Waiting
Procesando inicio de sesión ...

Trial ends in Request Full Access Tell Your Colleague About Jove

Biology

Measuring Relative Insulin Secretion using a Co-Secreted Luciferase Surrogate

Published: June 25, 2019 doi: 10.3791/59926

Summary

This protocol describes how to perform rapid low-cost luciferase assays at medium-throughput using an insulin-linked Gaussia luciferase as a proxy for insulin secretion from beta cells. The assay can be performed with most luminescence plate readers and multichannel pipettes.

Abstract

Performing antibody-based assays for secreted insulin post-sample collection usually requires a few hours to a day of assay time and can be expensive, depending on the specific assay. Secreted luciferase assays expedite results and lower the assay cost per sample substantially. Here we present a relatively underused approach to gauge insulin secretory activity from pancreatic β cells by using Gaussia luciferase genetically inserted within the C-peptide. During proteolytic processing of proinsulin, the C-peptide is excised releasing the luciferase within the insulin secretory vesicle where it is co-secreted with insulin. Results can be obtained within minutes after sample collection because of the speed of luciferase assays. A limitation of the assay is that it is a relative measurement of insulin secretion and not an absolute quantitation. However, this protocol is economical, scalable, and can be performed using most standard luminescence plate readers. Analog and digital multichannel pipettes facilitate multiple steps of the assay. Many different experimental variations can be tested simultaneously. Once a focused set of conditions are decided upon, insulin concentrations should be measured directly using antibody-based assays with standard curves to confirm the luciferase assay results.

Introduction

The method presented here allows insulin secretion from a genetically-modified beta cell line to be assayed rapidly and affordably in 96-well-plate format. The key to this protocol is a modified version of insulin with the naturally-secreted Gaussia luciferase (GLuc, ~18 kDa) inserted (see Figure 1) into the C-peptide to generate insulin-Gaussia (InsGLuc)1,2. Other larger proteins, such as GFP (~25 kDa), have been successfully inserted into the C-peptide of insulin and exhibited the expected post-translational processing from proinsulin-GFP to insulin and GFP-C-peptide3,4. For the assay in this protocol, GLuc has been codon-optimized for mammalian expression and two mutations have been introduced to enhance glow-like kinetics5,6. Multiple combinations and replicates of treatment conditions can be easily tested in 96-well-plate format and the secretion results can be obtained immediately following the experiment.

A major advantage, as previously noted2, is the low cost of this luciferase-based secretion measurement (< $0.01/well) which differentiates it from the relatively higher costs and technical aspects of enzyme-linked immunosorbent assays (ELISAs) (> $2/well) and homogenous time-resolved fluorescence (HTRF) or other Förster resonance energy transfer (FRET)-based antibody (> $1/well) assays. In comparison to these antibody-based assays, which measure the concentration of insulin by referencing a standard curve, the InsGLuc assay measures secretory activity as a relative comparison to control wells on the plate. For that reason, every experiment requires the inclusion of proper controls. This distinction is a trade-off to allow rapid and inexpensive measurements. However, InsGLuc secretion has been demonstrated to be highly correlated with insulin secretion as measured by ELISA1,2. This technology has been scaled up for high-throughput screening1,2,7 and has led to the identification of novel modulators of insulin secretion including a voltage-gated potassium channel inhibitor7 as well as a natural product inhibitor of β cell function, chromomycin A28. The use of InsGLuc is most appropriate for researchers who plan to continually test many different treatment conditions for their impact on insulin secretion. In follow-up experiments it is necessary to repeat key findings in a parental β cell line, and optimally in murine or human islets, and measure insulin secretion using an antibody-based assay.

Subscription Required. Please recommend JoVE to your librarian.

