BPDE and B[a]P induce mitochondrial compromise by ROS-mediated suppression of the SIRT1/TERT/PGC-1α pathway in spermatogenic cells both in vitro and in vivo

https://doi.org/10.1016/j.taap.2019.05.004Get rights and content

Highlights

  • Mitochondria are the target of male reproductive toxicity induced by BPDE/B[a]P.

  • BPDE and B[a]P induce mitochondrial compromise in spermatogenic cells.

  • SIRT1/TERT/PGC-1α pathway accounts for BPDE/B[a]P-induced mitochondrial damage.

  • BPDE/B[a]P-induced ROS causes the suppression of SIRT1/TERT/PGC-1α pathway.

Abstract

There is increasing evidence that indicates benzo[a]pyrene (B[a]P) and its active metabolite benzo[a]pyrene-7, 8-dihydrodiol-9, 10-epoxide (BPDE) are endocrine disruptors that can cause reproductive toxicity. Nevertheless, the underlying mechanisms are still obscure. The present study investigates the impacts of B[a]P and BPDE on mitochondria, a sensitive target affected by multiple chemicals, in spermatogenic cells. It showed that BPDE treatment induced mitochondrial dysfunction and the inhibition of mitochondrial biogenesis in mouse spermatocyte-derived cells (GC-2). These effects were efficiently mitigated by pretreatment with ZLN005, an activator of PGC-1α, in GC-2 cells. TERT knockdown and re-expression cell models were established to demonstrate that TERT regulated the BPDE-induced mitochondrial damage via PGC-1α signaling in GC-2 cells. Moreover, upregulating or knockdown SIRT1 expression attenuated or aggravated BPDE-induced mitochondrial compromise by activating or inhibiting, respectively, the TERT and PGC-1α molecules in GC-2 cells. Finally, we observed that BPDE markedly elevated oxidative stress in GC-2 cells. Resveratrol and N-acetylcysteine, as reactive oxygen species (ROS) scavengers, attenuated BPDE-mediated mitochondrial damage by increasing SIRT1 activity and expression in GC-2 cells. The in vitro results were corroborated by in vivo experiments in rats treated with B[a]P for 4 weeks. B[a]P administration caused mitochondrial damage and mitochondria-dependent apoptosis in spermatogenic cells, as well as the decreased expression of SIRT1, TERT, and PGC-1α. In summary, the results of the present study demonstrate that B[a]P and BPDE induce mitochondrial damage through ROS production that suppresses SIRT1/TERT/PGC-1a signaling and mediate B[a]P- and BPDE-mediated reproductive toxicity.

Introduction

Benzo[a]pyrene (B[a]P), as a ubiquitous and extensively studied polycyclic aromatic hydrocarbon (PAH), is mainly generated by the incomplete combustion of organic material (Yin et al., 2017). Besides well-documented carcinogenicity and mutagenicity, growing evidence indicates that B[a]P is a representative endocrine disruptor that may exert notable male reproductive toxicity (Banerjee et al., 2016; el SA et al., 2010). Previous epidemiological research efforts have confirmed the production of B[a]P-DNA adducts in human sperm and suggested that PAHs exposure in humans is associated with reduction in semen quality that might result in a higher risk of infertility (Oliveri Conti et al., 2017; Han et al., 2011). B[a]P following oral administration in rats can be metabolized in reproductive tissues, such as testis, and get transformed into various metabolites by cytochrome P450 enzymes, including the most-studied metabolic product benzo[a]pyrene-7, 8-dihydrodiol-9, 10-epoxide (BPDE) which has been identified as an active metabolite that can cause reproductive damage (Ramesh et al., 2001; Sipinen et al., 2010). Our previous study has implied that sub-chronic exposure to B[a]P results in the atrophy of seminiferous epithelium and decreases sperm count in male Sprague Dawley rats (Chen et al., 2011). Other animal studies have also revealed that exposure to B[a]P causes a significant decline in sperm count and motility (el SA et al., 2010; Meier et al., 2017). These particular evidences demonstrate that B[a]P can induce appreciable spermatogenic disorder, which may then exert further adverse effects on male reproductive health.

