Waiting
Login processing...

Trial ends in Request Full Access Tell Your Colleague About Jove

Immunology and Infection

A Calcium Bioluminescence Assay for Functional Analysis of Mosquito (Aedes aegypti) and Tick (Rhipicephalus microplus) G Protein-coupled Receptors

Published: April 20, 2011 doi: 10.3791/2732

Summary

This protocol provides instructions for clonal-cell line selection and a calcium bioluminescence assay to analyze the structure-activity relationships of synthesized arthropod neuropeptides on their cognate GPCRs. This assay can be used for receptor deorphanization and structure-activity relationship studies for synthetic analog design and peptide/drug-lead discovery.

Abstract

Arthropod hormone receptors are potential targets for novel pesticides as they regulate many essential physiological and behavioral processes. The majority of them belong to the superfamily of G protein-coupled receptors (GPCRs). We have focused on characterizing arthropod kinin receptors from the tick and mosquito. Arthropod kinins are multifunctional neuropeptides with myotropic, diuretic, and neurotransmitter function. Here, a method for systematic analyses of structure-activity relationships of insect kinins on two heterologous kinin receptor-expressing systems is described. We provide important information relevant to the development of biostable kinin analogs with the potential to disrupt the diuretic, myotropic, and/or digestive processes in ticks and mosquitoes.

The kinin receptors from the southern cattle tick, Boophilus microplus (Canestrini), and the mosquito Aedes aegypti (Linnaeus), were stably expressed in the mammalian cell line CHO-K1. Functional analyses of these receptors were completed using a calcium bioluminescence plate assay that measures intracellular bioluminescence to determine cytoplasmic calcium levels upon peptide application to these recombinant cells. This method takes advantage of the aequorin protein, a photoprotein isolated from luminescent jellyfish. We transiently transfected the aequorin plasmid (mtAEQ/pcDNA1) in cell lines that stably expressed the kinin receptors. These cells were then treated with the cofactor coelenterazine, which complexes with intracellular aequorin. This bond breaks in the presence of calcium, emitting luminescence levels indicative of the calcium concentration. As the kinin receptor signals through the release of intracellular calcium, the intensity of the signal is related to the potency of the peptide.

This protocol is a synthesis of several previously described protocols with modifications; it presents step-by-step instructions for the stable expression of GPCRs in a mammalian cell line through functional plate assays (Staubly et al., 2002 and Stables et al., 1997). Using this methodology, we were able to establish stable cell lines expressing the mosquito and the tick kinin receptors, compare the potency of three mosquito kinins, identify critical amino acid positions for the ligand-receptor interaction, and perform semi-throughput screening of a peptide library. Because insect kinins are susceptible to fast enzymatic degradation by endogenous peptidases, they are severely limited in use as tools for pest control or endocrinological studies. Therefore, we also tested kinin analogs containing amino isobutyric acid (Aib) to enhance their potency and biostability. This peptidase-resistant analog represents an important lead in the development of biostable insect kinin analogs and may aid in the development of neuropeptide-based arthropod control strategies.

