspase inhibitor, was determined by the MTT assay. The results shown in (A) are

    representative of three independent experiments. The data in B-E are expressed as the mean SD from three independent experiments with n = 3.

    p< 0.05 compared withthe control group (0 lM) in (BD) or with the corresponding z-VAD-group in E.

    L. Wang et al./ Chemico-Biological Interactions 223 (2014) 19 5

    http://-/?-http://-/?-
  • 8/10/2019 Astin B

    6/9

    24 h, the activities of caspases-3 and -9 were increased in concen-

    tration-dependent manners (Fig. 4C and D). Co-treatment of astin B

    with z-VAD-fmk (a pan-caspase inhibitor) could increase the via-

    bility of the L-02 cells to some extent, compared with the groups

    that were only treated by astin B with corresponding concentra-

    tions (Fig. 4E). These results suggest that astin B most likely

    mediates apoptotic cell death and that they do so mainly in a

    caspase-dependent manner.

    3.5. Astin B induces apoptotic cell death

    To confirm whether astin B induced cell death through apopto-

    sis, we analyzed the sub-G1 peaks, i.e., the population of apoptotic

    cells, of the L-02 cells. After treatment of the cells with astin B at 0,

    15, 30, 45, and 60 lM for 24 h, the percentage of the sub-G1 peakgradually increased in a concentration-dependent manner from

    4.09% to 14.22% (Fig. 5A and B), indicating apoptotic cell death

    induced by astin B in L-02 cells. This finding was further evidenced

    by the Annexin-V/PI staining method, which can clearly discrimi-

    nate among the four types of cells, i.e., unaffected cells (AV/

    PI), early apoptotic cells (AV+/PI), early necrotic and late apop-

    totic cells (AV+/PI+), and late necrotic cells (died without apopto-

    sis) (AV/PI+). After treatment with astin B at same

    concentrations for 24 h, the early apoptotic L-02 cells increased

    obviously in a concentration-dependent fashion from 8.03% to

    47.12%, along with lower increases of necrotic and late apoptotic

    cells from 4.64% to 16.33% (Fig. 5C and D). These results indicate

    that apoptosis is one of the major ways for astin B to induce L-02

    cell death, which may be one of the main hepatotoxic components

    in RAT.

    3.6. Astin B induces autophagy against apoptosis

    Autophagy is important for cell death decisions and can protect

    cells by preventing them from undergoing apoptosis [22]. There-

    fore, a method of AO staining was employed to detect autophagiceffects in L-02 cells. As shown in Fig. 6A, after exposure to astin

    B for 12 h, autophagic vacuoles were visible in the cells. Further-

    more, the expressions of LC3-I/LC3-II and p62, both key indicators

    for autophagosome formation[23,24],were examined by Western

    blot analysis. The results showed that the treatment of L-02 cells

    with astin B significantly enhanced LC3-II and reduced p62 expres-

    sion (Fig. 6B). These data indicate that the stimulus of astin B

    induces both autophagy and apoptosis in hepatocytes. In addition,

    the connection between autophagy and apoptosis was investigated

    with astin B-induced L-02 cells by co-treatment with 3-MA (an

    autophagy inhibitor) or z-VAD-fmk (a caspase inhibitor).

    Compared with treatment with astin B alone, co-treatment with

    3-MA obviously attenuated LC3-II expression, rapidly enhanced

    caspase-3 expression, and significantly reduced cell viability in

    L-02 cells (Fig. 6C and D). On the other hand, co-treatment with

    z-VAD-fmk resulted in an accrual of autophagic pathway evi-

    denced by increased LC3-II and decreased p62 (Fig. 6E), which

    may partially explain why the cell viability was only increased

    slightly on blocking apoptosis (Fig. 4E). These results indicated that

    astin B-induced LC3-II enhancement and p62 redaction were the

    0

    2

    4

    6

    8

    10

    12

    14

    16

    0 15 30 45 60

    Cellsofsub-G1peak(%)

    Astin B (M)

    *

    *

    **

    B

    A

    C

    0

    10

    20

    30

    40

    50

    60

    0 15 30 45 60

    Cellnumber(%)

    Astin B (M)

    AV+ PI- A V+PI+

    * * *

    *

    *

    *

    *

    *

    D

    Fig. 5. Astin B increased the apoptotic L-02 cells. Cells were treated with astin B for 24 h, and apoptotic cells were determined by flow cytometry. (A and B) The population of

    sub-G1 peaks (indicated by an arrow) was measured by propidium iodide staining, and (C and D) apoptotic and necrotic cells were clarified by Annexin V-FITC (AV) and

    propidium iodide (PI) staining. The results shown in (A) and (C) are representative of three independent experiments. The data in (B) and (D) are expressed as the mean SDfrom three independent experiments. p< 0.05 compared with the control group (0 lM).