Protocol

1. Preparation of reagents, media and buffers (Table 1)

  1. Prepare MIN6 complete media in 500 mL of high-glucose (4.5 g/L) Dulbecco's modified Eagle medium (DMEM) with the following additives: 15% fetal bovine serum (FBS), 100 units/mL penicillin, 100 μg/mL streptomycin, 292 μg/mL L-glutamine, and 50 μM β-mercaptoethanol.
    NOTE: The stable cell line in this case is maintained in 250 µg/mL of G418 antibiotic.
  2. Prepare Krebs-Ringer bicarbonate buffer (KRBH) by making a solution containing 5 mM KCl, 120 mM NaCl, 15 mM HEPES (pH 7.4), 24 mM NaHCO3, 1 mM MgCl2, 2 mM CaCl2, and 1 mg/mL radioimmunoassay-grade bovine serum albumin (BSA). Glucose is to be added where specified from a 2 M stock.
    NOTE: KRBH is used to incubate the cells with and without stimulation in order to assess insulin and/or Gaussia luciferase secretion.
  3. Prepare coelenterazine (CTZ) stock solution as follows. Prepare acidified methanol by adding 106 µL of concentrated HCl to 10 mL of methanol. Next, dissolve lyophilized CTZ in acidified methanol at 1 mg/mL and store at -80 °C in screw-cap tubes.
    NOTE: These stocks retain sufficient activity in routine luciferase assays, even after 1 year of proper storage.
  4. Prepare Gaussia luciferase (GLuc) assay buffer based upon the literature9 as well as patent information10 to aid in half-life of the Gaussia luciferase assay in 96 well plate format. Use the formula: 25 mM Tris pH 8, 1 mM EDTA, 5% glycerol, 1 mg/mL Na2PO4, 300 mM sodium ascorbate, 200 mM Na2SO3 in water. Freeze stocks at -20 °C. After thawing, store the buffer at 4 °C.
  5. To prepare Gaussia luciferase working solution, add 4.2 µL/mL of the 1 mg/mL (2.36 mM) CTZ stock solution to GLuc assay buffer. This results in a 2x working solution of 10 µM CTZ which will have a 5 µM final concentration in the assay.

2. Culture of InsGLuc MIN6 cells and seeding for secretion assays

  1. To culture MIN6 cells, trypsinize and seed the cells once per week using standard cell culture techniques. Change media on the cells every two to three days. Include appropriate selection antibiotic, such as 250 µg/mL of G418 in the media.
    1. To provide a sufficient number of cells weekly for experiments, maintain the cells in T75 flasks. Seeding 6 x 106 cells per T75 in 10 mL of media will typically yield 30 x 106 to 40 x 106 cells total per T75 after 7 days of culture.
  2. To prepare cells for plating into 96-well plates, wash a confluent T75 of InsGLuc MIN6 cells twice with PBS and add 2 mL of trypsin. Incubate at 37 °C for ~5 min or until the cells dissociate from the flask. Determine the cell concentration per milliliter.
    1. Dilute the cells in complete media to 1 x 106 cells/mL to result in 1 x 105 cells in 100 µL per well in a 96-well plate. The cells should be sufficiently confluent for the assay after 3–4 days.
      NOTE: To extend the culture period, half the cell concentration can be plated. Media changes are not required prior to the day of the assay unless the cells are to be subjected to experimental treatments.

3. Glucose-stimulated Gaussia luciferase secretion assay

  1. On the day of the assay prepare enough KRBH for the experiment (step 1.2). A minimum of 50 mL of KRBH per 96-well plate is required, therefore prepare at least 75 mL to ensure sufficient buffer for the experiment. Prepare extra buffer if different combinations of drug treatment conditions will require KRBH for dilution.
  2. Prepare a reservoir with glucose-free KRBH. Decant the medium from 96-well plate(s) by quickly inverting the plate over a laboratory sink and then blot firmly on a stack of paper towels to remove excess medium.
  3. Using either an electronic or manual 8-channel pipette, pipette 100 µL /well KRBH from the reservoir across the 96-well plate(s). Repeat for a total of two washes.
  4. (Optional step of acute compound treatments) If not performing drug treatments, proceed with steps 3.5 and 3.6 without modification. To test the effects of small molecules on insulin secretion, compounds can be added to the cells during the preincubation period, stimulation period, or both.
    1. One technique is to add compounds in batch to the KRBH in 1.5 mL tubes and use an adjustable 8-channel digital pipette to transfer the drug-KRBH from the tubes to cells in 96-well plates.
      NOTE: If an adjustable pipette is not available, a replica 96-well plate of drug-KRBH can be made and a standard 8-channel pipette can be used to transfer buffer. Further modifications to the treatment paradigm can be made to treat cells for 24 h in media prior to the assay, as previously described1,8.
  5. Add 100 µL of KRBH containing the desired concentration of glucose or compounds and place the dish in the 37 °C incubator for 1 h.
    NOTE: Depending on the experimental layout, it is extremely helpful to have an electronic multichannel pipette that allows transitioning the channel distances from a column of eight 1.5-mL tubes to the 8 rows of a 96-well plate.
  6. After the 1 h of preincubation, decant the buffer into the sink and blot firmly on paper towel. Add 100 µL of glucose-free KRBH per well to wash away accumulated background of Gaussia luciferase. Decant the plate again and add control and stimulatory conditions to the plate at 100 µL per well. Place the dish in the 37°C incubator for 1 h.
  7. Carefully collect 50 µL of supernatant using a multichannel pipette, changing tips between treatment conditions as necessary, and transfer the supernatant to a clean opaque white 96-well assay plate.
    NOTE: White-walled clear-bottom plates can be used if necessary, although a significant amount of luciferase signal will be lost.
  8. After collection of 50 µL of KRBH supernatant, the sample can be assayed immediately. If necessary, seal and store samples at 4 °C for a few days (GLuc activity half-life ~6 days) or -20 °C for up to one month11,12.