Protocol

1. Establishment of stable cell lines

  1. Clone your GPCR of interest and insert it into an expression vector incorporating a 5' Kozak consensus sequence (GCCA/GCCATGG) around the start codon for optimal ribosomal binding in a mammalian system (Kozak, 1986). Here we use the plasmid pcDNA3.1/Bm-KR for the tick kinin receptor, and the plasmid pcDNA3.1/Aedae-KR for the mosquito kinin receptor (Holmes et al., 2003; Pietrantonio et al., 2005). The pcDNA3.1(-) vector encodes ampicillin resistance for selection in bacteria and neomycin (G418) resistance for selection in mammalian cells.
  2. Grow CHO-K1 empty cells (without plasmids) (ATCC, Manassas, VA, USA) or other desired cell line in a T-25 flask (BD Falcon) at 37°C in a 5% CO2 humidified incubator (Holmes et al., 2003). All further incubations should be done under these conditions unless specified otherwise. Maintain the empty cells in growth medium (F12K medium with 10% fetal bovine serum) with 1X Antibiotic-Antimycotic (Invitrogen,CA).
  3. To split cells, warm up all solutions to 37 °C. Remove old medium in the T-25 flask and rinse with 5 ml PBS and then remove PBS. To trypsinize cells, add 2 ml PBS-Trypsin-EDTA (34 ml PBS, 2 ml 7.5% sodium bicarbonate, 4 ml 10X trypsin-EDTA) for 2 min. Add 3 ml medium and aspirate medium up and down to mix cells. Transfer medium with cells into a conical tube and centrifuge for 2 min at ˜200-300 g (1,000 rpm). Discard supernatant and re-suspend cells in 5 ml medium. Dilute the resuspended cells to a concentration of 1:5 or 1:10 with fresh medium and transfer 5 ml in a new T-25 flask.
  4. After the cells are growing healthily (around 2-3 days), seed the CHO-K1 cells into T-25 tissue culture flasks and grow them overnight in growth medium without antibiotics until they are about 30% confluent (approximately 18 hours). The degree of confluency can be determined by observing cells under inverted fluorescent microscopy.
  5. Prepare the following mixtures for each sample:
    1. Combine 1-2 μg DNA (example: use 4μl of 265μg/μl pcDNA3.1/Aedae-KR) with 100 μl Opti-MEM I Reduced Serum Medium (Invitrogen).
    2. Mix 6 μl Lipofectin Reagent (InvitrogenTM) into 100 μl F12K serum free medium, making a 1:1 ratio of Lipofectin to DNA. Incubate at room temperature for 30-45 min.
    Gently mix the solutions from 1.5.1 with 1.5.2 in a drop-wise fashion (total volume = 200 μL). Let stand at room temperature for 10-15 min.
  6. Remove the old growth medium from the cells and wash the cells with 5 ml F12K serum free medium and then remove the F12K serum free medium. In a 15 ml tube, gently mix the transfection mixture into 1.8 ml of fresh F12K medium in a drop-wise fashion. Then, washed the cells from step 1.5 with PBS and add this new transfection solution to the cells. Incubate for 18 hours.
  7. Change the medium to F12K medium plus 10% fetal bovine serum without antibiotics and incubate overnight. Split the cells into two T-25 flasks with F12K medium plus 10% fetal bovine serum without antibiotics for another 18 hours (for splitting cells please see step 1.3).
  8. Replace the medium with selective medium (F12K medium plus 10% fetal bovine serum with 800μg/ml GENETICIN, Invitrogen). Culture the cells using the selective medium for 3-4 weeks. Over time this will select for cells that have stably incorporated the plasmid into their genomic DNA. Continue maintaining the cells using maintenance medium (F12K medium plus 10% fetal bovine serum with 400μg/ml GENETICIN). Periodically freeze cell lines with 1:1 ratio of freezing media (selective medium with 20% DMSO) to prevent loses in case of unexpected contamination.
  9. Selecting clonal cell lines: as a first step, trypsinize and centrifuge the cells from step 1.8 with maintenance medium as in step 1.3. Re-suspend cells in 5 ml maintenance medium, take 0.5 ml of the cell suspend and add 4.5 ml of fresh maintenance medium to make a 10x dilution. Transfer the 10x dilution into 12 wells of a 96 well plate, adding 100 μl to each well to select for single cells.
  10. Continue to make 10x serial dilutions of these cells for a theoretical final suspension of one cell per 100 μl (normally the final dilution will be in the range of 10-11 to 10-19; the total number of 10x serial dilutions is ˜19). Immediately following each dilution, transfer 100 μl of the dilution into 12 wells of the 96 well plate. After 18 hours incubation, observe 96 well plates under an inverted light or fluorescence microscope and mark wells that appear to only contain one single cell or contain two cells that obviously divided from one single cell. Keep observing every day and change medium every three days.
  11. When the wells are 80% confluent (about a week), rinse marked wells with 200 μl PBS and trypsinize them with 100 μl of PBS-trypsin-EDTA solution. Transfer cells from each marked well into one well of a 6-well plate with 1 ml maintenance medium. Grow those cells for 3 days then transfer cells into individual T25 flask. Test these cells using the calcium bioluminescence assay with agonist peptides (please see section 2).
  12. Select 1 cell line from step 1.11 with the highest response in the calcium bioluminescence plate assay and perform for a second time the single cell selection following steps 1.9-1.11. Periodically freeze cell lines.
  13. From the secondary selection described in step 1.12, choose 2-3 cell lines with the highest response in the calcium bioluminescence plate assay and maintain them in culture for further calcium bioluminescence plate assays. Keep track of passage numbers. From time to time, freeze cell lines from early passages so that you can always go back to them if the cell lines with more passages stop performing consistently.

