32
Accepted Manuscript Protection of Quercetin against Triptolide-induced apoptosis by suppressing oxidative stress in rat Leydig cells Jie Hu, Qinwei Yu, Fang Zhao, Jinzi Ji, Zhenzhou Jiang, Xin Chen, Peng Gao, Yuran Ren, Shuai Shao, Luyong Zhang, Ming Yan PII: S0009-2797(15)30033-8 DOI: 10.1016/j.cbi.2015.08.004 Reference: CBI 7434 To appear in: Chemico-Biological Interactions Received Date: 4 March 2015 Revised Date: 6 August 2015 Accepted Date: 7 August 2015 Please cite this article as: J. Hu, Q. Yu, F. Zhao, J. Ji, Z. Jiang, X. Chen, P. Gao, Y. Ren, S. Shao, L. Zhang, M. Yan, Protection of Quercetin against Triptolide-induced apoptosis by suppressing oxidative stress in rat Leydig cells, Chemico-Biological Interactions (2015), doi: 10.1016/j.cbi.2015.08.004. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Protection of Quercetin against Triptolide-induced ...download.xuebalib.com/2vzvZSDvkbEg.pdf · T D ACCEPTED MANUSCRIPT Protection of Quercetin against Triptolide-induced apoptosis

  • Upload
    others

  • View
    7

  • Download
    0

Embed Size (px)

Citation preview

  • Accepted Manuscript

    Protection of Quercetin against Triptolide-induced apoptosis by suppressing oxidativestress in rat Leydig cells

    Jie Hu, Qinwei Yu, Fang Zhao, Jinzi Ji, Zhenzhou Jiang, Xin Chen, Peng Gao, YuranRen, Shuai Shao, Luyong Zhang, Ming Yan

    PII: S0009-2797(15)30033-8

    DOI: 10.1016/j.cbi.2015.08.004

    Reference: CBI 7434

    To appear in: Chemico-Biological Interactions

    Received Date: 4 March 2015

    Revised Date: 6 August 2015

    Accepted Date: 7 August 2015

    Please cite this article as: J. Hu, Q. Yu, F. Zhao, J. Ji, Z. Jiang, X. Chen, P. Gao, Y. Ren, S. Shao, L.Zhang, M. Yan, Protection of Quercetin against Triptolide-induced apoptosis by suppressing oxidativestress in rat Leydig cells, Chemico-Biological Interactions (2015), doi: 10.1016/j.cbi.2015.08.004.

    This is a PDF file of an unedited manuscript that has been accepted for publication. As a service toour customers we are providing this early version of the manuscript. The manuscript will undergocopyediting, typesetting, and review of the resulting proof before it is published in its final form. Pleasenote that during the production process errors may be discovered which could affect the content, and alllegal disclaimers that apply to the journal pertain.

    http://dx.doi.org/10.1016/j.cbi.2015.08.004

  • MAN

    USCR

    IPT

    ACCE

    PTED

    ACCEPTED MANUSCRIPT

  • MAN

    USCR

    IPT

    ACCE

    PTED

    ACCEPTED MANUSCRIPT

    Protection of Quercetin against Triptolide-induced apoptosis by

    suppressing oxidative stress in rat Leydig cells

    Jie Hua,1 , Qinwei Yua,1 , Fang Zhaoa, Jinzi Jia,c, Zhenzhou Jianga, b, Xin

    Chena, Peng Gaoa, Yuran Rend , Shuai Shaod, Luyong Zhanga, b, *, Ming

    Yana, *

    a Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical

    University, Nanjing 210009, China.

    b Key Laboratory of Drug Quality Control and Pharmacovigilance (China

    Pharmaceutical University), Ministry of Education, Nanjing 210009, PR

    China

    c Central Laboratory, General Clinical Research Center, Nanjing First

    Hospital, Nanjing 210006, PR China

    d School of Pharmacy, China Pharmaceutical University, Nanjing 210009,

    PR China

    1 These authors equally contributed to this work.

    *Corresponding authors at: Jiangsu Key Laboratory of Drug Screening,

    China Pharmaceutical University, Jiangsu Province, Nanjing 210009,

    China; Tel: +86 025 83271142; Fax: +86 025 83271142.

    E-mail addresses: [email protected] (Luyong Zhang), Abbreviations: TP, Triptolide; Que, Quercetin; ∆Ψm, mitochondrial membrane potential; GPx, glutathione peroxidase; SOD, superoxide dismutase; ROS, reactive oxygen Species; Nrf2, NF-E2-related factor; Cyt-C, cytochrome C; JNK, c-Jun Nterminal kinase.

  • MAN

    USCR

    IPT

    ACCE

    PTED

    ACCEPTED MANUSCRIPT

    [email protected] (Ming Yan).

    1. Introduction

    Triptolide (TP), derived from the medicinal plant Triterygium wilfordii Hook.f. (TWHF), is a

    diterpene triepoxide with variety biological activities, such as anti-inflammatory, anti-cancerogenic,

    immunomodulatory and pro-apoptotic activities [1, 2]. However, its potential toxicity to circulatory

    and reproductive system limits its clinical application [3]. A recent study on male reproductive

    toxicity showed that TP could induce decrease of testis and epididymis weights and apparent

    changes of seminiferous tubules and epididymides [4]. In addition, it was demonstrated that the

    sperm viability and motility in canda epididymal fluid could be reduced by TP, but the mechanism

    has been unknown [5]. One major disruptive factor of reproductive function in Leydig cells is

    oxidative stress, which is also the main reason of male infertility in pathology [6]. Similarly,

    spermatogenesis is also vulnerable to oxidative stress because of a low oxygen demand in

    physiology [7]. All of these damage are resulted from over-accumulation of reactive oxygen species

    (ROS). Therefore, anti-oxidant enzymes, such as superoxide dismutase (SOD) and glutathione

    peroxidase (GPx), and other free radical scavengers are demanded for protecting testis from free

    radical damage to maintain its normal function [8-10]. NF-E2-related factor (Nrf2) is a

    redox-sensitive transcription factor. With the increase of ROS, the dissociation of Nrf2 and Keap1

    causes Nrf2 translocating into nucleus and binding to antioxidant response elements (AREs), which

    leads protection against oxidative stress [11].