Mitochondria are eukaryotic organelles that produce energy through the Krebs cycle and the electron transport chain that then can result in the synthesis of adenosine triphosphate (ATP). Previous in vivo and in vitro research efforts have revealed that exposure to B[a]P as well as BPDE can induce obvious mitochondrial compromise in somatic cells (Zha et al., 2017; Zhang et al., 2015). Moreover, mitochondria in spermatogenic cells play importance roles in spermatogenesis and male fertility (Rajender et al., 2010). A previous cross-sectional study conducted by our laboratory found a significant relationship between urinary concentrations of PAH-metabolites and decreased mitochondrial DNA copy number (mtDNAcn) in sperm, which implied that mitochondria might be the target organelle involved in the B[a]P-induced spermatogenic disorder (Ling et al., 2017). However, the precise cellular and biochemical mechanisms by which B[a]P causes mitochondrial damage in spermatogenic cells and triggers spermatogenic disorder have not yet been identified.

The peroxisome proliferator-activated receptor gamma co-activator 1-alpha (PGC-1α), as a transcriptional coactivator, fundamentally regulates mitochondrial biogenesis as well as the mitochondrial function. Inactivated PGC-1α remains in its acetylated form in cells and is deacetylated by silent information regulator type-1 (SIRT1), which generates the active form of PGC-1α. Deacetylated PGC-1α cooperates with the nuclear respiratory factors (NRFs), such as NRF1, to trigger expression of the mitochondrial transcriptional factor A (Tfam) and various nuclear-encoded genes (Dorn 2nd et al., 2015). Besides SIRT1-mediated regulation, PGC-1α can also be regulated by telomerase reverse transcriptase (TERT), indeed a critical component of telomerases (Sahin et al., 2011). Telomerase, including the TERT and the telomerase RNA, is an RNA-containing reverse transcriptase that adds repeats of telomeric DNA to the ends of the chromosomes, which then cap and protect the integrity of the chromosomal ends.

Previous studies indicate that TERT deficiency causes telomeres dysfunction, which inhibits PGC-1α-mediated mitochondrial biogenesis and elevates oxidative stress through the activation of p53 (Sahin et al., 2011). Our study also previously reported that B[a]P exposure decreased the expression and activity of TERT, which caused senescence and apoptosis via the DNA damage response in the spermatogenic cells in vitro and in vivo (Ling et al., 2018). However, whether suppression of TERT is involved in B[a]P-induced reproductive damage via regulation of PGC-1α remains still largely unknown.

Mitochondria are the primary source of cellular ROS production (Yue et al., 2016). Although reactive oxygen species (ROS) is necessary for several vital signaling pathways to maintain physiologic homeostasis (Dostert et al., 2008; Tschopp and Schroder, 2010), the excessive production of ROS is closely related to the inhibition of mitochondrial biogenesis and mitochondrial dysfunction (Murphy, 2013; Soeur et al., 2015). Several studies have reported that exposure to B[a]P induces incremental ROS production both in vivo and in vitro (Banerjee et al., 2016; Omidian et al., 2017; Yang et al., 2017), and oxidative stress can decrease the expression of SIRT1 and agonism of SIRT1 through resveratrol or SIRT1 reconstitution can mitigate ROS-induced damages (Lin et al., 2018; Kerksick and Willoughby, 2005). Thus, further investigations are needed to elucidate whether excessive ROS production can cause mitochondrial compromise through a SIRT1 signal in B[a]P-induced male reproductive damage.

We hypothesized that mitochondrion is the sensitive targeted organelle that is involved in B[a]P-mediated spermatogenic cell damage. Meanwhile, B[a]P-induced mitochondrial compromise may be closely related to the alterations of SIRT1, TERT, and PGC-1α that can be regulated by elevated oxidative stress. To test this hypothesis, we examined mitochondrial damage in mouse spermatocyte-derived GC-2 spd (ts) cells (GC-2 cells) and Sprague-Dawley rats treated with BPDE and B[a]P, respectively. We also modified the ROS level as well as the molecules mentioned above to further ascertain they are involved in B[a]P-induced mitochondrial compromise and male reproductive toxicity.