2. The calcium bioluminescence plate assay

  1. Ligate the reporter gene of interest into an expression vector. Here we used aequorin plasmid mtAEQ/pcDNA1 (a gift from Drs. C. J. P. Grimmelikhuijzen and Michael Williamson, University of Copenhagen, Denmark). Transform the plasmid in E. coli cells MC1061/PS (Invitrogen) and purify them using a QIAprep spin miniprep kit (Qiagen Inc.). In the final step elute the plasmid with Tris buffer without EDTA, not water.
  2. Grow cell lines from step 1.13 expressing the desired receptor in maintenance medium. When the cells are 90% confluent, trypsinize, centrifuge and then re-suspend the cells in 5 ml maintenance medium as in step 1.3. Dilute cells (about 10x with maintenance medium) and count cell number with cell counter (Bright-Line Hemacytometer) under microscopy. Adjust the cell number to approximately 2 x 105 cells/ ml (average 20 cells in one of the 9 squares showed in the Hemacytometer). Seed 2 ml diluted cells in media into each well of a six well plate. Incubate for 24 hours (the cells should reach about 60% confluency after incubation).
  3. Change media in the 6 wells plate to OPTI-MEM medium. For each well, mix 96 μl OPTI-MEM with 4 μl FuGENE 6 Transfection reagent (Roche Biochemicals) in a microfuge tube and let sit at room temperature for 5 min. Add 1 μg of aequorin/pcDNA 1 plasmid DNA into each tube and then gently shake the sample for 1 min, incubate at room temperature for 15-20 min. Add each mixture into each well in a dropwise fashion while gently shaking the well plate. Incubate the plates for 6 hours and change the medium to F12K medium containing 10% fetal bovine serum without antibiotic.
  4. After incubating the cells in six well plate for 24 hours, trypsinize, centrifuge and re-suspend the cells as step 1.3. Count the cell number to 400,000 cells/ml as step 2.1 and transfer 100 μl (total 40,000 cells/100 μl) into each well of a 96-well white thin bottom microtitre plate (Costar 3610). Incubate for another 24 hours until about 80% cell confluency. This is the optimal concentration of cells for the bioluminescence assay.
  5. Prepare 90μl/well of a calcium-free DMEM media (Invitrogen) containing 5 μM coelenterazine (Invitrogen) in the dark (coelenterazine is light sensitive). Take the plate from 2.4, remove old media and add this 90μl into each well. Incubate plates for 3 hours in the dark at 37°C and 5% CO2, after which the cells in the plate are ready to be tested.

3. Instrument operation and data analysis

  1. Each bioluminescence plate reader is different. We perform our assay using a NOVOstar (BMG Labtechnologies) plate reader in bioluminensence mode. If you use a different instrument, you must adapt the protocol.
  2. Purge the plate reader pumps (or PRIME PUMPS) before use. Turn off the light in the room before putting the plate on the plate holder. Once the plate holder has closed, turn the lights on.
  3. Solubilize peptides (in a 1.5 ml Eppendorf tube) in the calcium-free DMEM media. Set the "Aspirate Depth" and "Position Determination" of the peptide solution before use. Challenge the cells with 10 μl (10x) of varying concentrations of the peptides (FFFSWG-NH2, Aedes-K1-3, or other desired peptide) and immediately begin recording the light emission. We have set the instrument to record the light emission (465 nm) for each well every 2 seconds for a total time of 50 seconds.
  4. Make sure to include a positive control such as an active analog (analog FFFSWGa has been used, Taneja-Bageshwar et al., 2009) and a negative control such as vector only transfected cells. The negative control will be necessary during data analysis to set the baseline threshold (see representative results). An unrelated, inactive peptide can also be added as a negative control.
  5. After the run is complete, wash the instrument pump (or PRIME PUMPS) then place your next peptide sample. Save your data and wash the pumps again.
  6. Data handling: Transfer the data of light emission from each well into a Microsoft Excel data sheet.
  7. Paste the data from Excel to Prism software 4.0 from GraphPad Software Inc. (San Diego, CA, USA). The various peptide concentrations is the X-axis and bioluminescence units is the Y-axis. To normalize the data, plot a log-response curve. Select a nonlinear regression curve fit analysis (sigmoidal dose-response equation with variable slope) to obtain concentration-response curves for each peptide. The program plots the values in the end and gives the EC50.
  8. Each experiment should be repeated three times for data analysis.

