Elsevier

Chemico-Biological Interactions

Volume 256, 25 August 2016, Pages 142-153
Chemico-Biological Interactions

Aldo-keto reductase 1B10 promotes development of cisplatin resistance in gastrointestinal cancer cells through down-regulating peroxisome proliferator-activated receptor-γ-dependent mechanism

https://doi.org/10.1016/j.cbi.2016.07.008Get rights and content

Highlights

  • AKR1B10 is up-regulated with CDDP resistance in gastrointestinal cancer cells.

  • PPARγ is down-regulated in the CDDP-resistant cells.

  • AKR1B10 detoxifies reactive aldehydes and suppresses PPARγ expression.

  • Treatment with AKR1B10 inhibitor and PPARγ ligand overcomes the CDDP resistance.

Abstract

Cisplatin (cis-diamminedichloroplatinum, CDDP) is one of the most effective chemotherapeutic drugs that are used for treatment of patients with gastrointestinal cancer cells, but its continuous administration often evokes the development of chemoresistance. In this study, we investigated alterations in antioxidant molecules and functions using a newly established CDDP-resistant variant of gastric cancer MKN45 cells, and found that aldo-keto reductase 1B10 (AKR1B10) is significantly up-regulated with acquisition of the CDDP resistance. In the nonresistant MKN45 cells, the sensitivity to cytotoxic effect of CDDP was decreased and increased by overexpression and silencing of AKR1B10, respectively. In addition, the AKR1B10 overexpression markedly suppressed accumulation and cytotoxicity of 4-hydroxy-2-nonenal that is produced during lipid peroxidation by CDDP treatment, suggesting that the enzyme acts as a crucial factor for facilitation of the CDDP resistance through inhibiting induction of oxidative stress by the drug. Transient exposure to CDDP and induction of the CDDP resistance decreased expression of peroxisome proliferator-activated receptor-γ (PPARγ) in MKN45 and colon cancer LoVo cells. Additionally, overexpression of PPARγ in the cells elevated the sensitivity to the CDDP toxicity, which was further augmented by concomitant treatment with a PPARγ ligand rosiglitazone. Intriguingly, overexpression of AKR1B10 in the cells resulted in a decrease in PPARγ expression, which was recovered by addition of an AKR1B10 inhibitor oleanolic acid, inferring that PPARγ is a downstream target of AKR1B10-dependent mechanism underlying the CDDP resistance. Combined treatment with the AKR1B10 inhibitor and PPARγ ligand elevated the CDDP sensitivity, which was almost the same level as that in the parental cells. These results suggest that combined treatment with the AKR1B10 inhibitor and PPARγ ligand is an effective adjuvant therapy for overcoming CDDP resistance of gastrointestinal cancer cells.

Introduction

Cisplatin (cis-diamminedichloroplatinum, CDDP), a first member of platinum-based chemotherapy drugs, is widely used to treat various types of cancer, such as malignant lymphomas and lung, gastrointestinal, esophageal, ovarian, bladder, prostate and cervical cancers. The platinum drug interferes with growth of the cancer cells through intra- and inter-strand crosslinking of the DNA helix and the resulting inhibition of replication and synthesis of nucleic acids, hereby leading to the cancer cell death [1], [2]. Besides inhibiting the cancer cell proliferation, CDDP is considered to damage the cancer cells by the enhanced formation of reactive oxygen species (ROS), which trigger apoptosis towards cancer cells through DNA injury, mitochondrial dysfunction, activation of p53- and caspase-signaling cascades, and cell cycle arrest [2], [3], [4]. Despite the potent anticancer action of CDDP, the continuous exposure easily gives rise to hyposensitivity to the drug, so-called chemoresistance, as well as onset and development of severe side-effects including nephrotoxicity [4]. Induction of chemoresistance in cancer cells limits efficacy of the anticancer drug and consequently imposes complexed multidrug therapy on the cancer patients. In many cases, it also forms the incurable malignant cancer cells featuring overgrowth and elevation of invasive and metastatic capacities. Therefore, it is hoped that novel medicine to inhibit function and/or expression of molecules requisite for the gain of CDDP resistance is developed. Mechanisms of chemoresistance including cellular uptake and efflux of CDDP and potentiation of the DNA-repair and cytoprotective capacity have been suggested so far [2], [5], [6]. In addition, caudal type homeobox 2 [7], autophagy-related gene-5 [8] and syndecan-1 [9] have been more recently proposed as the target molecules for developing the drug resistance.