    Quercetin (Que), as a plant phenolic compound, is a member of flavonoids discovered in many

    dietary sources [12] with pharmacological activities involving anti-cancerogenic, antiviral,

  • MAN

    USCR

    IPT

    ACCE

    PTED

    ACCEPTED MANUSCRIPT

    anti-ischemic, anti-inflammatory and antiallergenic [13]. It had been demonstrated that it had

    protective effect on melphalan-mediated oxidative stress in kidney and liver [14]. Meanwhile, the

    effect to protect embryonic chicken spermatogonial cells from oxidative damage was also be

    confirmed, which suggests its availability in male reproductive protection as free radical scavenger

    [15].

    Interaction between apoptosis and mitochondria damage is revealed by accumulated data. As

    one major source of ROS, mitochondria mediated an important signaling pathway in apoptosis [16].

    The decrease of mitochondria membrane potential (∆Ψm) and the release of cytochrome C (Cyt-C)

    are two major events in this mitochondrial pathway. Thereby, caspases, a family of cysteine

    proteases, are activated, especially caspase-3 and caspase-9 [17, 18]. Bcl-2 family proteins include

    both anti-apoptotic (BCL-2, BCL-XL) and pro-apoptotic (BAX, BAD, BAK, and BID) members,

    could regulate Cyt-C releasing, thereby control apoptosis [19]. Yao et al. [20] showed TP-induced

    cytotoxicity in human normal liver L-02 cells involved mitochondrial pathway.

    In this study, we investigated the reproductive toxicity of TP and to assess whether these effect

    can be ameliorated by pre-treatment with Que. The free radical damage and restoration were

    assessed by measurement of ROS, SOD and GPx. The evaluation of mitochondrial function was

    achieved by JC-1 assay. In addition, to identify the apoptotic action of TP and anti-apoptosis action

    of Que, the activities of cytosolic CytC, Bcl-2 family proteins (BAX, Bcl-2), caspase-3 and

    caspase-9 were measured. This study would guide a clinical approach to reduce the TP-mediated

    productive toxicity by anti-oxidant candidates.

    2. Materials and methods

    2.1 Materials and reagents

  • MAN

    USCR

    IPT

    ACCE

    PTED

    ACCEPTED MANUSCRIPT

    Triptolide and Quercetin (>98% purity) were purchased from the National Institute for the

    Control of Pharmaceutical and Biological Products (Beijing, China). Antibodies to Nrf2 were

    purchased from Santa Cruz Biotechnology (Santa Cruz, CA, USA). Antibodies to caspase-9,

    caspase-3 and bcl-2 were purchased from Cell Signaling Technology (CST, MA, USA).

    Anti-Cytochrome c antibody was purchased from GeneTex (GeneTex, CA, USA). Anti-Bax

    antibody was purchased from Signalway Antibody (SAB, CA, USA).

    2.2 Animals

    Sprague-Dawley (SD) rats (250±20g) were purchased from QingLongShan Laboratory

    Animal Company (Nanjing, China).

    2.3 Isolation and culture of Leydig cells

    Leydig cells were isolated as previously described [21] with some modifications. Briefly, the

    testis were dissociated in 1mg/ml collagenase II (SigmaAldrich, MO, USA) by shaking in an

    orbital miser incubator at 1.16 g for 30 min at 37°C. The isolation procedure involved collagenase

    digestion and Percoll (GE, CT, USA) density centrifugation according to a method described

    previously. Following testis digestion, the collecting filtrate was then subjected to Percoll density

    centrifugation and isolation of Leydig cells at a density between 1.070 and 1.088 g/ml. In general,

    Leydig cells were cultured in 6-well plates (1.0×106 cells/well) at 37°C and 5% CO2 under

    humidified conditions. The cells were cultured in DMEM/F12 medium (Invitrogen, CA, USA )

    supplied with 5% fetal bovine serum (HyClone, UT, USA), 1 mM sodium pyruvate (Invitrogen,

    CA, USA), antibiotics (Invitrogen, CA, USA), 10% BSA(Shengxing, Jiangsu, China ) and hCG

    (0.075 IU/ ml) for 18 h.

    The purity was assessed by 3β-HSD immune-staining. In a separate experiment, Leydig cells

  • MAN

    USCR

    IPT

    ACCE

    PTED

    ACCEPTED MANUSCRIPT

    were pre-incubated with Que for 1 h and then treated with TP for 24 h.

    2.4 AlamarBlue assay

    Leydig cells were seeded in a 96-well black plate at a density of 2×104 cells/well.

    Twenty-four hours later, the cells were treated with control (0.1% DMSO) or different

    concentrations of TP. Meanwhile, the cells were pre-incubated with Que for 1 h and then treated

    with different concentrations of TP for 24 h. The viability of Leydig cells was assessed by the

    AlamarBlue Cell Viability Assay (Invitrogen, CA, USA) according to manufacturer’s instructions.