4. Secreted Gaussia luciferase assay

  1. To prepare the GLuc assay working solution, pipette the required amount of CTZ stock solution (4.2 µL/mL) into GLuc assay buffer. To prevent warming the CTZ, pipette the CTZ quickly at the -80 °C freezer or keep the tube on dry ice.
  2. Using an electronic multichannel pipette, quickly add 50 µL of the GLuc assay working solution per well across the 96-well dish containing the collected KRBH supernatants. If there are any droplets on the sides of any wells, briefly spin the plate in a table-top swing-bucket centrifuge.
  3. Read the luminescence in a suitable plate reader within a few minutes and read each well with a 0.1 s integration time.

Subscription Required. Please recommend JoVE to your librarian.

Representative Results

To gauge the performance of the assay under control conditions, a simple glucose dose-response curve or a stimulation using the diazoxide paradigm can be completed. In the case of the former, pre-incubating the cells for 1 h in glucose-free conditions followed by treating for 1 h with increasing glucose concentrations should result in very little secretory activity at and below 5 mM, while increased secretion is observed above 8 mM glucose (Figure 2). Stimulation with 35 mM KCl also serves as a positive control for stimulated secretion. Inclusion of secretion-modulating drugs during the stimulation period should give the expected inhibition or potentiation of secreted GLuc activity (Figure 3). For example, diazoxide binds the KATP channel and prevents it from closing upon increased [ATP/ADP] ratio, blocking membrane depolarization and preventing secretion13. Phorbol esters like para-methoxyamphetamine (PMA) activate protein kinase C (PKC) and are known amplifiers of insulin secretion14. Finally, stimulation with 1 µM epinephrine activates α2A-adrenergic receptors which in turn activate the heterotrimeric G-protein complex Gi, inhibiting membrane depolarization and insulin secretion15. It is important to recognize that while MIN6 cells are an immortal cell line, they start to lose proper glucose-induced insulin secretion responses (such as left-shifting of the response curve) after extended passaging16. For this reason, it is good practice to routinely culture all MIN6 cell lines for up to eight weeks (splitting once per week) before starting over from liquid nitrogen stocks.

Figure 1
Figure 1: Description of the InsGLuc reporter. (A) First, a stable β cell line (in this case MIN6 cells) was generated expressing the insulin-Gaussia transgene from the rat insulin promoter. (B) The full protein is synthesized and packaged in insulin granules along with endogenous insulin. Prohormone convertases cleave the peptide, indicated by asterisks. (C) The processed insulin and Gaussia are co-secreted and the luciferase activity is detected by the addition of CTZ in an ATP-independent, oxygen-dependent reaction.  Please click here to view a larger version of this figure.