    6 L. Wang et al. / Chemico-Biological Interactions 223 (2014) 19

  • 8/10/2019 Astin B

    7/9

    0

    0.2

    0.4

    0.6

    0.8

    1

    1.2

    Relativeviability

    P 0.05

    DC

    E

    B

    A

    Fig. 6. Astin B induced autophagy in L-02 cells to protect from apoptosis. (A) After treatment with astin B (30lM) for 12 h, autophagy in the L-02 cells was detected byacridine orange (AO) staining, and orange-colored vacuoles indicated positive results, Earles balanced salt solution (EBSS) as positive control. (B) After treatment with astin B

    (15, 30, 60 lM) for 12 h, the levels of p62 and LC3-I/LC3-II in L-02 cells were determined by Western blot analysis. (C and D) L-02 cells were exposed to astin B (60 lM) with orwithout the presence of the autophagy inhibitor 3-MA (2 mM) for 12 h, the levels of LC3-I/LC3-II and procaspase-3/cleaved caspase-3 were determined by Western blot

    analysis, and cell viability was measured by the MTT assay. (E) L-02 cells were treated with astin B (60 lM) with or without the presence of the pan-caspase inhibitor z-VAD-fmk (50 lM) for 12 h, the levels of p62 and LC3-I/LC3-II were determined by Western blot analysis. The results shown in A are representative of three independentexperiments. The results shown in (B, C, and E) are representative of three independent experiments. The data in (D) are expressed as the mean SD from three independent

    experiments.

    **

    **

    0

    20

    40

    60

    80

    100

    120

    140

    160

    Control Astin B

    Enzymeactivity(U/L)

    ALT

    AST

    A B

    Fig. 7. Hepatotoxicity of astin B in mice. Ten mice were orally administered with vehicle (control group) or astin B at 10 mg/kg once daily for 7 consecutive days. (A) ALT and

    AST levels in serum were determined by commercial kits. (B) Slices of liver were stained with H&E for histopathological analysis. The data in (A) are expressed as themean SD, p< 0.01 compared with the control group; the results shown in (B) are representative of five independent experiments at a magnification of 200 .

    L. Wang et al./ Chemico-Biological Interactions 223 (2014) 19 7

    http://-/?-http://-/?-
  • 8/10/2019 Astin B

    8/9

    real events associated with autophagosomes and that astin B-

    induced apoptotic cell death will be accelerated in the absence of

    autophagy in hepatocytes.

    3.7. Astin B induces liver injury in mice

    To investigate whether astin B induces liver injury in vivo, we

    observed the effects of orally administered astin B on the liver cells

    in mice. Compared to the control group, the serum levels of ALT

    and AST in mice treated with astin B were elevated about 11.9

    and 2.26 times, respectively (Fig. 7A). The liver injury of astin B

    was further confirmed by histopathological examination, as

    evidenced by diffuse hydropic degeneration of hepatocytes with

    moderate inflammatory cell infiltration (Fig. 7B). The results

    indicated that, consistent with its action in vitro, astin B could also

    induce liver injuryin vivo.

    4. Discussion

    Our previous work suggested that pentapeptides, especially the

    cyclic ones, were most likely the principal toxic components in RAT

    [14]. Astins are a class of halogenated cyclic pentapeptides con-

    tained in RAT. Although astins have been reported to kill tumor

    cells and prevent colitis through regulation of apoptosis [1113],

    our results clearly showed that astin B, one of the cyclic pentapep-

    tides, induced hepatic cell death mainly by ROS-associated apopto-

    sis via a mitochondria-dependent pathway. Such cell death will

    lead to liver injury, which was evidenced by thein vivoexperiment.

    On the other hand, astin B also positively regulated autophagy, and

    this action partially attenuated apoptotic cell death.

    MTT assays showed that astin B effectively inhibited cell prolif-

    eration and survival. The inhibitory percentage reached 60.78% at

    60 lM after 48 h exposure. Apoptosis and necrosis are the mostfrequent events in xenobiotic metabolism-induced liver injury

    [25]. Astin B substantially increased cell death, while the leakageof LDH, an indicator of necrosis, only moderately increased in cells

    that were exposed to high concentrations. This finding indicates

    that apoptosis is most likely the major event that is responsible

    for the cell death.

    Uncontrolled ROS production and/or decreased antioxidant

    defenses result in oxidative stress, which has been considered to

    be an important pathogenic element for the initiation of hepato-

    toxicity[4,6]. GSH is a major antioxidant that guards cells against

    oxidative injury by diminishing ROS. Therefore alterations in the

    GSH level can be monitored as an indication of oxidative stress

    in cells [26]. Astin B treatment enhanced ROS production with

    downregulation of GSH levels in L-02 cells, indicating that oxida-

    tive stress occurred following the astin B stimulus. Changes in

    ROS and GSH act as intracellular signals and can evoke inflamma-tion by the regulation of transcription factor molecules through

    JNK activation [27]. Consistent with this view, we observed

    enhanced pJNK expression when the cells were exposed to astin

    B. Moreover, the inhibited cell proliferation and survival by astin

    B could be reversed by antioxidants, such as CGA, to some extent.

    All these alterations indicate that the astin B stimulus induces

    ROS-associated inflammation in hepatocytes.