    2.5 Detection of apoptosis

    Leydig cells (1×106 cells/well) plated in each well of 6-well plates were treated with

    concentrations of TP for 24 h after pre-incubating with 5 µM Que for 1 h. Cell apoptosis was

    determined via FACS Calibur flow cytometer (Becton Dickinson, San Jose, CA, USA) using

    Annexin V-FITC/PI Apoptosis detection kit (Vazyme, Jiangsu, China) according to the

    manufacturer’s instructions.

    2.6 Measurement of antioxidant enzymes activity (SOD and GPx)

    Leydig cells were seeded in a 6-well plate at a density of 1×106 cells/well, and then exposed

    to TP for 24 h after pre-incubating with 5 µM Que for 1 h. Cells were lysed in RIPA buffer

    (Vazyme, Jiangsu, China) and centrifuged at 12,000 g for 10 min. Total protein of each sample

    was determined by the BCA protein assay (Beyotime, Haimen, Jiangsu, China). SOD or GPx

    activity was determined using a SOD or GPx Detection Kit (Beyotime, Haimen, Jiangsu, China)

    according to the manufacturer’s instructions.

    2.7 Measurement of Reactive Oxygen Species (ROS)

    DCFH-DA (SigmaAldrich, MO, USA), a lipophilic dye was used to determine the

  • MAN

    USCR

    IPT

    ACCE

    PTED

    ACCEPTED MANUSCRIPT

    intracellular accumulation of ROS. Leydig cells (2×104 cells/well) were seeded in 96-welll blank

    plate and exposed to TP for 24 h following pre-incubating with Que for 1 h. After that, cells were

    incubated with DCFH-DA (10 µM) for 30 min at 37 °C in dark, and then washed in 100 µL

    ice-cold PBS. Fluorescence at Excitation: 488nm and Emission: 520nm were measured in a

    microplate reader (safire2, Tecan, Switzerland).

    2.8 Measurement of mitochondrial membrane potential (∆Ψm)

    Changes of ∆Ψm were assessed by lipophilic and cationic probe JC-1 (JC-1 Detection Kit

    (Beyotime Biotech, Nantong, China)) according to the manufacturer’s instructions. Briefly, after

    different treatments, cells (2×104 cells/well) were cultured in 96-well blank plate and incubated

    with 10 mM JC-1 for 30 min in a 5% CO2 incubator at 37 °C. Subsequently, cells were washed

    twice with cold JC-1 Buffer. The cells were observed by a Fluorescence microscope (OLYMPUS,

    Japan) with a single excitation (488nm) and dual emission (shift from 530 nm to 590 nm).

    2.9 RNA isolation and RT-PCR analysis

    Total RNA was extracted from the treated cells by the TRIzol® reagent (Vazyme, Jiangsu,

    China). Genomic DNA contamination was removed by treatment of the total RNA with

    RNase-free DNase (Vazyme, Jiangsu, China). The RNA concentration and integrity were

    determined at 260 and 280 nm by a GeneQuant Pro spectrophotometer (Amersham Biosciences,

    USA). The total RNA (2 µg) was reverse transcribed to cDNA with HiScript® Reverse

    Transcription kit (Vazyme, Jiangsu, China) with Oligo-dT primers (Vazyme, Jiangsu, China)

    according to the manufacturer’s instructions. The target fragments quantified by real-time PCR

    using AceQTMSYBR Green® PCR Kit (Vazyme, Jiangsu, China) were performed on a iCycler

    iQ™5 Multicolor Reai-Time PCR Detection System (Bio-Rad, USA). The specific primers are

  • MAN

    USCR

    IPT

    ACCE

    PTED

    ACCEPTED MANUSCRIPT

    described in Table 1. For the quantification of real-time PCR results, the threshold cycle Ct was

    determined for each reaction. Ct values for each gene were normalized to the housekeeping gene

    (GAPDH), Relative expression levels of the target genes were calculated based on 2-∆∆Ct

    according to the manufacture's specifications.

    Table 1

    Primer sequences for PCR amplification.

    Gene Primer sequence

    Gpx1 Forward: 5'-CGGACATCAGGAGAATGGCA-3'

    Gpx1 Reverse: 5'-GTAAAGAGCGGGTGAGCCTT-3'

    Gpx4 Forward: 5'-GCCGTCTGAGCCGCTTATT-3'

    Gpx4 Reverse: 5'-CGATGTCCTTGGCTGCGAAT-3'

    SOD1 Forward: 5'-AGGGCGTCATTCACTTCGAG-3'

    SOD1 Reverse: 5'-TCTGCAAGTGCATCATCGTT-3'

    SOD2 Forward: 5'-GCCTCAGCAATGTTGTGTCG-3'

    SOD2 Reverse: 5'-ATTGTTCACGTAGGTCGCGT-3'

    GAPDH Forward: 5'-AGAACATCATCCCTGCATCCA-3'

    GAPDH Reverse: 5'-CCGTTCAGCTCTGGGATGAC-3'

    2.10 Western blot analysis

    Cells were lysed in RIPA buffer (Vazyme, Jiangsu, China) containing protease inhibitors

    (Vazyme, Jiangsu, China) to prepare the total protein fractions. The nuclear extracts were prepared

    using a Nuclear Extract kit (Vazyme, Jiangsu, China), and the mitochondrial proteins were

    prepared using a Cell Mitochondria Isolation Kit (Beyotime, Jiangsu, China) following the

    manufacturer’s instructions. Protein concentration was measured by the BCA protein assay. 20 µg

    protein sample was separated by 10% resolving SDS-PAGE and transferred to nitrocellulose

    membranes. The membranes were incubated with the appropriate primary antibodies at 4°C

  • MAN

    USCR

    IPT

    ACCE

    PTED

    ACCEPTED MANUSCRIPT

    overnight following incubated with 5% non-fat milk for 1h at room temperature. After incubating

    with HRP-conjugated secondary antibodies for 1 h at room temperature, the membranes were

    visualized using chemiluminescence HRP substrate in ChemiDoc XRS imaging system (Bio-Rad,

    USA).