Section snippets

Chemicals and antibodies

We purchased B[a]P, N-acetylcysteine (NAC), and dimethylsulfoxide (DMSO) from the Sigma-Aldrich Chemical Company (St. Louis, MO, USA); BPDE from the Midwest Research Institute (Kansas City, MO, USA); ZLN005 and CAY10602 from MedChemExpress (MedChemExpress, USA); and resveratrol from the Sangon Biotechnology Company (Shanghai, China). The following commercially available antibodies were purchased from Abcam (Cambridge, UK) and used in the present study: TERT, SIRT1, PGC-1α, NRF1, Cytochrome C

BPDE induces mitochondrial dysfunction and mitochondria-dependent apoptosis in GC-2 cells

A JC-1 fluorescent probe was used to measure the MMP, a crucial indicator of mitochondrial function. Treatment of GC-2 cells with 50 nM, 100 nM, and 200 nM BPDE for 72 h significantly reduced MMP (Fig. 1A). Cox IV is a critical enzyme in the mitochondrial oxidative phosphorylation for ATP generation in eukaryotes. Cox IV and cellular levels of ATP in the GC-2 cells were thus measured by immunoblot analysis and an ATP Determination Kit, respectively. The results showed that the Cox IV protein

Discussion

We previously found that BPDE treatment for 72 h at concentrations of 50, 100, and 200 nM induced appreciable senescence and apoptosis via TERT-mediated DNA damage response in GC-2 cells. Spermatogenic cell apoptosis and testicular damage were also observed in male Sprague-Dawley rats upon B[a]P administration at doses of 5, 10, and 20 mg/kg for 7 days (Ling et al., 2018). Another previous study from our laboratory revealed that TERT expression was significantly decreased in testicular cells of

Conflicts of interest

The authors declare that there are no conflicts of interest.

Acknowledgments

This work was supported by the National Key Research and Development Plan (2017YFC1600202); and the National Natural Science Foundation of China (NO.81273105).

References (41)

  • K. Omidian et al.

    Polyphenol inhibition of benzo[a]pyrene-induced oxidative stress and neoplastic transformation in an in vitro model of carcinogenesis

    Food Chem. Toxicol.

    (2017 Aug)
  • S. Rajender et al.

    Mitochondria, spermatogenesis and male infertility

    Mitochondrion

    (2010 Aug)
  • A. Ramesh et al.

    Metabolism, bioavailability, and toxicokinetics of benzo(alpha)pyrene in F-344 rats following oral administration

    Exp. Toxicol. Pathol.

    (2001 Sep)
  • J. Soeur et al.

    Skin resistance to oxidative stress induced by resveratrol: from Nrf2 activation to GSH biosynthesis

    Free Radic. Biol. Med.

    (2015 Jan)
  • K. Sumii et al.

    Characterization of urocortin as an anti-apoptotic protein in experimental ischemia-reperfusion model of the rat testis

    Biochem. Biophys. Res. Commun.

    (2016 Oct 14)
  • T. Wenz

    Regulation of mitochondrial biogenesis and PGC-1alpha under cellular stress

    Mitochondrion

    (2013 Mar)
  • S. Yin et al.

    Environmental exposure to polycyclic aromatic hydrocarbons (PAHs): the correlation with and impact on reproductive hormones in umbilical cord serum

  • J. Zha et al.

    Benzo(a)pyrene-induced a mitochondria-independent apoptosis of liver in juvenile Chinese rare minnows (Gobiocypris rarus)

    Environ. Pollut. (Barking, Essex : 1987).

    (2017 Dec)
  • A. Brunet et al.

    Stress-dependent regulation of FOXO transcription factors by the SIRT1 deacetylase

    Science

    (2004 Mar 26)
  • J.H. Cho et al.

    Sirtuin signaling controls mitochondrial function in glycogen storage disease type Ia

    J. Inherit. Metab. Dis.

    (2018 May 8)
  • Cited by (25)

    • Benzo[a]pyrene inhibits testosterone biosynthesis via NDUFA10-mediated mitochondrial compromise in mouse Leydig cells: Integrating experimental and in silico toxicological approaches

      2022, Ecotoxicology and Environmental Safety
      Citation Excerpt :

      Each cell suspension was diluted to a concentration of 1 × 105 cells/ml. NAO fluorescence intensity for each cell suspension was detected in triplicate using a multimode microplate reader (Varioskan®LUX, Thermo Scientific, USA) (Ex/Em 488/525 nm) as previously described (Yang et al., 2019). Immunofluorescence assay was performed to assess colocalization of mitochondria and LC3B according to the standard protocols.

    View all citing articles on Scopus
    View full text