Members of the aldo-keto reductase (AKR) superfamily are NAD(P)(H)-dependent oxidoreductases that metabolize carbohydrates, steroids, and prostaglandins, as well as exogenous carbonyl compounds such as drugs and toxicants [10]. Among human 15 members of this superfamily, AKR1C1, AKR1C2 and AKR1C3 have been shown to be highly expressed and associated with resistance against anticancer drugs such as anthracyclines, CDDP and methotrexate [11], [12], [13]. The three AKR1C isoforms are hydroxysteroid dehydrogenases with broad substrate specificity for carbonyl compounds including 4-hydroxy-2-nonenal (HNE), a major product of lipid peroxidation through oxidative stress, but it remains unclear as to how the enzymes confer the drug resistance. In addition, AKR1B10, i.e. human aldose reductase-like protein, is up-regulated in many types of solid tumors [14], [15], [16], [17], [18], and its gene silencing inhibits replication of colorectal cancer cells [19] and tumor growth in vivo [18]. Although detailed mechanisms of the AKR1B10-initiated tumorigenic action are still unclear, it is inferred to be due to metabolizing retinoids [20] and isoprenoids [21], regulating fatty acid synthesis [22], and detoxifying cytotoxic aldehydes such as acrolein [19] and HNE [23]. Thus, AKR1B10 has been recognized as a potential molecular target for preventing cancer proliferation as well as a diagnostic tumor marker. Besides, AKR1B10 was found to be overexpressed in medulloblastoma cell lines resistant to cyclophosphamide [24]. We also showed that mitomycin C (MMC) [25] or doxorubicin (DOX) resistance in gastrointestinal cancer [26] as well as CDDP resistance in lung cancer cells [27] are mediated by up-regulation of AKR1B10, suggesting a close association of the enzyme up-regulation with cancer chemoresistance.

Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors that belong to the nuclear receptor superfamily [28]. PPARs regulate transcription of target genes by binding to their peroxisome proliferator-response elements as heterodimers with retinoid X receptor. The PPAR receptor family is comprised of the three subtypes, PPARα, PPARβ/δ, and PPARγ, each of which mediates important physiological actions in metabolism of lipids and sugars. In addition to the energy homeostasis, ligands for PPARs are considered to act as key regulators for proliferation of gastrointestinal cancer cells. For example, treatment with ligands of PPARα and PPARγ inhibits proliferation of the cancer cells [29], [30]. Especially, PPARγ is closely linked to growth suppression and death of cancer cells, and incubation with the ligands appears to potently induce apoptosis in many kinds of cancer cells such as breast, thyroid, bladder, esophageal, gastric, colon, lung, and prostate cancers and lymphomas [31]. In addition, PPARγ is also associated with CDDP resistance of ovarian cancer [32], [33], bladder cancer [34] and malignant glioma [35], and that treatment with its ligands sensitizes the cancer cells to the drug. Furthermore, AKR1C3 is reported to facilitate proliferation of cancer cells such as myeloid leukemia cell and skin squamous cell carcinoma via metabolizing prostaglandin D2 into prostaglandin F, not 15-deoxy-Δ12,14-prostaglandin J2, an endogenous PPARγ agonist [36], [37], [38], [39], [40]. However, it is uncertain whether the PPARγ-dependent mechanism is involved in acquisition of CDDP resistance and expression of other AKRs, except for AKR1C3, in gastrointestinal cancer cells.

In this study, we have generated CDDP-resistant variants of gastrointestinal cancer (MKN45 and LoVo) cells, monitored alterations in factors involved in antioxidant response of the cells with development of CDDP resistance, and found a significant up-regulation of AKR1B10 as a pivotal process for promoting the chemoresistance. We have also investigated an association of PPARγ with the CDDP resistance, and finally evaluated efficacy of combined treatment with AKR1B10 inhibitor and PPARγ ligand for overcoming resistance in the resistant cells.

Section snippets

Materials

CDDP was purchased from Wako Pure Chemical Industries (Osaka, Japan). TRIzol reagent, Superscript III reverse transcriptase, oligo (dT)12-18 primer and Lipofectamine 2000 were obtained from Invitrogen (Carlsbad, CA); rosiglitazone, N-acetyl Asp-Glu-Val-Asp-7-amido-4-methylcoumarin (AMC), N-succinyl-Leu-Leu-Val-Tyr-AMC and N-acetyl-l-cysteine (NAC) were from Sigma-Aldrich (St. Louis, MO); (S)-(+)-1,2,3,4-tetrahydro-1-naphthol (S-tetralol) and oleanolic acid (OA) were from Tokyo Chemical Industry

