Research Article
Decreased lactate dehydrogenase B expression enhances claudin 1-mediated hepatoma cell invasiveness via mitochondrial defects

https://doi.org/10.1016/j.yexcr.2011.02.011Get rights and content

Abstract

Aerobic lactate production of which the final step is executed by lactate dehydrogenase (LDH) is one of the typical phenotypes in invasive tumor development. However, detailed mechanism of how LDH links to cancer cell invasiveness remains unclear. This study shows that suppressed LDHB expression plays a critical role in hepatoma cell invasiveness by inducing claudin-1 (Cln-1), a tight junction protein, via mitochondrial respiratory defects. First, we found that all the SNU human hepatoma cells with increased glycolytic lactate production have the defective mitochondrial respiratory activity and the Cln-1-mediated high invasive activity. Similar results were also obtained with human hepatocellular carcinoma tissues. Unexpectedly, the increased lactate production was due to LDH isozyme shifts to LDH5 by LDHB down-expression rather than LDHA induction, implying the importance of LDHB modulation. Second, LDHB knockdown did not only trigger Cln-1 induction at the transcriptional level, but also induced respiratory impairment. Interestingly, most respiratory inhibitors except KCN induced Cln-1 expression although complex I inhibition by rotenone was most effective on Cln-1 induction. Respiratory defect-mediated Cln-1 induction was further confirmed by knockdown of NDUFA9, one of complex I subunits. Finally, ectopic expression of LDHB attenuated the invasiveness of both SNU 354 and 449 cells whereas LDHB knockdown significantly augmented the invasiveness of Chang cells with Cln-1induction. The increased invasive activity by LDHB modulation was clearly reversed by knocking-down Cln-1. Taken together, our results suggest that LDHB suppression plays an important role in triggering or maintaining the mitochondrial defects and then contributes to cancer cell invasiveness by inducing Cln-1 protein.

Introduction

Increased aerobic glycolysis, continuous conversion of glucose to lactic acid in the presence of oxygen (i.e. the Warburg effect), is a distinctive hallmark of solid tumors [1], [2]. Because it is closely associated with increased metastasis in some cancers [3], [4], aerobic glycolysis is further considered a metabolic signature for invasive cancer. The glycolytic phenotype may be the result of adaptation to environmental constraints such as intermittent hypoxia in premalignant lesions. This hypothesis well corresponds to the concept termed ‘Pasteur effect,’ which glycolysis is inhibited by the presence of oxygen [5]. However, enhanced glycolysis persists in metastatic malignant cancers, even after increased angiogenesis restores a normoxic environment. This suggests that increased glycolysis is not just a passive response, but an active cellular strategy to confer a selective advantage for malignant progression by affecting acid-mediated matrix degradation, immune protection, and the dominant growth of cancer cells in an altered cellular environment [2], [6], [7], [8]. In addition to the metastatic ability, cell growth of malignant tumor persistently depends on glycolysis in normoxic culture conditions [9]. This suggests that augmented glycolysis may positively suppress or impair aerobic mitochondrial ATP production machinery, resulting in cellular dependence on glycolytic ATP. Evidence supporting this active strategy is that many glycolytic enzymes are upregulated by oncogenes such as Ras, Src, and Her2/Neu [10], [11]. Therefore, glycolytic activation and consequent aerobic glycolysis may also play a critical role in tumor progression associated with genetic alterations [12]. However, the underlying mechanisms are poorly understood.

Cultured cells derived from tumors maintain this altered metabolism when cultivated under normoxic conditions [2], [3], indicating that aerobic glycolysis is not just a transient result due to in vivo tumor microenvironment, but may be constitutively upregulated through stable genetic or epigenetic changes. Regulation of lactate dehydrogenase (LDH) plays a key role in this, since LDH is the alternative supplier of NAD+ in the absence of mitochondrial oxidation. Moreover, LDH produces lactate: lactate causes chronic acidification of the intratumoral microenvironment, which in turn helps drive cancer cell metastasis [13], [14].

