The pharmacological targeting of ion channels is a promising approach to treating solid tumors. Detailed protocols are provided for characterizing ion channel function in cancer cells and assaying the effects of ion channel modulators on cancer viability.
Ion channels are critical for cell development and maintaining cell homeostasis. The perturbation of ion channel function contributes to the development of a broad range of disorders or channelopathies. Cancer cells utilize ion channels to drive their own development, as well as to improve as a tumor and to assimilate in a microenvironment that includes various non-cancerous cells. Furthermore, increases in levels of growth factors and hormones within the tumor microenvironment can result in enhanced ion channel expression, which contributes to cancer cell proliferation and survival. Thus, the pharmacological targeting of ion channels is potentially a promising approach to treating solid malignancies, including primary and metastatic brain cancers. Herein, protocols to characterize the function of ion channels in cancerous cells and approaches to analyze modulators of ion channels to determine their impact on cancer viability are described. These include staining a cell(s) for an ion channel(s), testing the polarized state of mitochondria, establishing ion channel function using electrophysiology, and performing viability assays to assess drug potency.
Membrane transport proteins are critical for communication between cells, as well as for maintaining cellular homeostasis. Amongst the membrane transport proteins, ion channels serve to drive the growth and development of cells and to maintain the state of cells in challenging and changing environments. Ion channels have also been reported to drive and support the development of solid tumors, both systemically and in the central nervous system (CNS)1,2. For example, KCa3.1 channels are responsible for regulating membrane potential and controlling cell volume, which is important in cell-cycle regulation. Defective KCa3.1 channels have been reported to contribute to the abnormal proliferation of tumor cells3. Further, ion channels may contribute to the metastatic dissemination of cancers. Transient receptor potential (TRP) channels, for example, are involved in Ca2+ and Mg2+ influx; this influx activates several kinases and heat shock proteins that function to regulate the extracellular matrix surrounding a tumor, which is, in turn, important for initiating cancer metastasis4.
Since ion channels can contribute to the development of cancers, they may also be targets for drug-related cancer treatment. For example, resistance to treatment modalities, including chemotherapy and novel immunotherapy, is related to ion channel function dysregulation5,6,7. In addition, ion channels are emerging as important drug targets to impede the growth and development of cancers, with repurposed small molecule (FDA-approved) drugs being examined, as well as biopolymers, including monoclonal antibodies1,2,8,9. While there has been much progress on this front, ion channel cancer drug discovery remains underdeveloped. This is partly due to the unique challenges of studying ion channels in cancer cells. For example, there are technical limitations in setting up electrophysiology assays for slow-acting compounds and temporal differences in channel activation and drug action. Further, the solubility of compounds can also impede progress, as most of the automated electrophysiology systems commonly in use today utilize hydrophobic substrates, which may contribute to artifacts as a result of compound adsorption. In addition, large bioorganic molecular therapeutics such as natural products, peptides, and monoclonal antibodies are technically challenging to screen using conventional electrophysiology assays10. Finally, the bioelectrical properties of cancer cells remain poorly understood11.
Meanwhile, the immunofluorescence staining of ion channels is often challenging. This is due, in part, to the complexity of their structures and their context in the membrane, which impact the ability to both generate and employ antibodies for microscopy studies. It is especially important that the antibodies used to stain ion channels are validated for specificity, affinity, and reproducibility. Commercial antibodies for ion channels should be considered based on their validation strategy and publication record. Experiments should include negative controls to demonstrate the lack of nonspecific binding by either knockdown or knockout of the target protein. Alternatively, cell lines in which the target protein is absent or in low abundance based on mRNA or protein determinations may serve as negative controls. For example, this study shows the localization of the (GABA) receptor subunit Gabra5 in a medulloblastoma cell line (D283). D283 cells with an siRNA knockdown and Daoy cells, another cerebellar medulloblastoma cell line, were stained for Gabra5 and showed no appreciatable staining (data not shown).
