Elsevier

Experimental Cell Research

Volume 293, Issue 1, 1 February 2004, Pages 129-143
Experimental Cell Research

Low extracellular pH augments TRAIL-induced apoptotic death through the mitochondria-mediated caspase signal transduction pathway

https://doi.org/10.1016/j.yexcr.2003.09.015Get rights and content

Abstract

Tumor necrosis factor-related apoptosis inducing ligand (TRAIL/APO-2L), a member of the tumor necrosis factor (TNF) gene family, is considered as one of the most promising cancer therapeutic agents due to its ability to selectively kill tumor cells. Although microenvironments of solid tumors (hypoxia, nutrient deprivation, and low pH) often affect the effectiveness of chemotherapy, few studies have been reported on the relationship between tumor microenvironments and TRAIL. In this study, we investigated whether low extracellular pH affects TRAIL-induced apoptotic death. When human prostate carcinoma DU145 cells were treated with 200 ng/ml His-tagged TRAIL for 4 h, the survival was approximately 10% at pH 6.3–6.6 and 61.3% at pH 7.4. Similar results were observed in human colorectal carcinoma CX-1 cell line. The TRAIL-mediated activation of caspase, cytochrome c release, and poly (ADP-ribose) polymerase (PARP) cleavage was promoted at low extracellular pH. Immunoprecipitation followed by western blot analysis shows that low extracellular pH enhances the association of truncated Bid with Bax during treatment with TRAIL. Western blot analysis also shows that the low extracellular pH-enhanced TRAIL cytotoxicity does not involve modulation of the levels of TRAIL receptors (DR4, DR5, and DcR2), FLIP, inhibitor of apoptosis (IAP), and Bcl-2. Overexpression of Bcl-2 effectively prevented low extracellular pH-augmented TRAIL cytotoxicity. Taken together, we propose that TRAIL-mediated cytotoxicity is greatly enhanced in low pH environments by promoting caspase activation.

Introduction

It is well known that severe architectural and functional abnormalities are commonly observed in the capillary network that develops during tumor growth [1]. These abnormalities cause insufficient blood supply and development of a pathophysiological tumor microenvironment. Previous studies with the micropore chamber sampling procedure [2] and tumor-isolated preparations [3] reveal differences in the constituents of serum (vascular compartment) compared to interstitial fluid (interstitial compartment). Vascular and interstitial compartments are two major compartments of the extracellular space of solid tumors. The tumor interstitial compartment is characterized by low oxygen tensions (hypoxia) [4], low glucose concentrations [5], high lactate concentrations [6], [7], and low extracellular pH [8]. These characteristic features, which occur transiently or chronically, can markedly affect the therapeutic response [9], [10]. Recently, we demonstrated that low glucose augments the effect of tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), a potent anticancer agent, which induces apoptosis [11]. This low glucose-induced augmentation is mediated through the ceramide–Akt–FLIP pathway [11]. In this study, we investigated whether low extracellular pH alters TRAIL-induced cytotoxicity. We believe that understanding the role of low extracellular pH in TRAIL-mediated cytotoxicity will be important for predicting the effectiveness of this anticancer agent and developing new drug targets.

TRAIL/APO-2L is a type II integral membrane protein belonging to the tumor necrosis factor (TNF) family. TRAIL is a 281-amino acid protein, related most closely to Fas–APO-1 ligand. Like Fas ligand (FasL) and TNF, the C-terminal extracellular region of TRAIL (amino acids 114–281) exhibits a homotrimeric subunit structure [12]. However, unlike FasL and TNF, it induces apoptosis in a variety of tumor cell lines more efficiently than normal cells [13]. This differential cytotoxicity suggests that TRAIL is a good candidate for use as an effective anticancer agent. However, recent studies reveal that a polyhistidine-tagged TRAIL induces apoptosis in normal human hepatocytes in culture [14]. This is probably due to an aberrant conformation and subunit structure of TRAIL in the presence of low zinc concentrations [15]. In contrast, native-sequence, non-tagged recombinant TRAIL, when produced under optimized zinc concentrations, is markedly more active against tumor cells than the polyhistidine-tagged ligand, but has minimal toxicity toward human hepatocytes in vitro [15]. Moreover, preclinical studies in mice and primates have shown that administration of TRAIL can induce apoptosis in human tumors, but no cytotoxicity to normal organs or tissue [16]. In addition, unlike TNF and FasL, TRAIL mRNA is expressed constitutively in many tissues [12], [17]. Recent studies also reveal that TRAIL, which is constitutively expressed on murine natural killer cells in the liver, plays an important role in surveillance of tumor metastasis [18]. Although TRAIL is an apoptosis-inducing member of the TNF gene family on the basis of amino acid homology to the TNF and FasL [12], [17], unlike TNF and FasL, TRAIL infusion does not cause a lethal inflammatory response.