    Given that the mitochondria are the major intracellular source

    of ROS, we wondered whether ROS-associated inflammation would

    impair mitochondrial function. Our experiments showed that astin

    B induces the collapse ofDWm with an increased release of cyt c

    from the mitochondria into the cytosol. This result suggests that

    mitochondrial dysfunction results in the opening of the mitochon-

    drial permeability transition pores, leading to the release of cyt cfrom the mitochondria.

    In view of the link between mitochondrial dysfunction and

    apoptosis, the participation of the Bcl-2 family (which contains

    both pro- and anti-apoptotic proteins) is a key event because of

    the regulative effects of these proteins on mitochondrial dysfunc-

    tion during apoptosis. In such regulation, Bcl-2 is an anti-apoptotic

    member, while Bax is a pro-apoptotic multidomain effector protein

    [28]. Increased pro-apoptotic proteins can actively permeabilize

    the outer mitochondrial membrane and promote the release ofintermembrane space proteins, such as cyt c, into the cytosol. Cyt

    c is required for the activation of caspase-9. Generally, activated

    caspase-9 is involved in the mitochondrial pathway, and it acti-

    vates downstream caspase-3, thereby triggering cell apoptosis

    [29]. The increased ratio of Bax/Bcl-2 observed in cells treated with

    astin B indicates that astin B induces mitochondrial outer mem-

    brane permeabilization, which discharges cyt c into the cytosol.

    Furthermore, the increased activities of caspases-9 and -3 observed

    in treated cells suggest that the astin B-induced apoptosis may fol-

    low a caspase-dependent intrinsic mitochondrial pathway. The ini-

    tiation of apoptosis induced by astin B was further characterized

    by an increased sub-G1 peak population during the cell cycle and

    a predominant population of early apoptotic cells among the dead

    cells.

    Autophagy functions as a cytoplasmic quality control mecha-

    nism to remove protein aggregates and damaged organelles.

    Although autophagy has been observed in many dying cells, it is

    generally accepted that autophagy is a pro-survival and protective

    pathway [30,31]. Apoptosis and autophagy are genetically regu-

    lated processes that regulate cell fate. The functional relationship

    between apoptosis and autophagy is quite complex. In several sce-

    narios, autophagy prevents cell death and de facto suppresses

    apoptosis; and it also appears that similar stimuli can induce either

    apoptosis or autophagy [32]. AO-stained autophagic vacuoles,

    enhanced LC3-II and reduced p62 in L-02 cells treated with astin

    B confirmed that astin B can also induce autophagy in hepatocytes.

    After co-treatment with the autophagy inhibitor 3-MA, decreased

    cell viability and dramatically increased expression of cleaved

    caspase-3 were observed. Conversely, co-treatment with caspaseinhibitor z-VAD-fmk led to an enhanced autophagy for astin

    B-treated cells evidenced by increased LC3-II and decreased p62

    expression. These results indicate that apoptosis and autophagy

    induced by astin B were interacted and that autophagy could

    effectively protect the cells from B-induced cell apoptosis. The

    protective pathway of autophagy,i.e., whether autophagy is a sec-

    ondary result of cell death that functions to remove damaged

    organelles or if it is directly induced by astin B in an apoptosis-

    independent manner, remains to be determined.

    In summary, our studies suggest that astin B will provoke oxi-

    dative stress-associated inflammation and induce hepatocyte

    apoptosis through a mitochondria-dependent pathway, leading to

    liver injury. However, astin B will also induce autophagy to protect

    cells from apoptosis and to alleviate hepatic injury to some extent.Furthermore, we found that an astin-rich fraction (astins were

    71.2% of the relative content)[14] induces apoptosis in the same

    fashion (data not shown). As a result, astins (including astin B)

    can be considered to be the major hepatotoxic substances in the

    herbal medicine derived from A. tataricus. Owing to their special

    and interesting structures, previous studies of astins focused on

    their biological activities, and their toxic or side effects were often

    overlooked. We hope that our finding will be helpful for providing

    advice regarding drug safety for the clinical use of RAT and the

    development of new pharmaceuticals.

    Conflict of Interest

    The authors declare that there are no conflicts of interest.

    8 L. Wang et al. / Chemico-Biological Interactions 223 (2014) 19

  • 8/10/2019 Astin B

    9/9

    Transparency Document

    TheTransparency documentassociated with this article can be

    found in the online version.

    Acknowledgments

    This work was financially supported by the National Natural

    Science Foundation of China (30772702) and the National New

    Drug Innovation Major Project of China (2011ZX09307-002-02).

    References

    [1] F. Stickel, E. Patsenker, D. Schuppan, Herbal hepatotoxicity, J. Hepatol. 43(2005) 901910.