Here, methods are presented to analyze and assay ion channel function, as well as the effect of ion channel modulators on cancer cells. Protocols are provided for (1) staining cells for an ion channel, (2) testing the polarized state of mitochondria, (3) establishing ion channel function using electrophysiology, and (4) in vitro drug validation. These protocols emphasize studies of the type A gamma-aminobutyric acid (GABAA) receptor2,12,13,14,15,16, a chloride anion channel and major inhibitory neurotransmitter receptor. However, the methods presented here apply to studying many other cancer cells and ion channels.
1. Immunolabeling ion channels in cultured cells
2. Testing the polarized state of mitochondria
NOTE: This protocol utilizes the TMRE (tetramethylrhodamine, ethyl ester) assay to label the membrane potential in active mitochondria, maintaining a negative charge21,22. TMRE is a cell-permeable, red-orange, positively charged dye accumulating in active mitochondria because of their relative negative charge. Inactive or depolarized mitochondria have reduced membrane potential and fail to proportionally sequester TMRE. FCCP (carbonyl cyanide 4-[trifluoromethoxy] phenylhydrazone), an ionophore uncoupler of oxidative phosphorylation (OXPHOS), depolarizes mitochondrial membranes, thus preventing the accumulation and sequestration of TMRE23. This is illustrated in Figure 2.
3. Establishing ion channel function using electrophysiology
NOTE: The procedure in this section describes the use of an automated electrophysiology assay to screen test compounds in a cancer cell line (Figure 3).
4. In vitro potency
NOTE: This procedure details a MTS assay to determine drug potency. The One Solution Cell Proliferation Assay combines all the required assay reagents into a prepared solution that can be added in one step to cell culture wells to assess the cell viability and proliferation after treatment with experimental compounds. The reagent is reconstituted as per the manufacturer's recommendations (see the Table of Materials), aliquoted, and stored at −20 °C. The section describes the use of the assay to determine the IC50 of test compounds in a particular cell line (Figure 4). This MTS reagent can also be used for the high-throughput screening of large numbers of compounds at known concentrations.
Above are select procedures that can be employed to characterize ion channels in cancerous cells. The first protocol highlights the staining of an ion channel. As detailed, there are many challenges when staining an ion channel or, for that matter, any protein that is present in the extracellular membrane. Shown in Figure 1 is the staining for a subunit of the pentameric GABAA receptor. The second protocol highlights the results of testing the polarized state of mitochondria in cancerous cells. Mitochondria play roles essential for cell viability and proliferation, as well as cell death. In mammalian cells, mitochondria activate apoptosis in response to cellular stress through the release of the Bcl-2 family proteins found in between the mitochondrial inner and outer membranes. In the cytosol, the Bcl-2 family proteins activate caspase proteases, which mediate programmed cell death. Alterations in the plasma membrane ion channel function can result in a disruption of intracellular ion homeostasis, including in terms of the ion levels within the mitochondria, which can lead to a loss of membrane potential, thus triggering apoptosis14. Levels of Ca2+, K+, Na+, and H+ are important determinants in signaling events that can trigger mitochondrial-initiated cell death. Shown in Figure 2 is staining with the cell-permeable, positively charged dye TMRE to label and image the membrane potential in active mitochondria, which maintain a negative charge21,22. TMRE is a red-orange dye that binds with active mitochondria because of their relative negative charge. Depolarized or inactive mitochondria have reduced membrane potential and, thus, fail to sequester TMRE. In this experiment, the ionophore uncoupler FCCP is an important control, as it depolarizes mitochondrial membranes, thereby preventing the accumulation of TMRE23. The third protocol highlights single-cell patch-clamp electrophysiology. Shown in Figure 3 are representative recordings of a trace recorded from the patient-derived medulloblastoma cell line D283. Finally, the fourth protocol highlights an assay to determine the state of proliferation of cancerous cells. Shown in Figure 4 are details on how the MTS assay works and an illustration of the plate and readout when incubated with an agent that impairs the viability of the cancer cells under study (in this case, DAOY).
Figure 1: Staining cells for ion channels. (A) Staining of the protein Gabra5, a subunit of the GABAA receptor, in D283 medulloblastoma cancer cells. (B) Fixed cells treated with the fluorescent stain 4′,6-diamidino-2-phenylindole (DAPI), which binds to DNA. (C) Merge of medulloblastoma cancer cells stained for both Gabra5 and DAPI. Scale bars = 10 µm. The figure is adapted from Kallay et al.14. Please click here to view a larger version of this figure.
