Abstract

Transient receptor potential (TRP) ion channels are essential for normal functions and health by acting as molecular sensors and transducing various stimuli into cellular and physiological responses. Growing evidence has revealed that TRP ion channels play important roles in a wide range of human diseases, including malignancies. In light of recent discoveries, it has been found that TRP melastatin-subfamily members, TRPM7 and TRPM8, are required for normal and cancerous development of exocrine pancreas. We are currently investigating the mechanisms which mediate the functional roles of TRPM7 and TRPM8 and attempting to develop these ion channels as clinical biomarkers and therapeutic targets for achieving the goal of personalized therapy in pancreatic cancer.

1. Introduction

The purpose of this paper is to examine the newfound roles of the transient receptor potential (TRP) ion channels, in particular the melastatin subfamily members TRPM7 and TRPM8, in pancreatic cancer and discuss their potential as clinical biomarkers and therapeutic targets. Pancreatic adenocarcinoma is a highly lethal disease; its incidence has been rising and the prognosis remains dismal [1]. In order to develop effective interventions for prevention, early detection, and treatment of this malignant disease, it is critical to identify the etiologic risk factors and understand the mechanism underlying formation and progression of pancreatic neoplasia [2]. Steady progress has been made in advancing knowledge in molecular biology and genetics of pancreatic development under normal and malignant conditions [2, 3]. Accumulating evidence has implicated that ion channels are important modulators of the pathogenic mechanisms underlying many human diseases such as cancer [4]. There is a paucity of knowledge about the role of ion channels in normal pancreatic development and during malignant transformation. By using the zebrafish model, we have discovered the developmental role of Trpm7 in exocrine pancreas [5]. Moreover, we have demonstrated the novel roles of the human orthologue TRPM7 and its sub-family member TRPM8 in pancreatic adenocarcinoma [58].

From embryogenesis to adult life, ion channels have been shown to play crucial roles in regulating the diverse cellular processes and physiological functions. Aberrant expression and/or activities of ion channels have been implicated in various disease states, including cancer. Ion channels, particularly the TRP family members, detect a wide range of stimuli in the cellular microenvironment [9]. These include light, changes in temperature and osmolarity, pain, mechanical stress, touch, acidic and alkaline pH, and taste. Functional TRP channels are assembled as tetramers, which consist of four identical TRP subunits (homotetramers) or similar TRP subunits (heterotetramers). Each TRP subunit contains six transmembrane segments S1 to S6, with the amino- and carboxyl-terminals facing the intracellular side of the membrane. Voltage sensing has been shown to occur in the region between segments S1 and S4, and the hydrophobic central pore through which ions flow is located between the 5th and 6th segments [10]. The TRP channels exhibit relative ionic selectivity, and they exert regulatory effects on cellular proliferation, survival, differentiation, and migration by maintaining cationic homeostasis and modulating signaling pathways [11]. There is a growing body of evidence indicating that aberrant expression of some of the TRP channels contributes to the uncontrolled proliferation and growth of malignant tumors [4].