    2.11 Statistical analysis

    The data were expressed as mean ± standard deviation (SD). The statistical value p < 0.05

    was considered statistically significant. Statistical analysis was done by two-way ANOVA using

    GraphPad Prism 5 software. Each experiment was performed in triplicate and replicated

    independently.

    3. Results

    3.1. Cytotoxicity of TP and Que pre-treatment countered the decrease in TP-induced Leydig cell

    viability and inhibited TP-induced apoptosis in Leydig cell

    To determine the cytotoxicity of TP in vitro, Leydig cells were treated with various

    concentrations of TP for 24 h, the cell viability was measured using AlamarBlueTM assay. As

    shown in Fig.1A, TP increased the viability of Leydig cells in a dose-dependent manner. Loss of

    cell viability showed 30% by 60 nM TP and reached up to 95% by 640 nM. Meanwhile, to

    determine the optimal dose of Que that countered the TP-induced decrease in Leydig cell viability,

    Leydig cells were co-incubated with different doses of Que and 60 nM TP (final 2.5, 5, 10, 20 µM)

    for 24 h. As shown in Fig.1B, compared with TP-treated cells, Que at 2.5 µM was not able to

    maintain a significant rise in cell viability. On the other hand, high doses (10 and 20 µM) of Que

    were not able to sustain the cell viability. Only 5 µM Que demonstrated the significant effect on

    cell survival. Based on this observation, 20, 40 and 60 nM TP concentration were selected for

  • MAN

    USCR

    IPT

    ACCE

    PTED

    ACCEPTED MANUSCRIPT

    general toxicity assessment impact on Leydig cells (Fig.1C). Further, 5 µM Que was used in

    subsequent experiments. The annexin V-FITC/PI double staining assay results showed that TP

    induced apoptosis in Leydig cells in a dose-dependent manner (Fig.1D). The TP induced apoptosis

    was significantly restored by pre-treatment with 5 µM Que (Fig.1E and F).

  • MAN

    USCR

    IPT

    ACCE

    PTED

    ACCEPTED MANUSCRIPT

  • MAN

    USCR

    IPT

    ACCE

    PTED

    ACCEPTED MANUSCRIPT

  • MAN

    USCR

    IPT

    ACCE

    PTED

    ACCEPTED MANUSCRIPT

    Fig.1. Inhibition of Que on TP-induced cytotoxicity in Leydig cell. Leydig cells were treated with

    TP at various concentrations or DMSO (0.1%) as control for 24 h (A). Leydig cells were

    co-incubated with 60 nM TP and various concentrations of Que (final 2.5, 5, 10, 20 µM) or

    DMSO (0.1%) as control for 24 h (B). Leydig cells were treated with various concentrations of TP

    (final 20, 40, 60 nM) with or without 5 µM Que for 24 h (C). Cell apoptotic was determined by

    annexin V-FITC/PI double staining assay. Leydig cells were treated with various concentrations of

    TP (final 20, 40, 60 nM) with or without 5 µM Que for 24 h (D and E). The percentage of cell

    apoptosis is shown in (F). Data are presented as the mean ± SD. “*” indicates significant

    difference between normal control and TP treated groups, “#” indicates significant difference

    between TP treated and Que pre-treated groups (*** P < 0.001, #P < 0.05, ###P < 0.001), n=3.

    3.2 Effects of Que on TP-induced decrease of antioxidant enzymes activities and generation of

    intracellular ROS

    In oxidative stress-induced organ pathophysiology, intracellular antioxidant enzymes are

    considered to be the first line of cellular defense as these enzymes protect biological

    macromolecules like DNA, proteins etc. from oxidative damage. In our study, we determined the

    activities of SOD and GPx in Leydig cells which treated with various concentrations of TP and

    pre-incubated with 5 µM Que separately. We observed that TP intoxication significantly decreased

    the SOD activity at concentration of 40 and 60 nM and GPx activity at concentration of 20, 40 and

    60 nM. However, compared to the TP-treated group, there were significantly increased the SOD

    and GPx activity in the Que pre-treated group (Fig. 2A and B). The results of gene expression

    were shown in Fig.2C to F. Meanwhile, TP exposure increased the accumulation of intracellular

    ROS and the increase was significantly restored by pre-treatment with 5 µM Que (Fig. 2G).

  • MAN

    USCR

    IPT

    ACCE

    PTED

    ACCEPTED MANUSCRIPT

  • MAN

    USCR

    IPT

    ACCE

    PTED

    ACCEPTED MANUSCRIPT

  • MAN

    USCR

    IPT

    ACCE

    PTED

    ACCEPTED MANUSCRIPT

  • MAN

    USCR

    IPT

    ACCE

    PTED

    ACCEPTED MANUSCRIPT

    Fig.2. Effects of Que on TP-induced decrease of antioxidant enzymes activities and generation of

    intracellular ROS. The activities (A and B) and expression (C to F) of GPx and SOD were

    measured. Leydig cells were treated with various concentrations of TP (final 20, 40, 60 nM) with

    or without 5 µM Que for 24 h. The ROS were detected using the fluorescent probe DCFH-DA (G).