Figure 2: Testing the polarized state of mitochondria. (A) Live D283 medulloblastoma cancer cells are treated with increasing concentrations of the drug QH-II-066. The cells are then treated with positively charged, cell-permeable TMRE (tetramethylrhodamine, ethyl ester), which accumulates in active (negatively charged) mitochondria. Depolarized or inactive mitochondria have reduced membrane potential and, therefore, fail to retain the TMRE dye; as a result, they show a low fluorescence signal. Imaged by fluorescence microscopy; FCCP (carbonyl cyanide 4-[trifluoromethoxy] phenylhydrazone). Peak: λex, 549 nm; λem, 575 nm. Scale bars = 10 µm. (B) Quantification of TMRE staining (images shown in panel A) using the software platform. Data are presented as the mean and standard error of the mean. The figure is adapted from Kallay et al.14. Please click here to view a larger version of this figure.
Figure 3: Establishing ion channel function using electrophysiology. (A) Shown is a Port-a-Patch setup or rig, consisting of a Faraday cage (a), recording chamber (b), and suction unit (c, right). (B) Top view of the Port-a-Patch rig highlighting the recording chamber equipped with a perfusion inlet (a), outlet (b), and reference electrode (c). (C) The Port-a-Patch is connected to a rapid solution exchange perfusion system with automated and manual modes of operation and solution reservoirs. (D) Representative current trace from a whole-cell patch-clamp electrophysiology recording using a Port-a-Patch rig (Nanion) and D283 medulloblastoma cancer cells. GABA (10 µM) was applied for 5 s with a holding potential of −80 mV. (E) Representative current trace from a whole-cell patch-clamp electrophysiology recording using a Port-a-Patch rig (Nanion) and D283 medulloblastoma cancer cells with the co-application of GABA (1 µM) and the GABAA receptor agonist (general anesthetic) propofol (50 µM), which potentiates the current induced by GABA alone. Please click here to view a larger version of this figure.
Figure 4: MTS assay to assess drug potency. (A) Chemical reactions underlying the "MTS assay" used to assess the potency of an agent, as reflected by a reduction in cell proliferation. Reduction of MTS tetrazolium by cells that are viable, generating the dye formazan. (B) A 96-well plate showing the colorimetric results of an MTS assay with increasing drug concentrations. In this experiment, the DAOY medulloblastoma cancer cells are treated with increasing concentrations of a pre-clinical drug, KRM-II-08, a positive allosteric modulator of the GABAA receptor. (C) A dose-response curve generated with the MTS assay (from the quantification of 96-well plate). Please click here to view a larger version of this figure.
Manual | Semi/fully automated | |
Through-put | Low | High |
Fast solution exchange | Possible | Yes |
Cost | High | Low |
Operator | Experienced | Beginner/Intermediate |
Cell type | All cells; tissues | Primary single cells; cell lines |
Cell numbers required | Minimum* | High* |
Drug/Solution volume | High | Low |
Resources/Utilities | High | Low |
Maintenance | High | Low |
Experiment control | Very good | Good |
Live cell imaging | Yes | No |
*Port-a-Patch, for example, requires at least a million cells/mL for recordings, while a manual set-up usually needs a few hundred cells on a coverslip. |
Table 1: Comparison of manual versus semi and/or fully automated electrophysiology setups.
Changes in ion channel function alter intracellular signaling cascades, which can impact the overall functioning of a cell. Over the past decade, it has become increasingly clear that ion channels are important to cancer cell growth and metastasis. Importantly, many ion channels are primary targets for approved therapeutics targeting a broad range of disorders24. Investigators have probed whether ion channels could be anti-cancer targets, and the initial results are promising2,16,25. The field is just beginning to investigate the role of ion channels in cancer development and as therapeutic targets, and the future looks bright on both fronts.