In the TRP family of ion channels, there are seven TRP subfamilies including TRPC (classical or canonical), TRPM (melastatin), TRPA (ankyrin), TRPV (vanilloid), TRPP (polycystin), TRPN (NOMP-C homologues), and TRPML (mucolipin). These TRP subfamilies of channels share common gross architecture, and each subfamily is characterized by unique structural features [10]. TRPM represents one of the largest and most diverse TRP subfamily, and there are eight members in the TRPM subfamily. The TRPM channels possess the TRP domain (also conserved in the TRPC and TRPN subfamilies), which is a conserved 25-amino acid sequence distal to the S6 region and proximal to the carboxyl-terminus. It has been proposed that the TRP domain contributes to tetramerization of the channel subunits and also gating of the channel. The TRPM channels are nonselectively permeable for cations, and they play diverse physiological roles by sensing physical and chemical stimuli in the microenvironment and responding through modulation of the intracellular ionic levels and signaling pathways. Accumulating evidence from morphological, biochemical, and genetic studies has begun to elucidate the functional roles of the TRPM channels in various aspects of human malignancies. Until recently, it was unknown if the TRPM channels had any influential role in pancreatic cancer. Using a combination by positional cloning and candidate testing, we discovered that the zebrafish sweetbread (swd) mutations affect Trpm7, and we identified a novel role of Trpm7 in controlling the size of exocrine pancreas through regulation of cell cycle progression and growth in pancreatic epithelia [5]. In attempt to test the hypothesis that deregulated expression and/or activities of developmental regulators contributes to carcinogenesis, we have discovered that TRPM7 and TRPM8 are aberrantly overexpressed in pancreatic adenocarcinoma and required for proliferation of the cancer cells [58]. These studies provide a novel link between cellular sensation and signal modulation in pancreatic cancer. With the goal of identifying and developing clinical biomarkers and therapeutic targets in pancreatic cancer, we are actively investigating the mechanistic roles of TRPM7 and TRPM8 ion channels in the initiation, growth, and invasion of pancreatic neoplasia.

2. TRPM7 Channel Kinase: Physiological Functions and Roles in Development and Cancer

TRPM7 is a divalent cation-permeable channel with an intrinsic kinase, and it carries out its physiological functions by acting as a cellular sensor and signal transducer through its ion channel and kinase activity [1215]. Among the TRP family of ion channels, only the TRPM7 channel and its closest homologue TRPM6 possess such protein kinase domain. The function of the TRPM7 channel requires the activity of its endogenous kinase, which is capable of phosphorylating the serine and threonine residues of protein [13, 14]. While interaction of the TRP domain of TRPM7 with phosphatidylinositol-4,5-bisphosphate (PIP2) leads to activation of the TRPM7 channel, stimulation of phospholipase C (PLC) results in hydrolysis of PIP2 and thus inactivation of the TRPM7 channel [15]. Moreover, interaction between the kinase domain and PLC regulates the channel activity of TRPM7 in a manner that is dependent on cAMP and protein kinase A [16]. TRPM7 kinase also has autophosphorylating ability, which is regulated by intracellular level of Mg2+-ATP [1619]. TRPM7 is ubiquitously expressed, and it controls cellular homeostasis of ions, particularly Mg2+ and Ca2+, and modulates the signaling pathways involved in cell cycle progression, cytodifferentiation, survival, and migration.

TRPM7 plays a regulatory role in a variety of cellular effects, which are dependent on the cell types. These include cell survival in lymphocytes, neurons, and mast cells [12, 20, 21], proliferation of osteoblasts and pancreatic epithelia and cancer cells [58, 22, 23]; cytodifferentiation in T-lymphocytes [24]. Moreover, TRPM7 has been shown to regulate cell adhesion in neuroblastoma [25], cell volume in epithelia of kidney and cervical uterus [26], and cell migration in osteoblasts and fibroblasts [23, 27]. Electrophysiological data indicate that cellular influx of Mg2+ through the TRPM7 channel results in changes of intracellular concentrations of Mg2+ or Ca2+. Hypothetically, the resulting perturbed ionic homeostasis leads to modulation of the signaling complex and produces various cellular responses of TRPM7 [14, 15, 20, 27].

In vertebrates, the functional roles of TRPM7 are organ-specific, as revealed by genetic studies in model organisms. This is illustrated by the requirement of Trpm7 in skeletal formation and skin pigmentation in zebrafish. Moreover, it has been shown that Trpm7 is required for proliferation and survival of melanoblasts and osteoblasts [5, 2830]. In mice, genetic deletion of Trpm7 produces lethality in the embryos, and disruption of Trpm7 in lymphoid tissues impairs lymphocytic development [24]. Recently, we have reported that the zebrafish sweetbread (swd) mutations affect Trpm7, the zebrafish orthologue of mammalian TRPM7 [5]. From a genome-wide ethylnitrosourea-induced mutagenesis screen for mutations affecting the exocrine pancreas, we previously identified and recovered the zebrafish swd alleles, , and [2, 30]. Besides, genetic deletions in Trpm7 were identified in the zebrafish touchtone (tct) mutants, which have reduced skin pigmentation and abnormally developed skeletons [28]. Examination of the and mutant larvae revealed that their exocrine pancreata are relatively small [2, 5, 30]. Consistent with this finding, the pancreatic acini and ducts in the mutants are hypomorphic; however, specification of cell fate and cytodifferentiation in exocrine pancreatic epithelia appear normal [2, 5, 30].