    Data are presented as the mean ± SD. “*” indicates significant difference between normal control

    and TP treated groups, “#” indicates significant difference between TP treated and Que pre-treated

    groups (*P < 0.05, *** P < 0.001, #P < 0.05, ##P < 0.01, ###P < 0.001), n=3.

    3.3. Effects of Que on TP-induced loss of mitochondrial membrane potential and release of Cyt

    C

    To elucidate the direct effect of TP on Leydig cell mitochondrial membrane potential (∆Ψm),

    the ∆Ψm was measured after incubating with JC-1. As showed in Fig. 3A, TP induced ∆Ψm loss as

    a dose-dependent manner compared to control, which was confirmed by High Content Screening.

    Only 40 and 60 nM of TP significantly inhibited ∆Ψm. Meanwhile, TP caused the release of Cyt C

    from mitochondria into cytosol as a dose-dependent manner, which was regarded as a key step of

  • MAN

    USCR

    IPT

    ACCE

    PTED

    ACCEPTED MANUSCRIPT

    the mitochondria-mediated pathway in apoptosis (Fig. 3B and C). Treatment with 5 µM que prior

    to the TP exposure however could significantly inhibit TP induced alterations of these parameters.

    Fig.3. Effects of Que on TP-induced loss of mitochondrial membrane potential and release of

    Cyt-C. Leydig cells were treated with various concentrations of TP with or without 5 µM Que for

  • MAN

    USCR

    IPT

    ACCE

    PTED

    ACCEPTED MANUSCRIPT

    24 h. Representative pictures of JC-1 staining and quantitative analysis are shown. In control

    non-apoptotic cells, the dye stains the mitochondria in red. Otherwise the cytosol was shown green

    (A). CCCP was used as a positive control. Cyt-C protein expressed in the mitochondria and

    cytosol were determined (B and C). Data are presented as the mean ± SD. “*” indicates significant

    difference between normal control and TP treated groups, “#” indicates significant difference

    between TP treated and Que pre-treated groups (** P < 0.01, *** P < 0.001, ##P < 0.01, ###P <

    0.001), n=3.

    3.4 Que counteracts TP induced alteration mitochondrion-dependent apoptosis pathway related

    protein expression in Leydig cells

    To investigate the effects of Que on TP-induced changes of apoptosis-related proteins

    expression. The expression of BAX, Bcl-2, caspase-9 and caspase-3 were measured in Leydig

    cells (Fig.4A). TP decreased protein levels of the apoptosis inhibitory protein Bcl-2, and increased

    the expression levels of Bax, caspase-9 and caspase-3 (Fig.4B to E). In addition, pre-treatment

    with Que down-regulated the expression of Bax, caspase-9 and caspase-3 and up-regulated the

    Bcl-2 induced by TP.

  • MAN

    USCR

    IPT

    ACCE

    PTED

    ACCEPTED MANUSCRIPT

  • MAN

    USCR

    IPT

    ACCE

    PTED

    ACCEPTED MANUSCRIPT

  • MAN

    USCR

    IPT

    ACCE

    PTED

    ACCEPTED MANUSCRIPT

    Fig.4. Resist of Que to TP-induced alteration of apoptosis-related protein expression in Leydig

    cells. Cells were treated with various concentrations of TP (final 20, 40, 60 nM) with or without 5

    µM Que for 24 h. The levels of the apoptotic-related proteins Bcl-2, Bax, caspase-9 and caspase-3

    were analyzed (A to E). Data are presented as the mean ± SD. “*” indicates significant difference

    between normal control and TP treated groups, “#” indicates significant difference between TP

    treated and Que pre-treated groups (** P < 0.01, *** P < 0.001, #P < 0.05, ##P < 0.01, ###P < 0.001),

    n=3.

    3.5 Effect of TP on the level of Nrf2

    To confirm the role of Nrf2 in the TP-induced oxidative stress, the expression of Nrf2 were

    measured in Leydig cells. TP decreased protein levels of Nrf2, and pre-treatment with Que resisted

    this effect (Fig.5).

  • MAN

    USCR

    IPT

    ACCE

    PTED

    ACCEPTED MANUSCRIPT

    Fig.5. Effect of TP on the regulation of Nrf2. Leydig cells were treated with various

    concentrations of TP (final 20, 40, 60nM) with or without 5 µM Que for 24h. The levels of the

    Nrf2 protein was analyzed. Data are presented as the mean ± SD. “*” indicates significant

    difference between normal control and TP treated groups, “#” indicates significant difference

    between TP treated and Que pre-treated groups (*** P < 0.001, ###P < 0.001), n=3.

    4. Discussions

    TP is a diterpene triepoxide with variety biological activities. Generally, tripterygium and

    tripterygium glycosides pieces are the common forms for clinical application, in which TP is the

    major component. In addition of anti-rheumatoid, TP also has pharmacological activities like

    anti-cancerogenesis and anti-inflammatory [22]. However, the side effects of TP, especially its

  • MAN

    USCR

    IPT

    ACCE

    PTED

    ACCEPTED MANUSCRIPT

    reproduction toxicity, have not been well illustrated. In the present study, we used Leydig cell as a

    cellular model to explore the toxic effect of TP and the role of Que in defense against TP-induced

    male reproductive injury. In Leydig cells, TP induced testosterone secretion decline by lowering

    the expression of hormone synthesis enzyme and reproductive toxicity, which was caused by

    accumulating of ROS and cell apoptosis. Meanwhile, Que is proved to protect Leydig cells from

    TP-induced toxicity by the counteraction of oxidative damage.