4. Representative Results:

When expressed in CHO-K1 cells, the mosquito Aedes aegypti kinin receptor behaved as a multiligand receptor and functionally responded to concentrations as low as 1 nM of the three endogenours Aedes kinins, Aedae kinins 1-3, tested singly using the calcium bioluminescence plate assay. Figure 1 shows that the rank order of potency obtained was Aedae-K-3 > Aedae-K-2 > Aedae-K-1, based on the respective EC50 values of Aedae-K-3, 16.04 nM; Aedae-K-2, 26.6 nM and Aedae-K-1, 48.85 nM, which were statistically significantly different (P < 0.05) (Pietrantonio et al., 2005).

We also used this assay to determine which kinin residues are critical for the kinin peptide-receptor interaction. Insect kinin peptides share a C-terminal pentapeptide that represents the minimal sequence required for biological activity, also known as core. In Table 1, the kinin peptide core analogs were synthesized as an Alanine replacement series of the core kinin pentapeptide FFSWGa and tested by a calcium bioluminescence plate assay (Taneja-Bageshwar et al., 2006). We found that the amino acids Phe1 and Trp4 were essential for activity of the insect kinins for both receptors.

The assay also can be used to test peptides designed for enhanced biostability. Table 2 shows the designed kinin analogs containing amino isobutyric acid (Aib) tested on the tick recombinant kinin receptor and Figure 2 shows the activity comparison of six alpha-amino isobutyric acid analogs on the tick kinin receptor expressing CHO-K1 cell line by a calcium bioluminescence plate assay (Taneja-Bageshwar et al., 2009). The hexapeptide analog FFFSWGa is added for a positive control for receptor activity. The analog FF[Aib]WGa resulted more active than this hexapeptide control analog. The analog with two aminoisobutyric acid replacements, [Aib]FF[Aib]WGa, was the most potent of the double-replacement analogs tested (Table 2 and Figure 2).

For more examples of how this assay has been and can be applied see Nachman and Pietantonio (2010), Nachman et al. (2009), Taneja-Bageshwar et al. (2008a), and Taneja-Bageshwar et al. (2008b).

Figure 1
Figure 1. Estimation of Aedes kinins effective concentration (EC50) on CHO-K1 E10 cells by a calcium-bioluminescence plate assay. The y-axis in the concentration-response curves was obtained from bioluminescence units expressed as a percentage of the maximal response observed for each peptide. Data points represent the average of six replicates obtained during three independent experiments. Bars represent the standard error. (A) Estimation of Aedae-K1 EC50 = 48 nM. (B) Estimation of Aedae-K2 EC50 = 26 nM. (C) Estimation of Aedae-K3 EC50 = 16 nM. EC50 Aedae-K3 < EC50 Aedae-K2 < EC50 Aedae-K1; P < 0.05. Statistical analysis and graphs were with the GraphPad Prism 4.0 software.

Figure 2
Figure 2. Activity comparison of six alpha-amino isobutyric acid analogs on the tick kinin receptor expressing CHO-K1 cell line by a calcium bioluminescence plate assay. The y-axis represents percent maximal bioluminescence units for each analog expressed as a percentage of bioluminescence observed at a concentration versus the maximal response observed among all concentrations tested for each analog. Statistical analysis and graphs were performed with GraphPad Prism 4.0 software.

  Tick receptor cell line Mosquito receptor cell line
Peptides EC50 (nM) Maximal bioluminescence response at 1 mM EC50 (nM) Maximal bioluminescence response at 1 mM
AFSWGa I I I I
FASWGa 586 5,600 N.D. 400
FFAWGa 64 12,800 621 3,050
FFSAGa I I I I
FFSWAa 417 10,600 2,800 1,830
FFSWGa 590 10,800 N.D. 525
FSWGa I I I I
FFSWa I I I I
FFSWG-OH I I I I
FFFSWGa 259 13,000 562 10,000
FF[Aib]WGa 29 12,700 445 9,300

Table 1. Estimated potencies (EC50) and maximal bioluminescence response of all the peptides tested on tick and mosquito receptor transfected cell lines*.