The apoptotic signal induced by TRAIL is transduced by its binding to the death receptors TRAIL-R1 (DR4) and TRAIL-R2 (DR5), which are members of the TNF receptor superfamily. Both DR4 and DR5 contain a cytoplasmic death domain that is required for TRAIL receptor-induced apoptosis. TRAIL also binds to TRAIL-R3 (DcR1) and TRAIL-R4 (DcR2), which act as decoy receptors by inhibiting TRAIL signaling [19], [20], [21], [22], [23], [24]. Unlike DR4 and DR5, DcR1 does not have a cytoplasmic domain and DcR2 retains a cytoplasmic fragment containing a truncated form of the consensus death domain motif [21]. The relative resistance of normal cells to TRAIL has been explained by the presence of many the decoy receptors on normal cells [25], [26]. Recently, this hypothesis has been challenged based on results showing poor correlations between DR4, DR5, and DcR1 expression and sensitivity to TRAIL-induced apoptosis in normal and cancerous breast cell lines [27] and melanoma cell lines [28]. This discrepancy indicates that other factors such as death inhibitors including FLICE-inhibitory protein (FLIP) [28], Fas-associated protein (FAP-1) [29], Bcl-2 [30], Bcl-XL[30], Bruton's tyrosine kinase (BTK) [31], silencer of death domain (SODD) [32], toso [33], inhibitor of apoptosis (IAP) [34], X-linked inhibitor of apoptosis (XIAP) [35], and survivin [36] are also involved in differential sensitivity to TRAIL. Previous studies show that chemotherapeutic agents [27], [28], [37] and ionizing radiation [38] can sensitize TRAIL-induced cytotoxicity by decreasing intracellular levels of FLIP [28] or increasing DR5 gene expression in response to genotoxic stress [37], [38], [39]. In this study, we observed that low extracellular pH augments TRAIL-induced apoptotic death by promoting the mitochondria-mediated caspase signal transduction pathway via facilitating the interaction between truncated Bid and Bax rather than changes in the levels of TRAIL receptors, FLIP, IAP, and Bcl-2.

Section snippets

Cell culture and survival assay

Human prostate adenocarcinoma DU-145 cells or human colorectal carcinoma CX-1 cells were cultured in DMEM medium (Gibco BRL, Gaithersburg, MD, USA) or RPMI medium (Gibco BRL), respectively, with 10% fetal bovine serum (HyClone, Logan, UT, USA) and 26 mM sodium bicarbonate for monolayer cell culture. The dishes containing cells were kept in a 37°C humidified incubator with a mixture of 95% air and 5% CO2. Two days before the experiment, cells were plated into 60-mm dishes. For trypan blue

TRAIL-induced cytotoxicity is enhanced at low extracellular pH

To investigate the effect of low extracellular pH on TRAIL-induced cytotoxicity, human prostatic adenocarcinoma DU-145 cells were treated with His-tagged TRAIL at various extracellular pH (6.3–7.4). Figs. 1A and 1B show that no cytotoxicity was observed at low extracellular pH alone. In contrast, TRAIL-induced cytotoxicity was promoted at low extracellular pH. Cells undergoing apoptosis during treatment with TRAIL showed cell surface blebbing and formation of apoptotic bodies (Fig. 1A). During

Discussions

Microelectrode measurements in human and rodent solid tumors reveal that the extracellular pH (pHe) of tumor has a broader distribution and is on average 0.5 pH units more acidic than normal tissue pHe[8], [48]; an average extracellular pHe is 7.4 in normal tissue and 6.8–7.2 in tumor tissue [8], [49], [50], [51], [52]. This is because tumor cells which are farther than 100–200 μm from the nearest functional blood vessel experience low influx of metabolites and a low efflux of potentially toxic

Acknowledgements

This work was supported by NIH–NCI (CA-48000, CA-95191, CA-96989, CA-64139), DOD-PC001283, DOD-PC010270, Elsa U. Pardee Foundation, Pittsburgh Foundation, and Competitive Medical Research Funds of The UPMC Health System.

References (76)

  • A. Saleh et al.