    The role of ROS and its metabolites in cellular physiology and pathogenesis of number of

    diseases is the subject of today's research [23]. SOD is a scavenger of superoxide, is the most

    important to resist the toxic effects of ROS defense mechanism. SOD accelerates the disproportion

    of hydrogen peroxide, prevents further producing free radicals. GPx is antioxidant enzyme

    containing selenium, existing in the cell cytoplasm or plasma. The main function of these enzymes

    is to remove soluble hydrogen peroxide and alkyl peroxide by using of glutathione as the substrate

    [24]. Decline in SOD activity showed superoxide surplus accumulation. Reduced GPx activity

    showed H2O2 accumulation. TP had been reported its inhibition of the activity of SOD and GPx in

    various tissues of rats [25]. In the present study, we observed that the content of intracellular ROS

    significantly increased under TP exposure (Fig.2G), and the activity of the SOD and GPx both

    reduced (Fig.2A and B). Meanwhile, the transcription of SOD1, SOD2, GPx1 and GPx4 genes

    also showed a decrease (Fig.2C to F). The whole results performed as a dose-dependent manner.

    Thus, we demonstrated that TP could induce oxidative stress in Leydig cells in vitro.

    The interaction between oxidative stress and mitochondrial damage was revealed by large

    amounts of data [8]. Loss of ∆Ψm is a common index of mitochondrial damage which was

    resulted from the surplus generation of ROS. This phenomenon was shown in the present study

  • MAN

    USCR

    IPT

    ACCE

    PTED

    ACCEPTED MANUSCRIPT

    (Fig.3A), which confirmed the dysfunction of mitochondria. As an important enzyme inducing

    apoptosis, Cyt-C was released from mitochondria to cytoplasm due to the ROS-induced

    mitochondrial damage. The substantial increase of Cyt-C and loss of ∆Ψm (Fig.3) suggested that

    mitochondria played a critical role in TP-induced apoptosis.

    Various motivators including oxidative stress foment the mitochondrial apoptosis pathway.

    The Bcl-2 family proteins, including pro-apoptotic protein Bax and anti-apoptotic protein Bcl-2,

    are involved in the apoptotic pathway. Following the expose to TP, the expressions of Bax and

    Bcl-2 were increased and decreased separately (Fig.4B and C). In addition, the loss of Bax/Bcl-2

    ratio demonstrated that mitochondrial permeability transition might be a reason of Cyt-C release

    [26]. Caspase family, including caspase-3 and caspase-9 were significant elements mediated

    apoptosis [17]. The release of Cyt-C which would bind to Apaf-1, an apoptosis-related protein,

    could mediate activation of caspase-9, and then lead to cleavage of procaspase-3. Evidently, the

    treatment of TP increased the expression of caspase-9 and caspase-3 (Fig.4D and E). These results

    confirmed the effect of TP on apoptosis in Leydig cells.

    Exogenous antioxidants, such as flavonoids, could resist oxidative stress by several ways,

    major of which is scavenging or inhibiting the generation of ROS [27]. Que is one of the most

    common flavonoids in the diet, distributed in many vegetables, fruits and other foods [28]. Que is

    an effective antioxidant which can directly remove oxide free radicals, inhibit lipid peroxidation

    and change in vivo and in vitro antioxidant defense pathways [12]. Zhang [29] and Robaszkiewicz

    et al. [30] showed that Que reduced reactive oxygen species in human non-small cell lung cancer

    cell lines A549 and chickens testicular germ cells. Kalender et al.[31] and Payne and Halles[32]

    reported that Que could protect SOD, CAT, GPx activities from chlorpyrifos-induced the toxic

  • MAN

    USCR

    IPT

    ACCE

    PTED

    ACCEPTED MANUSCRIPT

    effect of ROS in rat testis tissue. However, the protective effect of Que on TP toxicity is

    little-known. The present study shows that a certain dose of Que had no adverse effects in Leydig

    cells (Fig.1B). We observed the effect of TP exposure on Leydig cells could be restored by Que.

    The protective effect of Que was achieved by treating prior to TP. Compared to TP group, ROS

    generation was inhibited significantly (Fig.2G) and the activities (Fig.2A and B) and expressions

    (Fig.2C to F) of SOD and GPx were up-regulated comparatively. Ascent of ∆Ψm was observed

    (Fig.3A) and the content of Cyt-C was down-regulated to the physiological level (Fig.3B and C).

    Apoptosis (Fig.1D and E) and apoptosis-related proteins expression (Fig.4) were also inhibited.

    All of these results could be explained based on the antioxidant effect of Que. In that, Que could

    protect Leydig cells from TP-induced oxidant damage.

    Nrf2 is anti-oxidant transcription factor leading a protection against oxidative stress by

    translocating into the nucleus to associate with AREs. The effect of TP to inhibit Nrf2 pathway

    has been demonstrated in heart tissues [33]. We hypothesized it is also the target of TP in Leydig

    cells. As shown in Fig.5, the expression of Nrf2 decreased obviously. This finding indicates a

    promising strategy to protect reproduction from TP toxicity in clinical treatment.

    The results of the present study suggest that TP induces mitochondrial apoptosis by

    decreasing ∆Ψm and the expression of related protein (bcl-2, caspase-9 and caspase-3) which is

    partly due to oxidative stress. As an effective antioxidant, Pre-treatment of Que can effectively

    alleviate the above toxic reaction caused by TP in Leydig cells.