    [2] H. Malhi, G.J. Gores, J.J. Lemasters, Apoptosis and necrosis in the liver: a tale oftwo deaths?, Hepatology 43 (2006) S31S44

    [3] G. Kroemer, L. Galluzzi, P. Vandenabeele, J. Abrams, E.S. Alnemri, E.H.Baehrecke, M.V. Blagosklonny, W.S. Eldeiry, P. Golstein, D.R. Green, M.Hengartner, R.A. Knight, S. Kumar, S.A. Lipton, W. Malorni, G. Nunez, M.E.Peter, J. Tschopp, J. Yuan, M. Piacentini, B. Zhivotovsky, G. Melino, Classificationof cell death: recommendations of the Nomenclature Committee on cell death,Cell Death Differ. 16 (2009) 311.

    [4] J.L. Martindale, N.J. Holbrook, Cellular response to oxidative stress: signalingfor suicide and survival, J. Cell Physiol. 192 (2002) 115.

    [5] R. Heidari, H. Babaei, M. Eghbal, Mechanisms of methimazole cytotoxicity inisolated rat hepatocytes, Drug Chem. Toxicol. 36 (2013) 403411.

    [6] D. Pessayre, B. Fromenty, A. Berson, M.A. Robin, P. Lettron, R. Moreau, A.Mansouri, Central role of mitochondria in drug-induced liver injury, DrugMetab. Rev. 44 (2012) 3487.

    [7] M. Giorgio, E. Migliaccio, F. Orsini, D. Paolucci, M. Moroni, C. Contursi, G.Pelliccia, L. Luzi, S. Minucci, M. Marcaccio, P. Pinton, R. Rizzuto, P. Bernardi, F.Paolucci, P.G. Pelicci, Electron transfer between cytochrome c and p66 (Shc)generates reactive oxygen species that trigger mitochondrial apoptosis, Cell122 (2005) 221233.

    [8] S. Lim, S.J. Lee, K.W. Nam, K.H. Kim, W. Mar, Hepatoprotective effects ofreynosin against thioacetamide-induced apoptosis in primary hepatocytes andmouse liver, Arch. Pharm. Res. 36 (2013) 485494.

    [9] D. Han, L. Dara, S. Win, T.A. Than, L. Yuan, S.Q. Abbasi, Z.X. Liu, N. Kaplowitz,Regulation of drug-induced liver injury by signal transduction pathways:critical role of mitochondria, Trends Pharmacol. Sci. 34 (2013) 243253.

    [10] C. Bincoletto, A. Bechara, G.J. Pereira, C.P. Santos, F. Antunes, J. Peixoto da-Silva,M. Muler, R.D. Gigli, P.T. Monteforte, H. Hirata, A. Jurkiewicz, S.S. Smaili,Interplay between apoptosis and autophagy, a challenging puzzle: newperspectives on antitumor chemotherapies, Chem. Biol. Interact. 206 (2013)279288.

    [11] H. Morita, S. Nagashima, K. Takeya, H. Itokawa, Y. Iitaka, Structures andconformation of antitumour cyclic pentapeptides, astins A, B and C, from Astertataricus, Tetrahedron 51 (1995) 11211132.

    [12] R. Cozzolino, P. Palladino, F. Rossi, G. Cali, E. Benedetti, P. Laccetti,Antineoplastic cyclic astin analogues kill tumour cellsvia caspase-mediatedinduction of apoptosis, Carcinogenesis 26 (2005) 733739.

    [13] Y. Shen, Q. Luo, H.M. Xu, F.Y. Gong, X.B. Zhou, Y. Sun, X.F. Wu, W. Liu, G.Z. Zeng,N.H. Tan, Q. Xu, Mitochondria-dependent apoptosis of activated Tlymphocytes induced by astin C, a plant cyclopeptide, for preventing murineexperimental colitis, Biochem. Pharmacol. 82 (2011) 260268.

    [14] X.D. Liu, P.P. Cao, C.F. Zhang, X.H. Xu, M. Zhang, Screening and analyzingpotential hepatotoxic compounds in the ethanol extract of Asteris Radix byHPLC/DAD/ESI-MSn technique, J. Pharm. Biomed. Anal. 6768 (2012) 5162.

    [15] K. Terao, E. Ito, T. Tatsuno, Liver injuries induced by cyclochlorotine isolatedfromPenicillium islandicum, Arch. Toxicol. 55 (1984) 3946.

    [16] A. Cossarizza, M. Baccaranicontri, G. Kalashnikova, C. Franceschi, A new

    method for the cytofluorometric analysis of mitochondrial membranepotential using the J-aggregate forming lipophilic cation 5,50 ,6,60-tetrachloro-1,10 ,3,30-tetraethylbenzimidazolcarbocyanine iodide (JC-1),Biochem. Biophys. Res. Commun. 197 (1993) 4045.

    [17] V.A. Tyurin, Y.Y. Tyurina, W. Feng, A. Mnuskin, J.M. Jiang, Tang, X. Zhang, Q.Zhao, P.M. Kochanek, R.S. Clark, H. Bayir, V.E. Kagan, Mass-spectrometriccharacterization of phospholipids and their primary peroxidation products inrat cortical neurons during staurosporine-induced apoptosis, J. Neurochem.107 (2008) 16141633.