Enzymatically functional LDH consists of four subunits, and there are two types of subunits designated M (muscle-type; LDHA gene product) and H (heart-type; LDHB gene product). Normal cells can contain five different LDH isozymes with different substrate reactivities as a result of the five different combinations of the two different subunits: LDH1 (H4); LDH2 (MH3); LDH3 (M2H2); LDH4 (M3H); LDH5 (M4) [15]. The expression levels of LDHA and LDHB expression determine the cell's isozyme pattern. LDH5 effectively catalyzes the conversion of pyruvate to lactate [16], and an isozyme shift to LDH5 has been linked with metastatic cancer [17], [18]. Furthermore, the importance of LDHA modulation in cancer progression is highlighted by reports that oncogenes can upregulate LDHA in tumor cells. For example, LDHA transcription is directly activated by cMyc and is indirectly induced by other oncogenes via hypoxia-inducing factor α (HIF-1α) stabilization [19], [20], [21]. However, it is not clear whether LDHA upregulation fully explains the stable and persistent isozyme shift toward LDH5 in cancer cells.

Hepatocellular carcinoma (HCC) is an increasingly common malignant tumor, with worldwide incidence of 5.5–14.9 per 100,000 inhabitants [22], [23]. Currently, liver transplantation and tumor resection are the most effective standard therapies. Although these procedures provide 5-year survival rates of 70%, this rate is only for patients within the Milan criteria (single tumor  5 cm in size or up to three tumors  3 cm in size) [24]. More than 60% of patients do not fall into these criteria because HCC is often diagnosed only after the size of tumor becomes larger, thereby resulting in survival time raging from 3 to 16 months [23]. Moreover, these therapeutic procedures are not complete cure, as half of the treated patients experience tumor recurrence within 3 years. Thus, alternative drug-based therapies for HCC are currently being tested and applied despite their low survival rates. High lethality with drug-based therapies is mainly due to its resistance to existing anticancer agents [25]. Glycolysis inhibition has recently been proposed to overcome drug resistance [9], but it is unknown whether this strategy is reasonable and would also be effective in treating HCC. Present study demonstrated the link of LDH isozyme shift via LDHB suppression to mitochondrial respiratory defects and claudin-1 induction, and their contribution to hepatoma cell invasiveness. Our results provide a new insight into the metabolic and molecular backgrounds of hepatocellular carcinoma for developing a novel drug based on glycolysis inhibition.

Section snippets

Cell cultures and cell growth rates, and tumor samples

Human hepatoma cells (SNU-354, SNU-387, SNU-423, and SNU-449) were purchased from Korean Cell Line Bank (Seoul, Korea) and were propagated in GIBCO® RPMI1640 medium (Invitrogen, Carlsbad, CA, USA) supplemented with 10% GIBCO® fetal bovine serum (FBS) (Invitrogen) and antibiotics at 37 °C in a humidified incubator with 5% CO2. Chang cell, an immortalized human hepatocyte, was obtained from the American Tissue Culture Collections (ATCC, Rockville, MD, USA), and cultured in GIBCO® Dulbecco's

The metabolic transition to glycolytic lactate production is mainly associated with suppressed LDHB expression in HCC cells and tissues

To investigate the relationship between the metabolic shift toward aerobic glycolysis and liver cancer cell invasiveness, we first characterized the metabolic phenotype and underlying molecular backgrounds of four SNU HCC cell lines (SNU-354, SNU-387, SNU-423, and SNU-449) derived from human HCC [30], [31] and compared to those of a Chang cell clone which shows higher mitochondrial respiratory activity and certain liver-characteristics. When cultured under normoxic conditions, all HCC cell

Discussion

Aerobic glycolysis in cancer cells has long been regarded as a phenotype acquired in response to environmental constraints such as intermittent hypoxia in premalignant lesions [2], [12]. However, current thought is that this metabolic change is not simply an adaptation to a hypoxic environment but is an active cellular strategy that confers a significant advantage for proliferation and malignancy. In support of the latter view, there is an evidence that increased lactate release leads to

Acknowledgments

This work was supported by the Korean Science and Engineering Foundation (KOSEF), by a grant from the Korean government (MEST) (R13-2003-019-01007-0), by Mid-carrer Research Program through NRF grant funded by the MEST (No. 2009–0079076), and by a grant from the National R&D Program for Cancer Control, Ministry of Health & Welfare, Republic of Korea (0720400).