    5. Conclusions

    To the best of our knowledge, this is the first report demonstrating that TP-induced oxidative

    stress is the cause of its toxic effect in Leydig cells and the impact of TP on the expression of Nrf2

  • MAN

    USCR

    IPT

    ACCE

    PTED

    ACCEPTED MANUSCRIPT

    was first observed in this experiment. On the other hand, we found that Que provides protection

    against TP-induces reproductive toxicity through its Antioxidant properties. Furthermore, Que acts

    an anti-apoptotic activity by down-regulating the expression of the mitochondrial apoptotic

    pathways related proteins. However, reproductive toxicity caused by TP involves not only single

    factor but multiple signaling pathways including JNK and Nrf2 pathway.

    Disclosure

    None.

    Conflict of interest

    The authors declare that are no Conflict of interest.

    Acknowledgements

    The work was supported by the Major Scientific and Technological Special Project for

    Significant New Drugs Creation (No. 2012ZX09504001-001), the National Natural Science

    Foundation of China (NO.81102876, 81430082),the Fundamental Research Funds for the Central

    Universities (ZL15005,YD2014SK0002), 333 high level project of Jiangsu Province.

    References

    [1] L. Wen, Y. Chen, L.L. Zeng, F. Zhao, R. Li, Y. Liu, C. Zhang, Triptolide induces cell-cycle arrest and

    apoptosis of human multiple myeloma cells in vitro via altering expression of histone demethylase

    LSD1 and JMJD2B, Acta Pharmacol. Sin., 33 (2012) 109-119.

    [2] X.H. Jiang, B.C. Wong, M.C. Lin, G.H. Zhu, H.F. Kung, S.H. Jiang, D. Yang, S.K. Lam, Functional p53 is

    required for triptolide-induced apoptosis and AP-1 and nuclear factor-kappaB activation in gastric

    cancer cells, Oncogene, 20 (2001) 8009-8018.

    [3] X.J. Li, Z.Z. Jiang, L.Y. Zhang, Triptolide: progress on research in pharmacodynamics and toxicology, J.

    Ethnopharmacol., 155 (2014) 67-79.

    [4] B. Ni, Z. Jiang, X. Huang, F. Xu, R. Zhang, Z. Zhang, Y. Tian, T. Wang, T. Zhu, J. Liu, L. Zhang, Male

    reproductive toxicity and toxicokinetics of triptolide in rats, Arzneimittel-Forsch, 58 (2008) 673-680.

    [5] N. Singla, S. Challana, Reproductive toxicity of triptolide in male house rat, Rattus rattus, The

    Scientific World J., 2014 (2014) 879405.

  • MAN

    USCR

    IPT

    ACCE

    PTED

    ACCEPTED MANUSCRIPT

    [6] R.J. Aitken, S.D. Roman, Antioxidant systems and oxidative stress in the testes, Oxid. Med. Cell.

    Longev., 1 (2008) 15-24.

    [7] M. Perri, A. Pingitore, E. Cione, E. Vilardi, V. Perrone, G. Genchi, Proliferative and anti-proliferative

    effects of retinoic acid at doses similar to endogenous levels in Leydig MLTC-1/R2C/TM-3 cells,

    Biochim. Biophys. Acta, 1800 (2010) 993-1001.

    [8] T.T. Turner, J.J. Lysiak, Oxidative stress: a common factor in testicular dysfunction, J Androl, 29

    (2008) 488-498.

    [9] I. Gulcin, Antioxidant activity of food constituents: an overview, Arch. Toxicol., 86 (2012) 345-391.

    [10] I. Gulcin, S. Beydemir, Phenolic compounds as antioxidants: carbonic anhydrase isoenzymes

    inhibitors, Mini-Rev Med. Chem., 13 (2013) 408-430.

    [11] T. Nguyen, C.S. Yang, C.B. Pickett, The pathways and molecular mechanisms regulating Nrf2

    activation in response to chemical stress, Free Radical Bio. Med., 37 (2004) 433-441.

    [12] M. Anjaneyulu, K. Chopra, Quercetin, an anti-oxidant bioflavonoid, attenuates diabetic

    nephropathy in rats, Clin. Exp. Pharmacol. P., 31 (2004) 244-248.

    [13] M. Erden Inal, A. Kahraman, The protective effect of flavonol quercetin against ultraviolet a

    induced oxidative stress in rats, Toxicology, 154 (2000) 21-29.

    [14] E.T. Olayinka, A. Ore, O.S. Ola, O.A. Adeyemo, Protective effect of quercetin on

    melphalan-induced oxidative stress and impaired renal and hepatic functions in rat, Chemother. Res.

    Pract., 2014 (2014) 936526.

    [15] Y. Mi, C. Zhang, C. Li, S. Taneda, G. Watanabe, A.K. Suzuki, K. Taya, Quercetin protects embryonic

    chicken spermatogonial cells from oxidative damage intoxicated with 3-methyl-4-nitrophenol in

    primary culture, Toxicol. Lett., 190 (2009) 61-65.

    [16] H. Tsutsui, S. Kinugawa, S. Matsushima, Oxidative stress and mitochondrial DNA damage in heart

    failure, Circ. J., 72 Suppl A (2008) A31-37.

    [17] B. Baliga, S. Kumar, Apaf-1/cytochrome c apoptosome: an essential initiator of caspase activation

    or just a sideshow?, Cell Death Differ., 10 (2003) 16-18.

    [18] D.M. Stocco, X. Wang, Y. Jo, P.R. Manna, Multiple signaling pathways regulating steroidogenesis

    and steroidogenic acute regulatory protein expression: more complicated than we thought,

    Mol.Endocrinol., 19 (2005) 2647-2659.