By impairing cell cycle progression and cell growth, the and mutations cause diminished proliferation of exocrine pancreatic epithelia [5]. Consistent with the role of TRPM7 as a transporter of Mg2+, it was demonstrated that the impaired growth of exocrine pancreas in the and mutants can be partially improved by adding extra Mg2+ to the embryo medium. The supplementary Mg2+-induced partial rescue of the exocrine pancreatic phenotype in the trpm7 mutants is associated with repression of the mRNA levels of and cyclin G1 [5]. Furthermore, the and mutant larvae express elevated levels of suppressor of cytokine signaling 3a (socs3a), a negative regulator of epidermal growth factor (EGF) receptor-mediated signaling [5]. Exocrine pancreatic growth in the trpm7 mutants can be rescued by repression of socs3a by either supplementary Mg2+ or antisense oligos directed against socs3a mRNA. These effects are associated with improved cell cycle progression and cell growth in the exocrine pancreatic epithelia [5]. These studies support a novel role of Trpm7 in exocrine pancreas during early development, and the proliferative role of Trpm7 in exocrine pancreatic epithelia is mediated by Mg2+-sensitive mechanisms that involve Socs3a.

By translating the developmental studies of Trpm7 in zebrafish to humans, we have found the previously unidentified role of TRPM7 in pancreatic adenocarcinoma. In human pancreatic adenocarcinoma tissues, TRPM7 protein is aberrantly overexpressed [5] (Figure 1). Similarly, the mRNA levels of TRPM7 are elevated in the majority of human pancreatic adenocarcinoma cell lines being examined [5]. By using small interfering RNA (siRNA) to inhibit translation of TRPM7 mRNA in the pancreatic cancer cells, we provide evidence that TRPM7 is required for cellular proliferation by preventing cell cycle arrest in the G0/G1 phases [5]. In the TRPM7-deficient cells, the reduced proportions of cells in the S and G2/M phases correlate with upregulated expression of the cyclin-dependent kinase inhibitor and repression of cyclins G1 and B1 [5]. Consistent with the findings in zebrafish TRPM7 mutants and in agreement with the role of TRPM7 as a transporter or divalent cations, addition of extra Mg2+ in the culture medium rescues the proliferative defect of TRPM7-deficient pancreatic cancer cells [5].

In a recent report, we have demonstrated a novel role of TRPM7 in preventing replicative senescence. RNA interference-mediated silencing of TRPM7 did not cause an increase in apoptotic cell death, as analyzed by flow cytometry [7]. The pancreatic cancer cells with silenced expression of TRPM7 exhibit morphological features including enlarged cell size and multiple nuclei, suggestive of replicative senescence. This was confirmed by assaying for senescence associated -galactosidase (SA -gal) activity in the TRPM7-deficient cells, which were shown to have undergone accelerated cellular senescence [7]. Besides, our current studies using human pancreatic adenocarcinoma cells provide novel evidence that silenced expression of TRPM7 impedes cell migration (Yee NS, unpublished data), suggesting TRPM7 plays an important role in invasion and metastasis of pancreatic cancer.