In this work, detailed procedures are provided for analyzing ion channels in cancer cells and determining whether a channel is a therapeutic vulnerability. These assays serve as a guide to aid in studying ion channels in cancer cells. Methods have been described that focus on the visualization of ion channels in cancerous cells, the determination of how the modulation of ion channels alters the polarized state of cancerous cells, different approaches to analyze ion channel function using electrophysiology, and the measurement of cancer cell viability.
Immunofluorescence staining can be used to detect the presence and cellular localization of ion channels. The experimental conditions must be carefully optimized to accurately represent where a channel is found within the cell. Intuitively, one would expect the staining of ion channels to be relatively straightforward, as they are most likely solvent accessible. However, it is important to remember that the immunofluorescence epitope may not be easily accessible, depending on whether it is part of an extracellular or intracellular domain. Furthermore, ion channels are often densely clustered on cell membranes, so their detection by immunostaining may require more extensive optimization of the fixation and permeabilization procedures compared to other classes of proteins14,26.
The mitochondrial membrane potential is essential for many mitochondria-associated processes, including ATP synthesis, the generation of reactive oxygen species (ROS), calcium sequestration, the import of mitochondrial proteins, membrane dynamics, and triggering apoptosis. This protocol utilizes the TMRE assay to label membrane potential in active mitochondria in single cells. For high-throughput screens, the TMRE levels can be measured with a fluorescence plate in a microplate format.
Patch-clamp electrophysiology is the “gold standard” method for the study of ion channel kinetics. Whole-cell and single-channel methods are also the highest-resolution methods for accurately determining the structure-function relationships and pharmacology of ion channel modulators. This method utilizes the study of small electrical changes across a membrane caused by the movement of ions through ion channels27. Electrophysiology experiments are traditionally performed using manual patch-clamp electrophysiology recordings of a single cell at a time, resulting in a low-throughput approach that requires the user to possess a series of specialized skill sets. Automated patch-clamp electrophysiology techniques, such as Port-a-Patch, IonFlux Mercury, Patchliner, and/or Synchropatch technologies, offer multiple recordings at a time, and these techniques are equipped with a sophisticated perfusion setup for compound application, resulting in semi-high throughput or high throughput without requiring special skills. For some experiments, manual patch clamping is irreplaceable (Table 1). Still, automated patch-clamp technology has accelerated ion channel biophysics research and related drug discovery programs. One of the limitations of recording from primary/non-transfected cells is the contribution of leaky, non-specific endogenous currents. In our recordings, we observed slight baseline variations, suggesting a potential contribution of endogenous currents from D283 cells.
Cell proliferation assays measure the perturbation of cell growth activity in response to a chemical agent. Such assays are critical tools to assess the action of a drug on cell proliferation. To evaluate cell proliferation, investigators most commonly utilize MTT (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide), MTS (3-[4,5-dimethylthiazol-2-yl]-5-[3-carboxymethoxyphenyl]-2-[4-sulfophenyl]-2H-tetrazolium), and/or clonogenic assays. MTT and MTS assays measure the conversion (reduction) of a water-soluble tetrazolium salt (yellow) into a quantifiable formazan product (purple in the MTT assay), which is catalyzed by the dehydrogenase enzyme system of metabolically active cells. The intensity of the colored product provides an estimation of the number of “viable” (e.g., metabolically active) cells. In contrast, the clonogenic assay analyzes the ability of single cells in culture to form a colony (at least 50 cells or six successive divisions) following treatment with a drug28,29. MTT, MTS, and clonogenic assays each have advantages and disadvantages, and one should carefully assess the suitability of these assays to determine the potency of a drug(s) with a given cell line(s). Optimally, using more than one of these assays (e.g., clonogenic and MTT or MTS assays) is recommended to determine the potency of a drug(s).