Figure 2
Figure 2: The InsGLuc reporter is a faithful proxy of insulin secretion from MIN6 beta cells. (A) Response of MIN6 InsGLuc cells to increasing glucose concentrations and KCl (35 mM) with Gaussia luciferase secretion. Data are the mean fold luciferase activity ± SE compared to 0 mM glucose conditions for three independent experiments. *, P < 0.05. The figure has been modified with permission from Kalwat et al. ACS Sensors 20161. © 2016 American Chemical Society. (B) The InsGLuc reporter in MIN6 cells exhibits the expected secretory response to the diazoxide (Dz) paradigm where 250 µM Dz treatment holds the KATP channel open, blocking membrane depolarization unless extracellular KCl (35 mM) is provided to elicit the ‘triggering’ calcium influx. Further addition of glucose (20 mM) under the Dz + KCl condition reveals the metabolic amplification of secretion that occurs without further increases in calcium influx. Please click here to view a larger version of this figure.

Figure 3
Figure 3: Inclusion of secretion-modulating compounds during glucose-stimulates InsGLuc secretion. InsGLuc MIN6 cells plated in 96-well format as described were preincubated in glucose-free KRBH for 1 h. Cells were then treated with or without 20 mM glucose in the presence of dimethyl sulfoxide (DMSO) (0.1%), KCl (35 mM), diazoxide (250 µM), PMA (100 nM), or epinephrine (1 µM) for 1 h. Bar graph represent the mean ± SE of at least 3 independent experiments. Please click here to view a larger version of this figure.

Krebs-Ringer Bicarbonate Buffer (KRBH)
Stock solution or powder Stock Final Concentration 100 mL
KCl 0.25 M 5 mM 2 ml
NaCl 4 M 120 mM 3 ml
Hepes, pH 7.4 1 M 15 mM 1.5 ml
NaHCO3 0.5 M 24 mM 4.8 ml
MgCl2 1 M 1 mM 0.1 ml
CaCl2 1 M 2 mM 0.2 ml
water H2O 88.4 ml
RIA-grade BSA powder 1 mg/mL 100 mg
Make fresh on day of experiment.
Gaussia Assay Buffer*
Stock solution or powder Stock Final Concentration 50 mL
Disodium phosphate powder 0.1% (1 mg/mL) 50 mg
Glycerol 40% 5% 6.25 mL
Sodium Bromide powder 150 mM 772 mg
EDTA pH 8 0.5M 1 mM 100 µL
Tris-HCl pH 8 1M 25 mM 1.25 mL
Ascorbic Acid* powder 300 mM 2.64 g
Na2SO3** powder 200 mM 1.26 g
Water up to 50mL
Store in aliquots at -20 °C, thaw one at a time and keep at 4 °C.
*Modified recipe from Luft et al. BMC Biochemistry 2014, 15:14.
Acidified MeOH
Stock solution or powder Stock Final Concentration 10 mL
Methanol 100% 10 mL
HCl 11.65 M 1.06% 0.106 mL
Coelenterazine solution
Stock solution or powder Stock Final Concentration 1 mL
Coelenterazine powder 1 mg/mL (2.36 mM) 1 mg
Acidified methanol 1 mL
4.2 µL of stock per 1 mL of Gaussia Assay Buffer results in 10 µM CTZ to be used as a 2x working solution.
For example, add 50 µL of 2x working solution to 50 µL of KRBH sample containing secreted Gaussia.

Table 1: Buffer and stock solution recipes used to perform the presented assays.

Subscription Required. Please recommend JoVE to your librarian.

Discussion

Herein we present a method to rapidly assess glucose-stimulated insulin secretion responses from MIN6 β cells. For the best responses in the assay it is important to seed the MIN6 cells at the proper density and allow them to become 85-95% confluent. This improves β cell responses to glucose because of improved cell-cell contacts and synchronization, which  occurs both in primary islets17,18,19,20,21 as well as MIN6 cells16,18. To prevent losses in secretory response to glucose stimulation, it is important to maintain the cells at as low of a passage as possible and culture the cells for only 6-8 weeks prior to thawing a new vial from liquid nitrogen stocks. Modifications can be made to the plating strategy in protocol section 2 to adapt to the available equipment as necessary. Plating InsGLuc MIN6 cells into 96-well plates for secretion assays affords a large number of wells for experimental manipulations (including replicates) as well as maintaining accuracy of plating in a normal lab setting, as plating into dishes with higher well numbers often requires special equipment usually available in high-throughput screening cores.