Taken together, TRPM7 is required for Mg2+-dependent cellular proliferation in the developing exocrine pancreas and in pancreatic adenocarcinoma through modulation of the cell cycle regulators. Our current research focuses on the signaling mechanisms including the Socs3a pathway that mediate the functional roles of TRPM7 in exocrine pancreatic organogenesis as well as in the growth and invasion of pancreatic neoplasia. We hypothesize that aberrantly up-regulated expression of TRPM7 in pancreatic adenocarcinoma contributes to the uncontrolled proliferation and invasion of the cancer cells by facilitating cell cycle progression and cell migration and by preventing replicative senescence. In our working model, activation of TRPM7 by various stimuli modulates intracellular Mg2+ and/or Ca2+ levels and interact with the epidermal growth factors (EGF-) induced signaling pathways, resulting in cell cycle progression, cell survival, and cell migration in pancreatic adenocarcinoma (Figure 2). Besides pancreatic adenocarcinoma, the role of TRPM7 has also been implicated in cancers that arise in other organs, such as retinoblastoma [31], head and neck carcinoma [32], gastric carcinoma [33], breast carcinoma [34], and nasopharyngeal carcinoma [35]. While the mechanism underlying the aberrant overexpression of TRPM7 in malignant neoplasia has yet to be determined, the existing data support the exploration of its clinical significance in pancreatic adenocarcinoma and the other malignancies.

3. TRPM8 Ion Channel: Physiological Functions and Roles in Cancer

With identification of the role of TRPM7 in pancreatic development and cancer, we examined the expression of the other TRPM subfamily of ion channels in a panel of human pancreatic adenocarcinoma cell lines. We discovered that TRPM8, a nonselective, voltage-gated, and Ca2+ permeable ion channel is aberrantly expressed with a functional role in pancreatic cancer [6, 8]. For the TRPM8 channel to be functional, the coiled coil domain and a region between the amino acids 40 and 86 of the amino-terminus are essential [36]. Both the amino- and carboxyl-terminal regions are dispensable for assembly of the channel tetramer and its trafficking to the membrane [36]. Cold temperature (15°C to 25°C) or application of cooling compounds (such as menthol, eucalyptol, and icilin) has been shown to activate the TRPM8 channel, leading to an increase in intracellular concentration of Ca2+ [37, 38]. Indeed, the TRPM8 channel is permeable to both monovalent ions (e.g., Na+, K+) and divalent ions (e.g., Ca2+, Ba2+). Opening of the TRPM8 channel is dependent on voltage, and the probability of channel opening is enhanced with depolarization of the membrane.

TRPM8 is selectively expressed in human adult tissues. A relatively high level of TRPM8 mRNA is present in the prostate gland [39] and a discernible level in the liver, dorsal root ganglion, and trigeminal ganglion neurons [40]. Aberrant expression of TRPM8 has been reported in neoplastic tissues, some of which include prostate carcinoma, breast adenocarcinoma, lung cancer, colorectal cancer, melanoma, urinary bladder cancer, neuroblastoma, neuroendocrine tumor [4147], and in the most recent finding pancreatic adenocarcinoma [6, 8]. In a panel of seven human pancreatic adenocarcinoma cell lines, expression of TRPM8 is consistently up-regulated, as determined by real-time polymerase chain reaction and compared to the immortalized pancreatic ductal epithelia [6]. Analysis of human pancreatic adenocarcinoma tissues by immunohistochemistry using specific anti-TRPM8 antibodies shows that TRPM8 is aberrantly expressed [6, 8] (Figure 3). For comparison, there is no detectable immunoreactivity against TRPM8 in normal or nonmalignant pancreatic ducts [6].

The aberrant expression of TRPM8 in pancreatic adenocarcinoma is associated with a function role. SiRNA-mediated silencing of TRPM8 in pancreatic cancer cells reduced their ability to proliferate and progress through the cell cycle [6]. Such proliferative defect is associated with elevated mRNA levels of the cyclin-dependent kinase inhibitors and [6]. Moreover, phase contrast microscopy and DAPI staining revealed that TRPM8-deficient cells are enlarged and flat, containing multiple nuclei and cytoplasmic vacuoles [6]. These morphological features suggest that the TRPM8-deficient cells underwent nonapoptotic cell death via replicative senescence. This hypothesis was tested by repressing TRPM8 with siRNA and then assaying for activity of SA -gal as a marker of cellular senescence. Result of this study indicates that SA -gal is activated in TRPM8-deficient cells but not in the cells treated with nontargeting control siRNA [8].