Facilitation of antioxidant properties with CDDP resistance

Treatment of MKN45 cells for 24 h with CDDP provoked the viability loss, which was remarkable at its concentrations of more than 50 μM (Fig. 1A). In addition, the high concentrations of CDDP activated caspase-3, a downstream target for the ROS-dependent apoptotic signaling cascade [48] (Fig. 1B). Moreover, concomitant treatment with an antioxidant NAC significantly reduced the cellular damage by CDDP, although the inhibitory effect of the antioxidant is incomplete. The results clearly indicate

Discussion

In the present study, we showed that resistance development of gastrointestinal cancer cells against CDDP elevates the antioxidant capacity through increases in glutathione content, AKR expressions and 26S proteasome-based proteolytic activity. Since up-regulation of AKR1B10 was the most remarkable event among the alterations in the antioxidant factors (Fig. 2, Fig. 3), we highlight the importance of AKR1B10 as a crucial factor for acquiring CDDP resistance of gastrointestinal cancer cells.

Conflict of interest

All authors declare that there are no conflicts of interest concerning this work.

Acknowledgement

This work is supported in part by a Grant-in-Aid for Scientific Research (C) (25460068 and 15K10601) from the Japan Society for the Promotion of Science.

References (63)

  • C.L. Doig et al.

    Knockdown of AKR1C3 exposes a potential epigenetic susceptibility in prostate cancer cells

    J. Steroid Biochem. Mol. Biol.

    (2016)
  • N. Hevir et al.

    Decreased levels of AKR1B1 and AKR1B10 in cancerous endometrium compared to adjacent non-cancerous tissue

    Chem. Biol. Interact.

    (2013)
  • B.H. Rovin et al.

    15-Deoxy-Δ12,14-prostaglandin J2 regulates mesangial cell proliferation and death

    Kidney Int.

    (2002)
  • M. Soda et al.

    Design, synthesis and evaluation of caffeic acid phenethyl ester-based inhibitors targeting a selectivity pocket in the active site of human aldo-keto reductase 1B10

    Eur. J. Med. Chem.

    (2012)
  • S. Endo et al.

    Kinetic studies of AKR1B10, human aldose reductase-like protein: endogenous substrates and inhibition by steroids

    Arch. Biochem. Biophys.

    (2009)
  • T. Matsunaga et al.

    Nitric oxide mitigates apoptosis in human endothelial cells induced by 9,10-phenanthrenequinone: role of proteasomal function

    Toxicology

    (2010)
  • M.E. Anderson

    Determination of glutathione and glutathione disulfide in biological samples

    Methods Enzymol.

    (1985)
  • T. Matsunaga et al.

    Ceramide-induced intracellular oxidant formation, iron signaling, and apoptosis in endothelial cells: protective role of endogenous nitric oxide

    J. Biol. Chem.

    (2004)
  • T. Matsunaga et al.

    Pathophysiological roles of aldo-keto reductases (AKR1C1 and AKR1C3) in development of cisplatin resistance in human colon cancers

    Chem. Biol. Interact.

    (2013)
  • S. Cho et al.

    Protective role of glutathione synthesis in response to oxidized low density lipoprotein in human vascular endothelial cells

    Free Radic. Biol. Med.

    (1999)
  • J.Y. Chan et al.

    Impaired expression of glutathione synthetic enzyme genes in mice with targeted deletion of the Nrf2 basic-leucine zipper protein

    Biochim. Biophys. Acta

    (2000)
  • A.M. Pickering et al.

    Nrf2-dependent induction of proteasome and Pa28αβ regulator are required for adaptation to oxidative stress

    J. Biol. Chem.

    (2012)
  • J. Alam et al.

    Nrf2, a Cap’n’Collar transcription factor, regulates induction of the heme oxygenase-1 gene

    J. Biol. Chem.

    (1999)
  • Y. Zhang et al.

    All-trans retinoic acid potentiates the chemotherapeutic effect of cisplatin by inducing differentiation of tumor initiating cells in liver cancer

    J. Hepatol.

    (2013)
  • N. Najafzadeh et al.

    Low-dose all-trans retinoic acid enhances cytotoxicity of cisplatin and 5-fluorouracil on CD44(+) cancer stem cells

    Biomed. Pharmacother.

    (2015)
  • Y.T. Chung et al.

    Overexpression and oncogenic function of aldo-keto reductase family 1B10 (AKR1B10) in pancreatic carcinoma

    Mod. Pathol.

    (2012)
  • H. Baruah et al.