    [19] M.M. Janjic, N.J. Stojkov, S.A. Andric, T.S. Kostic, Anabolic-androgenic steroids induce apoptosis

    and NOS2 (nitric-oxide synthase 2) in adult rat Leydig cells following in vivo exposure, Reprod Toxicol,

    34 (2012) 686-693.

    [20] J. Yao, Z. Jiang, W. Duan, J. Huang, L. Zhang, L. Hu, L. He, F. Li, Y. Xiao, B. Shu, C. Liu, Involvement of

    mitochondrial pathway in triptolide-induced cytotoxicity in human normal liver L-02 cells, Biol. Pharm.

    Bull., 31 (2008) 592-597.

    [21] A. Steinberger, G. Klinefelter, Sensitivity of Sertoli and Leydig cells to xenobiotics in in vitro models,

    Reprod. Toxicol., 7 Suppl 1 (1993) 23-37.

    [22] Q. Liu, Triptolide and its expanding multiple pharmacological functions, Int. Immunopharmacol.,

    11 (2011) 377-383.

    [23] P.S. Azevedo, D.R. Duarte, M.F. Minicucci, B.B. Matsubara, L.S. Matsubara, R. Novo, E.L. Novelli,

    A.O. Campana, S.A. Paiva, L.A. Zornoff, Role of lipoperoxidation in the remodeling intensification

    induced by beta-carotene after infarction, Arq. Bras. Cardiol., 93 (2009) 34-38.

    [24] F.N. Bebe, M. Panemangalore, Exposure to low doses of endosulfan and chlorpyrifos modifies

    endogenous antioxidants in tissues of rats, J. Environ. Sci. Health B, 38 (2003) 349-363.

  • MAN

    USCR

    IPT

    ACCE

    PTED

    ACCEPTED MANUSCRIPT

    [25] F. Yang, L. Ren, L. Zhuo, S. Ananda, L. Liu, Involvement of oxidative stress in the mechanism of

    triptolide-induced acute nephrotoxicity in rats, Exp. Toxicol. Pathol., 64 (2012) 905-911.

    [26] D.G. Breckenridge, D. Xue, Regulation of mitochondrial membrane permeabilization by BCL-2

    family proteins and caspases, Curr. Opin. Cell Biol., 16 (2004) 647-652.

    [27] K. Bhatt, S.J. Flora, Oral co-administration of alpha-lipoic acid, quercetin and captopril prevents

    gallium arsenide toxicity in rats, Environ. Toxicol. Pathol., 28 (2009) 140-146.

    [28] F.G. Uzun, F. Demir, S. Kalender, H. Bas, Y. Kalender, Protective effect of catechin and quercetin on

    chlorpyrifos-induced lung toxicity in male rats, Food. Chem. Toxicol., 48 (2010) 1714-1720.

    [29] Y.M. Zhang, Protective effect of quercetin on aroclor 1254-induced oxidative damage in cultured

    chicken spermatogonial cells, Toxicol. Sci., 88 (2005) 545-550.

    [30] A. Robaszkiewicz, A. Balcerczyk, G. Bartosz, Antioxidative and prooxidative effects of quercetin on

    A549 cells, Cell Bio. Int. , 31 (2007) 1245-1250.

    [31] Y. Kalender, S. Kaya, D. Durak, F.G. Uzun, F. Demir, Protective effects of catechin and quercetin on

    antioxidant status, lipid peroxidation and testis-histoarchitecture induced by chlorpyrifos in male rats,

    Environ. Toxicol. Pathol., 33 (2012) 141-148.

    [32] A.H. Payne, D.B. Hales, Overview of steroidogenic enzymes in the pathway from cholesterol to

    active steroid hormones, Endocr Rev, 25 (2004) 947-970.

    [33] J. Zhou, C. Xi, W. Wang, X. Fu, L. Jinqiang, Y. Qiu, J. Jin, J. Xu, Z. Huang, Triptolide-induced oxidative

    stress involved with Nrf2 contribute to cardiomyocyte apoptosis through mitochondrial dependent

    pathways, Toxicol. Lett., 230 (2014) 454-466.

  • MAN

    USCR

    IPT

    ACCE

    PTED

    ACCEPTED MANUSCRIPT

    HIGHLIGHTS

    • Reproductive toxicity induced by Triptolide in vivo were due to

    oxidative stress in Leydig cells.

    • Protective effect of Quercetin on oxidative stress and Apoptosis

    induced by Triptolide in Leydig cells.

  • 本文献由“学霸图书馆-文献云下载”收集自网络,仅供学习交流使用。

    学霸图书馆(www.xuebalib.com)是一个“整合众多图书馆数据库资源,

    提供一站式文献检索和下载服务”的24 小时在线不限IP

    图书馆。

    图书馆致力于便利、促进学习与科研,提供最强文献下载服务。

    图书馆导航:

    图书馆首页 文献云下载 图书馆入口 外文数据库大全 疑难文献辅助工具

    http://www.xuebalib.com/cloud/http://www.xuebalib.com/http://www.xuebalib.com/cloud/http://www.xuebalib.com/http://www.xuebalib.com/vip.htmlhttp://www.xuebalib.com/db.phphttp://www.xuebalib.com/zixun/2014-08-15/44.htmlhttp://www.xuebalib.com/

    Protection of Quercetin against Triptolide-induced apoptosis by suppressing oxidative stress in rat Leydig cells.学霸图书馆link:学霸图书馆