Taken together, the TRPM8 channel is required for cellular proliferation in pancreatic adenocarcinoma by preventing cell cycle arrest and replicative senescence. Although it is unclear how TRPM8 becomes aberrantly expressed in pancreatic adenocarcinoma, current data suggest that TRPM8 plays a functional role in pancreatic tumor by contributing to its uncontrolled growth and progression. We propose a working model that will form the basis for testing our hypotheses regarding the signaling mechanisms that mediate the proliferative and prosurvival effects of TRPM8 in pancreatic adenocarcinoma by using cultured cells and animal models (Figure 4).

4. Potential Roles of TRPM7 and TRPM8 as Biomarkersand Targets for Personalized Therapies in Pancreatic Cancer

The aberrant over-expression and proliferative roles of the TRPM7 and TRPM8 ion channels in pancreatic adenocarcinoma suggest a unique opportunity of their development as clinical biomarkers and therapeutic targets (Figure 5). As potential biomarkers of pancreatic cancer, TRPM7 and TRPM8 channels can be exploited to characterize the molecular phenotype of tumors, to facilitate early detection of primary and metastatic tumors, to monitor and predict treatment responses, to determine patient prognosis, and to aid targeted delivery of cancer-specific therapeutics. As a potential therapeutic approach in pancreatic adenocarcinoma, we can target the TRPM7 and TRPM8 ion channels via chemical and/or genetic modulation of their channel activities and their associated signaling pathways. Such approach can also be pursued in combination with cytotoxic drugs [7] or other molecularly targeted agents. Therefore, immense potential exists for TRPM7 and TRPM8 channels to be developed as clinically useful biomarkers and valid therapeutic targets with the hope of accomplishing personalized therapy for patients with pancreatic adenocarcinoma and other malignant diseases [48].

5. Summary and Prospective

The novel findings of Trpm7 ion channel in the growth control of exocrine pancreas in zebrafish prompted discovery of the aberrant expression as well as the proliferative and migratory roles of TRPM7 and TRPM8 channels in human pancreatic adenocarcinoma. Such experimental evidence supports the idea that developmental regulators play important roles in oncogenesis [2, 30, 4954] and provide a novel link of cellular sensors to pancreatic cancer. The focus of our basic research studies is to determine the signaling mechanisms that mediate the proliferative and migratory roles of TRPM7 and TRPM8 in pancreatic epithelia and cancer cells. These results are expected to provide mechanistic insights into pancreatic carcinogenesis and generate new hypotheses regarding the impact of physical and chemical stimuli on the formation and progression of pancreatic neoplasia.

The findings of TRPM ion channels in pancreatic development and adenocarcinoma also provide support for translation of developmental modulators of exocrine pancreas into tumor biomarkers and molecular targets for therapy [2, 51, 55, 56]. The focuses of our translational research studies are (i) to develop TRPM7 and TRPM8 channels as clinical biomarkers, with the goals of improving molecular phenotype of pancreatic tumor and predicting their response to treatment and (ii) to exploit TRPM7 and TRPM8 channels as therapeutic targets and also as ligands for directing tumor-specific delivery of anticancer therapeutics. We hope that the complimentary approaches used in our studies will ultimately lead to developing novel and effective personalized strategies for improving treatment of patients with pancreatic cancer and other malignant tumors.

Acknowledgments

N. S. Yee is supported by the Physician Scientist Stimulus Package from the Pennsylvania State University, College of Medicine. The research work in the laboratory of N. S. Y. has been funded by Penn State Hershey Cancer Institute, University of Iowa Carver College of Medicine, Holden Comprehensive Cancer Center at The University of Iowa, National Institutes of Health (NIDDK and NCI), American Cancer Society, and Fraternal Orders of Eagles.