Current assays for insulin secretion, other than the indirect luciferase assay described here, include: ELISAs that use colorimetric readouts (direct assay), radioimmunoassays (competition assay) which use radioactive readouts, FRET-based antibody competition assay22 and HTRF23 which uses FRET between dye-linked antibodies to measure insulin directly, and DNA aptamers24. Each of these methods has its own advantages, but in general they are more expensive and/or time-consuming than a luciferase assay. One key limitation of the InsGLuc assay is the fact that luminescent activity of the co-secreted luciferase is only a proxy for actual insulin secretion. Additionally, there is no expected difference in luciferase activity between Gaussia with fragments of C-peptide on its N- and C-termini or Gaussia within the proinsulin protein, as Gaussia luciferase has been successfully used as a tag to measure the secretion of other proteins25. This highlights the requirement of confirmation studies using assays that measure processed insulin specifically. Alternatives to direct and indirect measurements of insulin secretion can also be used to assess β cell function. A variety of optical reporters exist for readouts including ATP:ADP ratio, calcium influx, NAD+/NADH ratio, extracellular signal-regulated kinases (ERK) activation, or cyclic adenosine monophosphate (cAMP) levels26.

The future applications of InsGLuc in particular appear to be in high-throughput screening. This assay has already been used in a handful of published small screens1,2 and unpublished larger screens are either completed7 or underway. Development of other iterations of this technology may involve tagging of other secreted islet hormones with luciferases to facilitate rapid measurements, such as for glucagon or somatostatin. Modifications could be made in any case where the cell line is recreated using alternate approaches including CRISPR/Cas9, lentivirus, or transposase-mediated insertion in any suitable beta cell line. Additional possible modifications to the original reporter may include substituting alternate secreted luciferases for Gaussia or combining multiple different secreted luciferases linked to different hormones for a multiplexed assay. Beyond cell culture, CRISPR/Cas9 technology presents the possibility of generating a mouse model where a suitable luciferase is knocked in to the C-peptide coding region of Ins2 in the genome. Such a mouse would be feasible given that transgenic mice have been created with GFP knocked in to the same C-peptide site27 and would allow measurement of endogenous β cell function with a luciferase assay in vivo or ex vivo.

Subscription Required. Please recommend JoVE to your librarian.

Disclosures

The authors have nothing to disclose.

Acknowledgments

The authors thank all current and former members of the Cobb laboratory for valuable work and discussions, and Dionne Ware for administrative assistance. Michael Kalwat is supported by a Juvenile Diabetes Research Foundation SRA-2019-702-Q-R. This work was made possible through NIH R37 DK34128 and Welch Foundation Grant I1243 to Melanie Cobb. Early parts of this work were also supported by an NIH F32 DK100113 to Michael Kalwat.

Materials

Name Company Catalog Number Comments
Cell culture materials
rIns-GLuc stable MIN6 cells Parental MIN6 cell line stably expressing pcDNA3.1+rInsp-Ins-eGLuc and maintained in 250 ug/ml G418
DMEM Sigma D6429 4.5 g/L glucose media
fetal bovine serum, heat-inactivated Sigma F4135
Penicillin/Streptomycin Thermo-Fisher Scientific SV30010
beta-mercaptoethanol Thermo-Fisher Scientific BP 176-100
glutamine Thermo-Fisher Scientific BP379-100
Trypsin-EDTA Sigma T3924-500
G418 Gold Biotechnology G418-10 Stock solution 250 mg/mL in water. Freeze aliquots at -20C.
T75 tissue culture flasks Fisher Scientific 07-202-000
96 well tissue culture plates Celltreat 229196
Reagent reservoirs (50 mL) Corning 4870
Name Company Catalog Number Comments
Secretion assay reagents
BSA (RIA grade) Thermo-Fisher Scientific 50-146-952
D-(+)-Glucose Sigma G8270-1KG
KCl Thermo-Fisher Scientific P217-500
NaCl Thermo-Fisher Scientific S271-3
Hepes, pH 7.4 Thermo-Fisher Scientific 50-213-365
NaHCO3 Thermo-Fisher Scientific 15568414
MgCl2 Thermo-Fisher Scientific M9272-500G
CaCl2 Sigma C-7902
Name Company Catalog Number Comments
Optional drugs for stimulation experiments
Diazoxide Sigma D9035 Stock solution: 50 mM in 0.1N NaOH. Add equal amount of 0.1N HCl to any buffer where diazoxide is added.
epinephrine (bitartrate salt) Sigma E4375 Stock solution: 5 mM in water
PMA (phorbol 12-myristate) Sigma P1585 Stock solution: 100 µM in DMSO
Name Company Catalog Number Comments
Guassia assay materials
Disodium phosphate (Na2HPO4) Thermo-Fisher Scientific S374-500
Glycerol Thermo-Fisher Scientific G334
Sodium Bromide Thermo-Fisher Scientific AC44680-1000
EDTA Thermo-Fisher Scientific AC44608-5000 Stock solution: 0.5 M pH 8
Tris base RPI T60040-1000.0 Stock solution: 1 M pH 8
Ascorbic Acid Fisher Scientific AAA1775922 US Patent US7718389 suggested ascorbate can increase coelenterazine stability.
Na2SO3 Sigma S0505-250G US Patent US8367357 suggested sulfite may decrease background due to BSA
Coelenterazine (native) Nanolight / Prolume 3035MG Stock solution: 1 mg/ml in acidified MeOH (2.36 mM)
OptiPlate-96, White Opaque 96-well Microplate Perkin Elmer 6005290 Any opaque white 96 well plate should be sufficient. Clear bottom plates will also work, however some signal will be lost.
Name Company Catalog Number Comments
Equipment
Synergy H1 Hybrid plate reader or equivalent BioTek 8041000 A plate reader with luminescence detection and 96-well plate capabilities is required.
8-channel VOYAGER Pipette (50-1250 µL) Integra 4724 An automated multichannel pipette is extremely useful for rapid addition of luciferase reagents and plating cells in 96 well format
8-channel 200 µL pipette Transferpette S 20-200 µL 2703710