    Platinum-intercalator conjugates: from DNA-targeted cisplatin derivatives to adenine binding complexes as potential modulators of gene regulation

    Curr. Top. Med. Chem.

    (2004)
  • G. Wang et al.

    Molecular basis of cellular response to cisplatin chemotherapy in non-small cell lung cancer

    Oncol. Rep.

    (2004)
  • M.K. Choi et al.

    Platinum transporters and drug resistance

    Arch. Pharm. Res.

    (2006)
  • L.H. Yan et al.

    Overexpression of CDX2 in gastric cancer cells promotes the development of multidrug resistance

    Am. J. Cancer Res.

    (2014)
  • J. Ge et al.

    Upregulation of autophagy-related gene-5 (ATG-5) is associated with chemoresistance in human gastric cancer

    PLoS One

    (2014)
  • Cited by (28)

    • Moving toward a new horizon for the aldose reductase inhibitor epalrestat to treat drug-resistant cancer

      2022, European Journal of Pharmacology
      Citation Excerpt :

      For example, it would be interesting to combine a proteasome inhibitor like bortezomib with EPA because bortezomib has been shown to dramatically increases mRNA levels of AKR1B10 (32-fold) and AKR1B1 (5-fold) in SW-480 colon cancer cells (Loeffler-Ragg et al., 2009; Ebert et al., 2011). Combinations of an AKR1B10 inhibitor with different platinum derivatives (cisplatin) or a taxane (docetaxel) could be proposed as well, to reduce drug resistance (Matsunaga et al., 2016; Endo et al., 2017). EPA is a well-known drug, used in human for decades for the treatment of diabetic peripheral neuropathy.

    • Caffeic acid and its derivatives as potential modulators of oncogenic molecular pathways: New hope in the fight against cancer

      2021, Pharmacological Research
      Citation Excerpt :

      Doxorubicin (DOX) is another chemotherapeutic agent that can induce cell cycle arrest in cancer cells via inhibiting topoisomerase activity and preventing DNA and RNA transcription [28]. Increasing evidence have revealed the fact that proteasomal activity could be enhanced in DOX- and CP-resistant cancer cells [191–193]. In stomach and colon cancer cells, 26 S proteasome-based proteolytic activity enhances, especially chymotrypsin-like activity, mediating DOX and CP resistance.

    • Pathophysiological roles of autophagy and aldo-keto reductases in development of doxorubicin resistance in gastrointestinal cancer cells

      2019, Chemico-Biological Interactions
      Citation Excerpt :

      In addition, AKR1B10 and AKR1C3 have been recently proposed as potential targets for development of resistance towards some anticancer drugs such as DOX, mitomycin-C (MMC), cis-diamminedichloroplatinum (CDDP) and oxaliplatin (LOHP), because of aberrant expression of the enzymes in some kinds of anticancer drug-resistant cancer cells [9,14]. Indeed, our previous studies showed that AKR1B10 and/or AKR1C3 are associated with development of gastrointestinal cancer cell resistance to DOZ, MMC and CDDP, and suggested that the two enzymes function as regulators for the development of chemoresistance [13,15–17]. Autophagy is considered to play a wide variety of physiological and pathophysiological roles in development, differentiation, infection, and onset and progression of several diseases including cancer.

    • New insights into molecular mechanisms of rosiglitazone in monotherapy or combination therapy against cancers

      2018, Chemico-Biological Interactions
      Citation Excerpt :

      Cisplatin is one of the most effective chemotherapeutic drugs that are used for treatment of patients with colon cancer, but its continuous administration often evokes the development of chemoresistance. ROSI could increase the sensitivity to cisplatin in cisplatin resistant colon cancer LoVo cells [75]. Moreover, the combination of ROSI and non-traditional chemotherapy drugs could also have a synergistic anticancer activity.

    • The influence of peroxisome proliferator-activated receptor γ (PPARγ) ligands on cancer cell tumorigenicity

      2018, Gene
      Citation Excerpt :

      On the other hand, Cisplatin resistant cancer cells reduce Cisplatin-induced oxidative stress through up-regulating of alde-keto reductase 1B10 (AKR1B10) and down-regulating of PPARγ, with an AKR1B10-dependent mechanism. Hence, it shows that combined treatment with AKR1B10 inhibitor and/or PPARγ agonist can be effective for eliminating Cisplatin-resistant cancer cells (Matsunaga et al., 2016). Anti-tumor effects of 6-iodolactone (6IL), an iodinated arachidonic acid derivative have been reported to mediate by PPARγ.

    View all citing articles on Scopus
    View full text