*The EC50 estimates the concentration required to induce a half-maximal response. I: Inactive if bioluminescence response is less than 300 units (level of vector-only transfected cells). A: The position where the respective residue in the peptide FFSWGa has been replaced by alanine. N.D.: The analog was tested but was either not very active or was not active at lower molarities, thus an EC50 could not be determined.

K-Aib-1 [Aib]FF[Aib]WGa
K-Aib-2 [α MeF]FF[Aib]WGa
K-Aib-3 Ac-R[Aib]FF[Aib]WGa
K-Aib-4 Ac-R[β3F]FF[Aib]WGa
K-Aib-5 [Aib]RFF[Aib]WGa
K-Aib-6 [Aib-Aib-Aib-Aib]RFF[Aib]WGa

Table 2. Kinin analogs (K) containing amino isobutyric acid (Aib) tested on the tick recombinant kinin receptor. Ac: acetyl; α Me: α methyl- phenylalanine; β3F: β3 -phenylalanine; a: amide.

Discussion

We were able to perform the functional characterization of the first neuropeptide receptor discovered from the Arachnida (ticks, mites and spiders), the tick kinin receptor, using this protocol. This method has three primary applications. First, the technique can be applied for receptor deorphanization through ligand activity measurements. Second, the assay can resolve ligand-receptor structure-activity relationships (SAR). Third, the methods can be used in drug discovery. Furthermore, this protocol can be used to study the activity of agonists or antagonists on almost any GPCR. We are beginning to adapt this protocol for screening of small libraries. The cell line we utilized does not express the ubiquitous G protein G16. We did not need it because arthropod kinin receptors signal through the Gq protein and the intracellular calcium cascade and they conserve this signaling properties in mammalian cells as shown here.

Disclosures

No conflicts of interest declared.

Acknowledgments

Drs. C. J. P. Grimmelikhuijzen and Michael Williamson from University of Copenhagen (Denmark) are appreciated for providing the aequorin plasmid. Our collaborator, Dr. Ronald J. Nachman from ARS-USDA (TX, USA), is acknowledged for peptide synthesis and for providing the NOVOstar plate reader.

Materials

Name Company Catalog Number Comments
DMEM Invitrogen ABCD1234
CHO-K1 cells ATCC CCL-61 Manassas, VA, USA
F12K medium Invitrogen 21127
Fetal Bovine Serum Sigma-Aldrich F0643
Trypsin-EDTA (10x) Invitrogen 15400
Antibiotic-Antimycotic Invitrogen 15240
Opti-MEM I Reduced Serum Medium Invitrogen 31985
Lipofectin Reagent Invitrogen 18292-011
GENETICIN Invitrogen 10131035
MC1061/P3 Ultracomp Invitrogen C663-03
QIAprep spin miniprep kit Qiagen 19064
FuGENE 6 Transfection reagent Roche Group 11 814 443 001
96-well white thin bottom microtitere plate Costar 3610
calcium-free DMEM media Invitrogen 21068
C–lenterazine Invitrogen C-2944
Bright-Line Hemacytometer Hausser Scientific Horsham, PA
NOVOstar BMG Labtechnologies
Prism software 4.0 GraphPad Software Inc. San Diego, CA, USA
T-25 and T-75 Flasks BD Biosciences 353014 and 353135