DOWNLOAD MATERIALS LIST

References

  1. Kalwat, M. A., et al. Insulin promoter-driven Gaussia luciferase-based insulin secretion biosensor assay for discovery of beta-cell glucose-sensing pathways. ACS Sensors. 1 (10), 1208-1212 (2016).
  2. Burns, S. M., et al. High-throughput luminescent reporter of insulin secretion for discovering regulators of pancreatic Beta-cell function. Cell Metabolism. 21 (1), 126-137 (2015).
  3. Rajan, S., et al. In vitro processing and secretion of mutant insulin proteins that cause permanent neonatal diabetes. American Journal of Physiology - Endocrinology and Metabolism. 298 (3), E403-E410 (2010).
  4. Watkins, S., et al. Imaging Secretory Vesicles by Fluorescent Protein Insertion in Propeptide Rather Than Mature Secreted Peptide. Traffic. 3 (7), 461-471 (2002).
  5. Welsh, J. P., Patel, K. G., Manthiram, K., Swartz, J. R. Multiply mutated Gaussia luciferases provide prolonged and intense bioluminescence. Biochemical Biophysical Research Communications. 389 (4), 563-568 (2009).
  6. Tannous, B. A., Kim, D. E., Fernandez, J. L., Weissleder, R., Breakefield, X. O. Codon-optimized Gaussia luciferase cDNA for mammalian gene expression in culture and in vivo. Molecular Therarpy. 11 (3), 435-443 (2005).
  7. Compounds and methods for regulating insulin secretion. World patent. Burns, S. M., Wagner, B. K., Vetere, A. , WO2018175324A1 (2018).
  8. Kalwat, M. A., et al. Chromomycin A2 potently inhibits glucose-stimulated insulin secretion from pancreatic beta cells. Journal of General Physiology. , (2018).
  9. Luft, C., et al. Application of Gaussia luciferase in bicistronic and non-conventional secretion reporter constructs. BMC Biochemistry. 15 (1), 14 (2014).
  10. Ohmiya, Y., Wu, C. Stabilizing composition and stabilizing method of coelenterazine solution for high-throughput measurement of luciferase activity. U.S. Patent. , 7,718,389 B2 (2010).
  11. Tannous, B. A. Gaussia luciferase reporter assay for monitoring biological processes in culture and in vivo. Nature Protocols. 4 (4), 582-591 (2009).
  12. Wurdinger, T., et al. A secreted luciferase for ex vivo monitoring of in vivo processes. Nature Methods. 5 (2), 171-173 (2008).
  13. Henquin, J. C. Triggering and amplifying pathways of regulation of insulin secretion by glucose. Diabetes. 49 (11), 1751-1760 (2000).
  14. Mourad, N. I., Nenquin, M., Henquin, J. C. Amplification of insulin secretion by acetylcholine or phorbol ester is independent of beta-cell microfilaments and distinct from metabolic amplification. Molecular & Cellular Endocrinology. 367 (1-2), 11-20 (2013).
  15. Straub, S. G., Sharp, G. W. Evolving insights regarding mechanisms for the inhibition of insulin release by norepinephrine and heterotrimeric G proteins. American Journal of Physiology - Cell Physiology. 302 (12), C1687-C1698 (2012).
  16. Cheng, K., et al. High passage MIN6 cells have impaired insulin secretion with impaired glucose and lipid oxidation. PLoS One. 7 (7), e40868 (2012).