    [18] S. Paglin, T. Hollister, T. Delohery, N. Hackett, M. McMahill, E. Sphicas, D.Domingo, J. Yahalom, A novel response of cancer cells to radiation involvesautophagy and formation of acidic vesicles, Cancer Res. 61 (2001) 439444.

    [19] A. Kapil, I.B. Koul, O.P. Suri, Antihepatotoxic effects of chlorogenic acid fromAnthocephalus cadamba, Phytother. Res. 9 (1995) 189193.

    [20] Z.N. Oltval, C.L. Milliman, S.J. Korsmeyer, Bcl-2 heterodimerizes in vivo with aconserved homolog, Bax, that accelerates programmed cell death, Cell 74(1993) 609619.

    [21] M. Boyce, A. Degterev, J. Yuan, Caspases: an ancient cellular sword ofDamocles, Cell Death Differ. 11 (2004) 2937.

    [22] A. Thorburn, Apoptosis and autophagy: regulatory connections between twosupposedly different processes, Apoptosis 13 (2008) 19.

    [23] Y. Kouroku, E. Fujita, I. Tanida, T. Ueno, A. Isoai, H. Kumagai, S. Ogawa, R.J.Kaufman, E. Kominami, T. Momoi, ER stress (PERK/eIF2 alpha phosphorylation)mediates the polyglutamine-induced LC3 conversion, an essential step forautophagy formation, Cell Death Differ. 14 (2007) 230239.

    [24] E. Itakura, N. Mizushima, P62 targeting to the autophagosome formation siterequires self-oligomerization but not LC3 binding, J. Cell Biol. 192 (2011) 1727.

    [25] H. Malhi, M.E. Guicciardi, G.J. Gores, Hepatocyte death: a clear and presentdanger, Physiol. Rev. 90 (2010) 11651194.

    [26] L. Yuan, N. Kaplowitz, Glutathione in liver diseases and hepatotoxicity, Mol.Aspects Med. 30 (2009) 2941.

    [27] S.J. Lee, M.S. Kim, J.Y. Park, J.S. Woo, Y.K. Kim, 15-Deoxy-delta (12,14)-prostaglandin J (2) induces apoptosis via JNK-mediated mitochondrialpathway in osteoblastic cells, Toxicology 248 (2008) 121129 .

    [28] R.J. Youle, A. Strasser, The Bcl-2 protein family: opposing activities thatmediate cell death, Nat. Rev. Mol. Cell Biol. 9 (2008) 4759.

    [29] N.N. Danial, S.J. Korsmeyer, Cell death: critical control points, Cell 116 (2004)205219.

    [30] P. Boya, R.A. Gonzalez-Polo, N. Casares, J.L. Perfettini, P. Dessen, N. Larochette,D. Metivier, D. Meley, S. Souquere, T. Yoshimori, G. Pierron, P. Codogno, G.Kroemer, Inhibition of macroautophagy triggers apoptosis, Mol. Cell Biol. 25(2005) 10251040.

    [31] N. Mizushima, B. Levine, A.M. Cuervo, D.J. Klionsky, Autophagy fights diseasethrough cellular self digestion, Nature 451 (2008) 10691075.

    [32] M.C. Maiuri, E. Zalckvar, A. Kimchi, G. Kroemer, Self-eating and self-killing:crosstalk between autophagy and apoptosis, Nat. Rev. Mol. Cell Biol. 8 (2007)741752.