DOWNLOAD MATERIALS LIST

References

  1. Kozak, M. An analysis of 5'-noncoding sequences from 699 vertebrate messenger RNAs. Nucleic Acids Research. 15, 8125-8148 (1987).
  2. Nachman, R., Pietrantonio, P. V. Interaction of mimetic analogs of insect kinin neuropeptides with arthropod receptors. Neuropeptide systems as Targets for Parasite and Pest Control. Geary, T. , Landes Bioscience. Madamme Curie Library. (2010).
  3. Nachman, R. J., Pietrantonio, P. V., Coast, G. M. Towards the development of novel pest management agents based upon insect kinin neuropeptide analogs. Annals of the New York Academy of Sciences. , 1163-11251 (2009).
  4. Taneja-Bageshwar, S., Strey, A., Isaac, R. E., Coast, G. M., Zubrzak, P., Pietrantonio, P. V., Nachman, R. J. Biostable agonists that match and/or exceed the activity of insect kinins on recombinant arthropod GPCRs. General and Comparative Endocrinology. 162, 122-128 (2009).
  5. Taneja-Bageshwar, S., Strey, A., Kaczmarek, K., Zabrocki, J., Pietrantonio, P. V., Nachman, R. J. Comparison of insect kinin analogs with cis-peptide bond, type VI-turn motifs identifies optimal stereochemistry for interaction with a recombinant arthropod kinin receptor from the southern cattle tick Boophilus microplus. Peptides. 29, 295-301 Forthcoming.
  6. Taneja-Bageshwar, S., Strey, A., Zubrzak, P., Williams, H., Reyes-Rangel, G., Juaristi, E., Pietrantonio, P. V., Nachman, R. J. Identification of selective and non-selective, biostable beta-amino acid agonists of recombinant insect kinin receptors from the southern cattle tick Boophilus microplus and mosquito Aedes aegypti. Peptides. 29, 302-309 Forthcoming.
  7. Taneja-Bageshwar, S., Strey, A., Zubrzak, P., Pietrantonio, P. V., Nachman, R. J. Comparative structure-activity analysis of insect kinin core analogs on recombinant kinin receptors drom southern cattle tick Boophilus microplus (Acari: Ixodidae) and mosquito Aedes aegypti (Diptera: Culicidae). Archives of Insect Biochemistry and Physiology. 62, 128-140 (2006).
  8. Pietrantonio, P. V., Jagge, C., Taneja-Bageshwar, S., Nachman, R. J., Barhoumi, R. The mosquito Aedes aegypti (L.) leucokinin receptor is a multiligand receptor for the three Aedes kinins. Insect Molecular Biology. 14, 55-67 (2005).
  9. Holmes, S. P., Barhoumi, R., Nachman, R. J., Pietrantonio, P. V. Functional analysis of a G protein-coupled receptor from the Southern cattle tick Boophilus microplus (Acari: Ixodidae) identifies it as the first arthropod myokinin receptor. Insect Molecular Biology. 12, 27-38 (2003).
  10. Staubli, F., Jorgensen, T. J. D., Cazzamali, G., Williamson, M., Lenz, C., Sondergaard, L., Roepstorff, P., Grimmelikhuijzen, C. J. P. Molecular identification of the insect adipokinetic hormone receptors. Proceedings of the National Academy of Sciences USA. 99, 3446-3451 (2002).
  11. Stables, J., Green, A., Marshall, F., Fraser, N., Knight, E., Sautel, M., Milligan, G., Lee, M., Rees, S. A bioluminescent assay for agonist activity at potentially any G-protein coupled receptor. Analytical Biochemistry. 252, 115-126 (1997).

Tags

Calcium Bioluminescence Assay Functional Analysis Mosquito Aedes Aegypti Tick Rhipicephalus Microplus G Protein-coupled Receptors Arthropod Hormone Receptors Pesticides Kinin Receptors Neuropeptides Structure-activity Relationships Heterologous Kinin Receptor-expressing Systems Biostable Kinin Analogs Diuretic Processes Myotropic Processes Digestive Processes Southern Cattle Tick Boophilus Microplus Aedes Aegypti CHO-K1 Cell Line Calcium Levels Peptide Application Recombinant Cells Aequorin Protein
A Calcium Bioluminescence Assay for Functional Analysis of Mosquito (<em>Aedes aegypti</em>) and Tick (<em>Rhipicephalus microplus</em>) G Protein-coupled Receptors
Play Video
PDF DOI DOWNLOAD MATERIALS LIST

Cite this Article

Lu, H., Kersch, C. N.,More

Lu, H., Kersch, C. N., Taneja-Bageshwar, S., Pietrantonio, P. V. A Calcium Bioluminescence Assay for Functional Analysis of Mosquito (Aedes aegypti) and Tick (Rhipicephalus microplus) G Protein-coupled Receptors. J. Vis. Exp. (50), e2732, doi:10.3791/2732 (2011).

Less
Copy Citation Download Citation Reprints and Permissions
View Video

Get cutting-edge science videos from JoVE sent straight to your inbox every month.

Waiting X
Simple Hit Counter