  17. Head, W. S., et al. Connexin-36 Gap Junctions Regulate In Vivo First- and Second-Phase Insulin Secretion Dynamics and Glucose Tolerance in the Conscious. Diabetes. 61 (7), 1700-1707 (2012).
  18. Benninger, R. K. P., Head, W. S., Zhang, M., Satin, L. S., Piston, D. W. Gap junctions and other mechanisms of cell-cell communication regulate basal insulin secretion in the pancreatic islet. The Journal of Physiology. 589 (22), 5453-5466 (2011).
  19. Konstantinova, I., et al. EphA-Ephrin-A-mediated beta cell communication regulates insulin secretion from pancreatic islets. Cell. 129 (2), 359-370 (2007).
  20. Jaques, F., et al. Dual effect of cell-cell contact disruption on cytosolic calcium and insulin secretion. Endocrinology. 149 (5), 2494-2505 (2008).
  21. Calabrese, A., et al. Connexin 36 Controls Synchronization of Ca2+ Oscillations and Insulin Secretion in MIN6 Cells. Diabetes. 52 (2), 417-424 (2003).
  22. Bielefeld-Sevigny, M. AlphaLISA immunoassay platform- the "no-wash" high-throughput alternative to ELISA. Assay and Drug Development Technologies. 7 (1), 90-92 (2009).
  23. Aslanoglou, D., George, E. W., Freyberg, Z. Homogeneous Time-resolved Forster Resonance Energy Transfer-based Assay for Detection of Insulin Secretion. Journal of Visualized Experiments. (135), (2018).
  24. Rafati, A., Zarrabi, A., Abediankenari, S., Aarabi, M., Gill, P. Sensitive colorimetric assay using insulin G-quadruplex aptamer arrays on DNA nanotubes coupled with magnetic nanoparticles. Royal Sociecy Open Science. 5 (3), 171835 (2018).
  25. Hulleman, J. D., Brown, S. J., Rosen, H., Kelly, J. W. A high-throughput cell-based Gaussia luciferase reporter assay for identifying modulators of fibulin-3 secretion. Journal of Biomolecular Screening. 18 (6), 647-658 (2013).
  26. Frank, J. A., et al. Optical tools for understanding the complexity of beta-cell signalling and insulin release. Nature Reviews Endocrinology. 14 (12), 721-737 (2018).
  27. Zhu, S., et al. Monitoring C-Peptide Storage and Secretion in Islet beta-Cells In Vitro and In Vivo. Diabetes. 65 (3), 699-709 (2016).

Tags

Measuring Relative Insulin Secretion Co-secreted Luciferase Surrogate Assay Beta Cell Lines Drug Discovery Characterization Multiple Treatments Immediate Results Affordable Small Molecule Modulators Diabetes Therapies Beta Cell Function Protein Secretion Neurotransmitter Secretion Lentiviral System Human Islets Cell Culture Multichannel Pipettes Glucose Response Insulin ELISA Krebs-Ringer Bicarbonate Buffer Gaussia Luciferase Working Solution MIN6 Cells Plating
Measuring Relative Insulin Secretion using a Co-Secreted Luciferase Surrogate
Play Video
PDF DOI DOWNLOAD MATERIALS LIST

Cite this Article

Kalwat, M., Cobb, M. H. MeasuringMore

Kalwat, M., Cobb, M. H. Measuring Relative Insulin Secretion using a Co-Secreted Luciferase Surrogate. J. Vis. Exp. (148), e59926, doi:10.3791/59926 (2019).

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