    L. Wang et al./ Chemico-Biological Interactions 223 (2014) 19 9

    http://dx.doi.org/10.1016/j.cbi.2014.09.003http://refhub.elsevier.com/S0009-2797(14)00251-8/h0005http://refhub.elsevier.com/S0009-2797(14)00251-8/h0005http://refhub.elsevier.com/S0009-2797(14)00251-8/h0005http://refhub.elsevier.com/S0009-2797(14)00251-8/h0010http://refhub.elsevier.com/S0009-2797(14)00251-8/h0010http://refhub.elsevier.com/S0009-2797(14)00251-8/h0015http://refhub.elsevier.com/S0009-2797(14)00251-8/h0015http://refhub.elsevier.com/S0009-2797(14)00251-8/h0015http://refhub.elsevier.com/S0009-2797(14)00251-8/h0015http://refhub.elsevier.com/S0009-2797(14)00251-8/h0015http://refhub.elsevier.com/S0009-2797(14)00251-8/h0015http://refhub.elsevier.com/S0009-2797(14)00251-8/h0020http://refhub.elsevier.com/S0009-2797(14)00251-8/h0020http://refhub.elsevier.com/S0009-2797(14)00251-8/h0020http://refhub.elsevier.com/S0009-2797(14)00251-8/h0025http://refhub.elsevier.com/S0009-2797(14)00251-8/h0025http://refhub.elsevier.com/S0009-2797(14)00251-8/h0025http://refhub.elsevier.com/S0009-2797(14)00251-8/h0030http://refhub.elsevier.com/S0009-2797(14)00251-8/h0030http://refhub.elsevier.com/S0009-2797(14)00251-8/h0030http://refhub.elsevier.com/S0009-2797(14)00251-8/h0030http://refhub.elsevier.com/S0009-2797(14)00251-8/h0035http://refhub.elsevier.com/S0009-2797(14)00251-8/h0035http://refhub.elsevier.com/S0009-2797(14)00251-8/h0035http://refhub.elsevier.com/S0009-2797(14)00251-8/h0035http://refhub.elsevier.com/S0009-2797(14)00251-8/h0035http://refhub.elsevier.com/S0009-2797(14)00251-8/h0040http://refhub.elsevier.com/S0009-2797(14)00251-8/h0040http://refhub.elsevier.com/S0009-2797(14)00251-8/h0040http://refhub.elsevier.com/S0009-2797(14)00251-8/h0045http://refhub.elsevier.com/S0009-2797(14)00251-8/h0045http://refhub.elsevier.com/S0009-2797(14)00251-8/h0045http://refhub.elsevier.com/S0009-2797(14)00251-8/h0050http://refhub.elsevier.com/S0009-2797(14)00251-8/h0050http://refhub.elsevier.com/S0009-2797(14)00251-8/h0050http://refhub.elsevier.com/S0009-2797(14)00251-8/h0050http://refhub.elsevier.com/S0009-2797(14)00251-8/h0050http://refhub.elsevier.com/S0009-2797(14)00251-8/h0055http://refhub.elsevier.com/S0009-2797(14)00251-8/h0055http://refhub.elsevier.com/S0009-2797(14)00251-8/h0055http://refhub.elsevier.com/S0009-2797(14)00251-8/h0055http://refhub.elsevier.com/S0009-2797(14)00251-8/h0055http://refhub.elsevier.com/S0009-2797(14)00251-8/h0060http://refhub.elsevier.com/S0009-2797(14)00251-8/h0060http://refhub.elsevier.com/S0009-2797(14)00251-8/h0060http://refhub.elsevier.com/S0009-2797(14)00251-8/h0060http://refhub.elsevier.com/S0009-2797(14)00251-8/h0060http://refhub.elsevier.com/S0009-2797(14)00251-8/h0065http://refhub.elsevier.com/S0009-2797(14)00251-8/h0065http://refhub.elsevier.com/S0009-2797(14)00251-8/h0065http://refhub.elsevier.com/S0009-2797(14)00251-8/h0065http://refhub.elsevier.com/S0009-2797(14)00251-8/h0070http://refhub.elsevier.com/S0009-2797(14)00251-8/h0070http://refhub.elsevier.com/S0009-2797(14)00251-8/h0070http://refhub.elsevier.com/S0009-2797(14)00251-8/h0070http://refhub.elsevier.com/S0009-2797(14)00251-8/h0075http://refhub.elsevier.com/S0009-2797(14)00251-8/h0075http://refhub.elsevier.com/S0009-2797(14)00251-8/h0075http://refhub.elsevier.com/S0009-2797(14)00251-8/h0075http://refhub.elsevier.com/S0009-2797(14)00251-8/h0080http://refhub.elsevier.com/S0009-2797(14)00251-8/h0080http://refhub.elsevier.com/S0009-2797(14)00251-8/h0080http://refhub.elsevier.com/S0009-2797(14)00251-8/h0080http://refhub.elsevier.com/S0009-2797(14)00251-8/h0080http://refhub.elsevier.com/S0009-2797(14)00251-8/h0080http://refhub.elsevier.com/S0009-2797(14)00251-8/h0080http://refhub.elsevier.com/S0009-2797(14)00251-8/h0080http://refhub.elsevier.com/S0009-2797(14)00251-8/h0080http://refhub.elsevier.com/S0009-2797(14)00251-8/h0080http://refhub.elsevier.com/S0009-2797(14)00251-8/h0080http://refhub.elsevier.com/S0009-2797(14)00251-8/h0085http://refhub.elsevier.com/S0009-2797(14)00251-8/h0085http://refhub.elsevier.com/S0009