The MTT and MTS assays assess the metabolic activity of living cells, which can vary by cell type and assay condition. The MTT or MTS assays are advantageous, as these assays are easy to perform, can be performed in replicate, and are amenable to high-throughput screening30. The MTT or MTS assays can be completed in 3-4 days, while the clonogenic assay can take 10-21 days, depending on the characteristics of the cell line used28. In contrast, the disadvantages of both the MTS and MTT assays are possible interference of the reagents required for performing the assay with culture medium, the difficulty of using the assay, and the potential variability between replicates due to the cell metabolic status and culture conditions31. Regarding the choice between the MTT and MTS assays, one should consider that the MTT assay requires a solubilization step to lyse the cells and allow the formazan crystals to dissolve in the medium and shows absorbance at 570 nm, whereas the MTS assay is a “one-step assay”, in which the formazan is directly solubilized into the medium without the intermittent steps (e.g., the cell lysis step). The formazan product in the MTS assay is darker in color and possesses a more sensitive absorbance value range (490-500 nm), thus making it easier to ascertain a positive response. The MTS assay is faster than the MTT assay, as 2-3 h is needed for the reaction versus 4 h for the reaction plus 1-2 h solubilization in the MTT assay. Additionally, the MTS assay formazan product remains in suspension, and the assay is more suitable for suspension cells than the MTT assay, since no medium change is required during the MTS assay32. However, a modified MTT assay has been developed in which the solubilization step has been improved using a combination of DMSO and SDS lysis solution. This modified MTT assay can be used for both adherent and suspension cells. If one opts to use the MTT assay, then care should be taken in selecting the cell culture and the conditions/reagents used for cell growth. For example, the results will vary if the cells are grown as a monolayer, differentiated, as a confluent monolayer, or senescent. In addition, certain non-mitochondrial dehydrogenases, like some intracellular reductases and flavin oxidases, can reduce the MTT reagent, possibly eliciting false-positive readings33. Further, the MTT reagent has a degree of toxicity, so the incubation time should be limited. The rate of MTT reduction can also change with the culture conditions, such as the pH and glucose content of the medium and the physiological state of the cells32,34. For example, the presence of ascorbic acid reportedly reduces MTT to formazan, and this effect is enhanced in the presence of retinol35. Regarding potential issues with the clonogenic assay, it is important to be aware that following drug addition, the cells can become prematurely senescent and “clonogenically inactive” (i.e., they do not form colonies) but remain metabolically active and exhibit activity in the MTT or MTS assays. Another point to remember is that the clonogenic assay is limited to the study of adherent cells, and not all adherent cells can form colonies when plated at low cell densities, since cell-cell communication is lost36. Due to inadequate cell-cell communication and the limitations of self-produced growth factors, the clonogenic assay responds to lower drug doses than the MTT and MTS assays.
The authors have nothing to disclose.
The authors acknowledge support from the Thomas E. & Pamela M. Mischell Family Foundation to S.S. and the Harold C. Schott Foundation funding of the Harold C. Schott Endowed Chair, UC College of Medicine, to S.S.
ABS SpectraMax Plate Reader | Molecular Devices | ABS | |
Accutase | Invitrogen | 00-4555-56 | |
Alexa Flor 488 | Invitrogen | A32723 | Goat Anti-Rabbit |
Antibiotic-Antimycotic | Gibco | 15240-062 | 100x |
B27 Supplement | Gibco | 12587-010 | Lacks vitamin A |