References (41)

  • S. Walenta et al.

    High lactate levels predict likehood of metastases, tumor recurrence, and restricted patient survival in human cervical cancers

    Cancer Res.

    (2000)
  • E. Racker

    History of the Pasteur effect and its pathobiology

    Mol. Cell. Biochem.

    (1974)
  • R.A. Gatenby et al.

    Acid-mediated tumor invasion: a multidisciplinary study

    Cancer Res.

    (2006)
  • R.H. Xu et al.

    Inhibition of glycolysis in cancer cells: a novel strategy to overcome drug resistance associated with mitochondrial respiratory defect and hypoxia

    Cancer Res.

    (2005)
  • E. Laughner et al.

    Her2(neu) signaling increases the rate of hypoxia-inducible factor 1alpha (HIF-1alpha)synthesis: novel mechanism for HIF-1-mediated vascular endothelial growth factor expression

    Mol. Cell. Biol.

    (2001)
  • R.J. Gillies et al.

    Causes and consequences of increased glucose metabolism of cancers

    J. Nucl. Med.

    (2008)
  • R. Martínez-Zaguilán et al.

    Acidic pH enhances the invasive behavior of human melanoma cells

    Clin. Exp. Metastasis

    (1996)
  • O.K. Schlappack et al.

    Glucose starvation and acidosis: effect on experimental metastatic potential, DNA content and MTX resistance of murine tumour cells

    Br. J. Cancer

    (1991)
  • M. Drent et al.

    Usefulness of lactate dehydrogenase and its isoenzyme as indicators of lung damage or inflammation

    Eur. Respir. J.

    (1996)
  • M.I. Koukourakis et al.

    Lactate dehydrogenase 5 (LDH5) relates to up-regulated hypoxia inducible factor pathway and metastasis in colorectal cancer

    Clin. Exp. Metastasis

    (2005)
  • Cited by (46)

    • Claudins and hepatocellular carcinoma

      2024, Biomedicine and Pharmacotherapy
    • Physical Exercise and Tumor Energy Metabolism

      2022, Cancer Treatment and Research Communications
      Citation Excerpt :

      The changes are important because the hypoxic microenvironment of tumor tissue leads to increased LDH-A expression, whereas LDH-A silencing suppresses tumorigenicity of breast cancer [58]. Furthermore, the increase in lactate production is related to the down-regulation of LDH-B [59]. In a study with an animal model and chemical induction of breast cancer, aerobic and voluntary physical exercise reduced the incidence of breast cancer and induced changes in hormones and growth factors, reducing insulin concentration, IGF-I, corticosterone, and leptin [60].

    • Lactic acidosis caused by repressed lactate dehydrogenase subunit B expression down-regulates mitochondrial oxidative phosphorylation via the pyruvate dehydrogenase (PDH)-PDH kinase axis

      2019, Journal of Biological Chemistry
      Citation Excerpt :

      Some studies show that LDH5 activity is mainly enhanced because of increased LDHA expression via the interplay of HIF-1α and cMyc (15, 29). However, we recently reported an LDH isozyme shift toward LDH5 in hepatoma cells, which was unexpectedly achieved by LDHB down-expression rather than LDHA up-expression (30). Moreover, this LDHB suppression restrained mitochondrial respiration, providing a model to elucidate the mechanism by which enhanced glycolysis regulates mitochondrial respiratory activity.

    • Lactate-mediated mitoribosomal defects impair mitochondrial oxidative phosphorylation and promote hepatoma cell invasiveness

      2017, Journal of Biological Chemistry
      Citation Excerpt :

      The MRPL13 knockdown also significantly decreased OCR and enhanced cell invasion activity (Fig. 3, A–C). We previously demonstrated that CLN1 is a key regulator of hepatoma cell invasion activity in response to OXPHOS defect (23). Similarly, mitoribosomal defects, caused by siRNA-mediated MRPL13 knockdown or DOX treatment, clearly induced CLN1 expression (Fig. 3, D and E).

    View all citing articles on Scopus
    1

    These two authors have contributed equally.

    View full text