-2797(14)00251-8/h0085http://refhub.elsevier.com/S0009-2797(14)00251-8/h0085http://refhub.elsevier.com/S0009-2797(14)00251-8/h0085http://refhub.elsevier.com/S0009-2797(14)00251-8/h0085http://refhub.elsevier.com/S0009-2797(14)00251-8/h0090http://refhub.elsevier.com/S0009-2797(14)00251-8/h0090http://refhub.elsevier.com/S0009-2797(14)00251-8/h0090http://refhub.elsevier.com/S0009-2797(14)00251-8/h0095http://refhub.elsevier.com/S0009-2797(14)00251-8/h0095http://refhub.elsevier.com/S0009-2797(14)00251-8/h0095http://refhub.elsevier.com/S0009-2797(14)00251-8/h0100http://refhub.elsevier.com/S0009-2797(14)00251-8/h0100http://refhub.elsevier.com/S0009-2797(14)00251-8/h0100http://refhub.elsevier.com/S0009-2797(14)00251-8/h0105http://refhub.elsevier.com/S0009-2797(14)00251-8/h0105http://refhub.elsevier.com/S0009-2797(14)00251-8/h0110http://refhub.elsevier.com/S0009-2797(14)00251-8/h0110http://refhub.elsevier.com/S0009-2797(14)00251-8/h0115http://refhub.elsevier.com/S0009-2797(14)00251-8/h0115http://refhub.elsevier.com/S0009-2797(14)00251-8/h0115http://refhub.elsevier.com/S0009-2797(14)00251-8/h0115http://refhub.elsevier.com/S0009-2797(14)00251-8/h0120http://refhub.elsevier.com/S0009-2797(14)00251-8/h0120http://refhub.elsevier.com/S0009-2797(14)00251-8/h0120http://refhub.elsevier.com/S0009-2797(14)00251-8/h0125http://refhub.elsevier.com/S0009-2797(14)00251-8/h0125http://refhub.elsevier.com/S0009-2797(14)00251-8/h0130http://refhub.elsevier.com/S0009-2797(14)00251-8/h0130http://refhub.elsevier.com/S0009-2797(14)00251-8/h0135http://refhub.elsevier.com/S0009-2797(14)00251-8/h0135http://refhub.elsevier.com/S0009-2797(14)00251-8/h0135http://refhub.elsevier.com/S0009-2797(14)00251-8/h0135http://refhub.elsevier.com/S0009-2797(14)00251-8/h0135http://refhub.elsevier.com/S0009-2797(14)00251-8/h0140http://refhub.elsevier.com/S0009-2797(14)00251-8/h0140http://refhub.elsevier.com/S0009-2797(14)00251-8/h0145http://refhub.elsevier.com/S0009-2797(14)00251-8/h0145http://refhub.elsevier.com/S0009-2797(14)00251-8/h0150http://refhub.elsevier.com/S0009-2797(14)00251-8/h0150http://refhub.elsevier.com/S0009-2797(14)00251-8/h0150http://refhub.elsevier.com/S0009-2797(14)00251-8/h0150http://refhub.elsevier.com/S0009-2797(14)00251-8/h0155http://refhub.elsevier.com/S0009-2797(14)00251-8/h0155http://refhub.elsevier.com/S0009-2797(14)00251-8/h0160http://refhub.elsevier.com/S0009-2797(14)00251-8/h0160http://refhub.elsevier.com/S0009-2797(14)00251-8/h0160http://refhub.elsevier.com/S0009-2797(14)00251-8/h0160http://refhub.elsevier.com/S0009-2797(14)00251-8/h0160http://refhub.elsevier.com/S0009-2797(14)00251-8/h0160http://refhub.elsevier.com/S0009-2797(14)00251-8/h0155http://refhub.elsevier.com/S0009-2797(14)00251-8/h0155http://refhub.elsevier.com/S0009-2797(14)00251-8/h0150http://refhub.elsevier.com/S0009-2797(14)00251-8/h0150http://refhub.elsevier.com/S0009-2797(14)00251-8/h0150http://refhub.elsevier.com/S0009-2797(14)00251-8/h0150http://refhub.elsevier.com/S0009-2797(14)00251-8/h0145http://refhub.elsevier.com/S0009-2797(14)00251-8/h0145http://refhub.elsevier.com/S0009-2797(14)00251-8/h0140http://refhub.elsevier.com/S0009-2797(14)00251-8/h0140http://refhub.elsevier.com/S0009-2797(14)00251-8/h0135http://refhub.elsevier.com/S0009-2797(14)00251-8/h0135http://refhub.elsevier.com/S0009-2797(14)00251-8/h0135http://refhub.elsevier.com/S0009-2797(14)00251-8/h0130http://refhub.elsevier.com/S0009-2797(14)00251-8/h0130http://refhub.elsevier.com/S0009-2797(14)00251-8/h0125http://refhub.elsevier.com/S0009-2797(14)00251-8/h0125http://refhub.elsevier.com/S0009-2797(14)00251-8/h0120http://refhub.elsevier.com/S0009-2797(14)00251-8/h0120http://refhub.elsevier.com/S0009-2797(14)00251-8/h0120http://refhub.elsevier.com/S0009-2797(14)00251-8/h0115http://refhub.elsevier.com/S0009-2797(14)00251-8/h0115http://refhub.elsevier.com/S0009-2797(14)00251-8/h0115http://refhub.elsevier.com/S0009-2797(14)00251-8/h0115http://refhub.elsevier.com/S0009-2797(14)00251-8/h0110http://refhub.elsevier.com/S0009-2797(14)00251-8/h0110http://refhub.elsevier.com/S0009-2797(14)00251-8/h0105http://refhub.elsevier.com/S0009-2797(14)00251-8/h0105http://refhub.elsevier.com/S0009-2797(14)00251-8/h0100http://refhub.elsevier.com/S0009-2797(14)00251-8/h0100http://refhub.elsevier.com/S0009-2797(14)00251-8/h0100http://refhub.elsevier.com/S0009-2797(14)00251-8/h0095http://refhub.elsevier.com/S0009-2797(14)00251-8/h0095http://refhub.elsevier.com/S0009-2797(14)00251-8/h0090http://refhub.elsevier.com/S0009-2797(14)00251-8/h0090http://refhub.elsevier.com/S0009-2797(14)00251-8/h0090http://refhub.elsevier.com/S0009-2797(14)00251-8/h0085http://refhub.elsevier.com/S0009-2797(14)00251-8/h0085http://refhub.elsevier.com/S0009-2797(14)00251-8/h0085http://refhub.elsevier.com/S0009-2797(14)00251-8/h0085http://refhub.elsevier.com/S0009-2797(14)00251-8/h0085http://refhub.elsevier.com/S0009-2797(14)00251-8/h0080http://refhub.elsevier.com/S0009-2797(14)00251-8/h0080http://refhub.elsevier.com/S0009-2797(14)00251-8/h0080http://refhub.elsevier.com/S0009-2797(14)00251-8/h0080http://refhub.elsevier.com/S0009-2797(14)00251-8/h0080http://refhub.elsevier.com/S0009-2797(14)00251-8/h0080http://refhub.elsevier.com/S0009-2797(14)00251-8/h0080http://refhub.elsevier.com/S0009-2797(14)00251-8/h0080http://refhub.elsevier.com/S0009-2797(14)00251-8/h0080http://refhub.elsevier.com/S0009-2797(14)00251-8/h0075http://refhub.elsevier.com/S0009-2797(14)00251-8/h0075http://refhub.elsevier.com/S0009-2797(14)00251-8/h0070http://refhub.elsevier.com/S0009-2797(14)00251-8/h0070http://refhub.elsevier.com/S0009-2797(14)00251-8/h0070http://refhub.elsevier.com/S0009-2797(14)00251-8/h0070http://refhub.elsevier.com/S0009-2797(14)00251-8/h0065http://refhub.elsevier.com/S0009-2797(14)00251-8/h0065http://refhub.elsevier.com/S0009-2797(14)00251-8/h0065http://refhub.elsevier.com/S0009-2797(14)00251-8/h0065http://refhub.elsevier.com/S0009-2797(14)00251-8/h0060http://refhub.elsevier.com/S0009-2797(14)00251-8/h0060http://refhub.elsevier.com/S0009-2797(14)00251-8/h0060http://-/?-http://refhub.elsevier.com/S0009-2797(14)00251-8/h0055http://refhub.elsevier.com/S0009-2797(14)00251-8/h0055http://refhub.elsevier.com/S0009-2797(14)00251-8/h0055http://-/?-http://refhub.elsevier.com/S0009-2797(14)00251-8/h0050http://refhub.elsevier.com/S0009-2797(14)00251-8/h0050http://refhub.elsevier.com/S0009-2797(14)00251-8/h0050http://refhub.elsevier.com/S0009-2797(14)00251-8/h0050http://refhub.elsevier.com/S0009-2797(14)00251-8/h0050http://-/?-http://refhub.elsevier.com/S0009-2797(14)00251-8/h0045http://refhub.elsevier.com/S0009-2797(14)00251-8/h0045http://refhub.elsevier.com/S0009-2797(14)00251-8/h0045http://refhub.elsevier.com/S0009-2797(14)00251-8/h0040http://refhub.elsevier.com/S0009-2797(14)00251-8/h0040http://refhub.elsevier.com/S0009-2797(14)00251-8/h0040http://refhub.elsevier.com/S0009-2797(14)00251-8/h0035http://refhub.elsevier.com/S0009-2797(14)00251-8/h0035http://refhub.elsevier.com/S0009-2797(14)00251-8/h0035http://refhub.elsevier.com/S0009-2797(14)00251-8/h0035http://refhub.elsevier.com/S0009-2797(14)00251-8/h0035http://refhub.elsevier.com/S0009-2797(14)00251-8/h0030http://refhub.elsevier.com/S0009-2797(14)00251-8/h0030http://refhub.elsevier.com/S0009-2797(14)00251-8/h0030http://refhub.elsevier.com/S0009-2797(14)00251-8/h0025http://refhub.elsevier.com/S0009-2797(14)00251-8/h0025http://refhub.elsevier.com/S0009-2797(14)00251-8/h0020http://refhub.elsevier.com/S0009-2797(14)00251-8/h0020http://refhub.elsevier.com/S0009-2797(14)00251-8/h0015http://refhub.elsevier.com/S0009-2797(14)00251-8/h0015http://refhub.elsevier.com/S0009-2797(14)00251-8/h0015http://refhub.elsevier.com/S0009-2797(14)00251-8/h0015http://refhub.elsevier.com/S0009-2797(14)00251-8/h0015http://refhub.elsevier.com/S0009-2797(14)00251-8/h0015http://refhub.elsevier.com/S0009-2797(14)00251-8/h0010http://refhub.elsevier.com/S0009-2797(14)00251-8/h0010http://refhub.elsevier.com/S0009-2797(14)00251-8/h0005http://refhub.elsevier.com/S0009-2797(14)00251-8/h0005http://-/?-http://-/?-http://dx.doi.org/10.1016/j.cbi.2014.09.003http://-/?-