Biosafety Cabinet | LABCONCO | 302381101 | Class II, Type A2 |
Bovine Serum Albumin | Fisher Scientific | BP1606-100 | |
CO2 Incubator | Fisher Scientific | 13-998-211 | Heracell VIOS 160i |
Calcium Chloride | Fisher Scientific | C7902 | Dihydrate |
Cell Culture Dishes, 150 mm | Fisher Scientific | 12-600-004 | Cell culture treated |
Cell Culture Flasks, 75 cm2 | Fisher Scientific | 430641U | Cell culture treated |
Cell Culture Plates, 6 well | Fisher Scientific | 353046 | Cell culture treated |
Cell Culture Plates, 96 well | Fisher Scientific | 353072 | Cell culture treated |
Centrifuge | Eppendorf | EP-5804R | Refrigerated |
Corning CoolCell | Fisher Scientific | 07-210-0006 | |
Coverslips, 22 x 22 mm | Fisher Scientific | 12-553-450 | Corning brand |
D283 Med | ATCC | HTB-185 | |
DABCO Mounting Media | EMS | 17989-97 | |
D-Glucose | Sigma Life Sciences | D9434 | |
Dimethyl Sulfoxide | Sigma Aldrich | D2650 | Cell culture grade |
DMEM/F12, base media | Fisher Scientific | 11330-032 | With phenol red |
DMEM/F12, phenol red free | Fisher Scientific | 21041-025 | |
EGTA | Sigma Aldrich | E4378 | |
Epidermal Growth Factor | STEMCELL | 78006.1 | |
FCCP | Abcam | AB120081 | |
Fetal Bovine Serum, Qualified | Gibco | 10437-028 | |
Fibroblast Growth Factor, Basic | Millipore | GF003 | |
GARBA5 Antibody | Aviva | ARP30687_P050 | Rabbit Polyclonal |
Glutamax | Gibco | 35050-061 | |
Glycerol Mounting Medium | EMS | 17989-60 | With DAPI+DABCO |
Hemocytometer | Millipore Sigma | ||
Heparin | STEMCELL | 7980 | |
HEPES | HyClone | SH3023701 | Solution |
HEPES | Fisher Scientific | BP310-500 | Solid |
ImageJ | Open platform | With Fiji plugins | |
Immuno Mount DAPI | EMS | 17989-97 | |
KRM-II-08 | experimental compounds not available from a commercial source | ||
Leica Application Suite X | Leica Microsystems | ||
Leukemia Inhibitory Factor | Novus | N276314100U | |
L-Glutamine | Gibco | 25030-081 | |
Magnesium Chloride | Sigma Aldrich | M9272 | Hexahydrate |
Microscope, Confocal | Leica | SP8 | |
Microscope, Light | VWR | 76382-982 | DMiL Inverted |
MTS – Promega One Step | Promega | G3581 | |
Multi-channel pipette, 0.5-10 µL | Eppendorf | Z683914 | |
Multi-channel pipette, 10-100 µL | Eppendorf | Z683930 | |
Multi-channel pipette, 30-300 µL | Eppendorf | Z683957 | |
Nest-O-Patch | Heka | ||
Neurobasal-A Medium | Gibco | 10888022 | Without vitamin A |
Neurobasal-A Medium | Gibco | 12348-017 | Phenol red free |
Non-Essential Amino Acids | Gibco | 11140-050 | |
NOR-QH-II-66 | experimental compounds not available from a commercial source | ||
Parafilm | Fisher Scientific | 50-998-944 | 4 inch width |
Paraformaldehyde | EMS | RT-15710 | |
PATHCHMASTER | Heka | ||
Penicillin-Streptomycin | Gibco | 15140-122 | |
Perfusion System | Nanion | 4000120 | |
PFA | EMS | RT-15710 | |
Phosphate Bufered Saline | Fisher Scientific | AAJ75889K2 | Reagent grade |
Poly-D-Lysine | Fisher Scientific | A3890401 | |
Poly-L-Lysine | Sigma Life Sciences | P4707 | |
Port-a-Patch | Nanion | 21000072 | |
Potassium Chloride | Sigma Life Sciences | P5405 | |
Primary Antibody | Invitrogen | MA5-34653 | Rabbit Monoclonal |
Prism | GraphPad | ||
Propofol | Fisher Scientific | NC0758676 | 1 mL ampule |
QH-II-66 | experimental compounds not available from a commercial source | ||
Reagent Reservoirs | VWR | 89094-664 | Sterile |
Slides, 75 x 25 mm | Fisher Scientific | 12-544-7 | Frosted one side |
Sodium Bicarbonate | Corning | 25-035-Cl | |
Sodium Chloride | Fisher Scientific | S271-3 | |
Sodium Pyruvate | Gibco | 11360-070 | |
Synth-a-Freeze Medium | Gibco | R00550 | Cryopreservation |
TMRE | Fisher Scientific | 50-196-4741 | Reagent |
TMRE Kit | Abcam | AB113852 | Kit |
Triton X-100 | Sigma Aldrich | NC0704309 | |
Trypan Blue | Gibco | 15-250-061 | Solution, 0.4% |
Trypsin/EDTA | Gibco | 25200-072 | Solution, 0.25% |
Vortex Mixer | VWR | 97043-562 | |
Whatman Filter Paper | Fisher Scientific | 09-927-841 |