    Cytochrome c and dATP-mediated oligomerization of Apaf-1 is a prerequisite for procaspase-9 activation

    J. Biol. Chem.

    (1999)
  • H. Zou et al.

    An APAF-1 cytochrome c multimeric complex is a functional apoptosome that activates procaspase-9

    J. Biol. Chem.

    (1999)
  • X. Luo et al.

    Bid, a Bcl2 interacting protein, mediates cytochrome c release from mitochondria in response to activation of cell surface death receptors

    Cell

    (1998)
  • L.E. Gerweck

    Tumor pH: implications for treatment and novel drug design

    Semin. Radiat. Oncol.

    (1998)
  • A. Thistlethwaite et al.

    pH distribution in human tumours

    Int. J. Radiat. Oncol., Biol., Phys.

    (1985)
  • J.G. Emery et al.

    Osteoprotegerin is a receptor for the cytotoxic ligand TRAIL

    J. Biol. Chem.

    (1998)
  • Y. Gazitt et al.

    Apoptosis-induced by TRAIL AND TNF-alpha in human multiple myeloma cells is not blocked by BCL-2

    Cytokine

    (1999)
  • F.C. Kischkel et al.

    Apo2L/TRAIL-dependent recruitment of endogenous FADD and caspase-8 to death receptors 4 and 5

    Immunity

    (2000)
  • A.A. Kuang et al.

    FADD is required for DR4- and DR5-mediated apoptosis: lack of trail-induced apoptosis in FADD-deficient mouse embryonic fibroblasts

    J. Biol. Chem.

    (2000)
  • M.R. Sprick et al.

    FADD/MORT1 and caspase-8 are recruited to TRAIL receptors 1 and 2 and are essential for apoptosis mediated by TRAIL receptor 2

    Immunity

    (2000)
  • H. Li et al.

    Cleavage of BID by caspase 8 mediates the mitochondrial damage in the Fas pathway of apoptosis

    Cell

    (1998)
  • S.L. Schendel et al.

    Ion channel activity of the BH3 only Bcl-2 family member, BID

    J. Biol. Chem.

    (1999)
  • P. Li et al.

    Cytochrome c and dATP-dependent formation of Apaf-1/caspase-9 complex initiates an apoptotic protease cascade

    Cell

    (1997)
  • T.H. Kim et al.

    Bid-induced cytochrome c release is mediated by a pathway independent of mitochondrial permeability transition pore and Bax

    J. Biol. Chem.

    (2000)
  • P. Vaupel et al.

    Blood flow, oxygen and nutrient supply, and metabolic microenvironment of human tumors: a review

    Cancer Res.

    (1989)
  • P.M. Gullino et al.

    The interstitial fluid of solid tumors

    Cancer Res.

    (1964)
  • P.M. Gullino et al.

    Studies on the exchange of fluids between host and tumor: I. A method for growing “tissue-isolated” tumors in laboratory animals

    J. Natl. Cancer Inst.

    (1961)
  • P. Vaupel et al.

    Oxygenation of human tumors: evaluation of tissue oxygen distribution in breast cancers by computerized O2 tension measurements

    Cancer Res.

    (1991)
  • P.M. Gullino

    Extracellular compartments of solid tumors

  • G. Schwickert et al.

    Correlation of high lactate levels in human cervical cancer with incidence of metastasis

    Cancer Res.

    (1995)
  • S. Walenta et al.

    Correlation of high lactate levels in head and neck tumors with incidence of metastasis

    Am. J. Pathol.

    (1997)
  • W.F. Mueller-Klieser et al.

    Tumor physiology and cellular microenvironments

  • A.C. Sartorelli

    Therapeutic attack of hypoxic cells of solid tumors: presidential address

    Cancer Res.

    (1988)
  • S.Y. Nam et al.

    Low glucose-enhanced TRAIL cytotoxicity is mediated through the ceramide–Akt–FLIP pathway

    Oncogene

    (2002)
  • A. Ashkenazi et al.

    Death receptors: signaling and modulation

    Science

    (1998)
  • M. Jo et al.

    Apoptosis induced in normal human hepatocytes by tumor necrosis factor-related apoptosis-inducing ligand

    Nat. Med.

    (2000)
  • D. Lawrence et al.

    Differential hepatocyte toxicity of recombinant Apo2L/TRAIL versions

    Nat. Med.

    (2001)
  • H. Walczak et al.

    Tumoricidal activity of tumor necrosis factor-related apoptosis-inducing ligand in vivo

    Nat. Med.

    (1999)
  • Cited by (0)

    View full text