logo
AAT Bioquest

Cal-520 ® , Cal-590 ™, and Cal-630™ Calcium Detection Reagents

Introduction


Cal-520® , Cal-590™ and Cal-630™ provide the most robust homogeneous fluorescence-based assay tools for detecting intracellular calcium mobilization. They are fluorogenic calcium-sensitive dyes with a significantly improved signal to noise ratio and intracellular retention compared to the existing calcium indicators (such as Fluo-3 AM, Fluo-4 AM and Rhod-2 AM). Cells expressing a GPCR or calcium channel of interest that signals through calcium can be preloaded with Cal-520® AM, Cal-590™ AM or Cal-630™ AM which can cross cell membrane. Once inside the cell, the lipophilic blocking groups of Cal 520® AM, Cal-590™ AM or Cal-630™ AM are cleaved by intracellular esterases, resulting in a negatively charged fluorescent dye that stays inside cells. Their fluorescence is greatly enhanced upon binding to calcium. When cells stimulated with agonists, the receptor signals the release of intracellular calcium, which significantly increase the fluorescence of Cal-520® , Cal-590 ™ or Cal-630™. The characteristics of high sensitivity and >100 times fluorescence enhancement make Cal-520® AM, Cal-590™ AM or Cal-630™ AM ideal indicators for the measurement of cellular calcium. The high S/N ratio and better intracellular retention make the Cal-520® , Cal-590 ™ or Cal-630™ calcium assay a robust tool for evaluating GPCR and calcium channel targets as well as for screening their agonists and antagonists.

Besides their convenient excitation wavelengths and large fluorescence enhancement by calcium, Cal-520® , Cal590™, and Cal-630™ are predominantly localized in cytosols unlike Rhod-2 that is mainly localized in mitochondria. In addition, the long Ex/Em wavelengths of Cal-590™ and Cal-630™ make these dyes perfect calcium indicators compatible for multicolor detection with green fluorescent protein (GFP) cell lines. In addition, Cal-520® , Cal-590™ or Cal-630™ calcium assays are optimized to be compatible with most of the existing fluorescence instruments. Cal-520® can be well excited at 488 nm, and used with FITC filter set. Cal-590™ is optimized to be excited at 555 nm, and used with TRITC/Cy3 filter set. Cal-630™ is optimized to be excited at 594 nm, and used with Texas Red® filter set. The spectral and calcium binding properties are summarized below (see Table 1).

 

Table 1. Spectral and Ca2+ –Binding Properties of Cal-520®, Cal-590™ or Cal-630™ Ca2+ Detection Reagents

Ca2+ Indicator
Excitation (nm)
Emission (nm)
Kd (nM)
Cal-520®, AM492 nm514 nm320
Cal-590™ AM558 nm584 nm561
Cal-630™ AM607 nm623 nm792

 

Use of Cal-520® AM, Cal-590™ AM, or Cal-630™ AM Esters


  1. Load Cells with Cal-520®, Cal-590™ or Cal-630™ AM Esters: AM esters are non-polar esters that can readily cross live cell membranes, and are rapidly hydrolyzed by cellular esterases inside live cells. AM esters are widely used for loading a variety of polar fluorescent probes into live cells noninvasively. However, cautions must be exercised when AM esters are used since they are susceptible to hydrolysis, particularly in solution. They should be reconstituted just before use in high-quality, anhydrous dimethylsulfoxide (DMSO). DMSO stock solutions may be stored desiccated at –20°C and protected from light. Under these conditions, AM esters should be stable for several months. Following is our recommended protocol for loading Cal-520® AM, Cal-590™ AM or Cal-630™ AM esters into live cells. This protocol only provides a guideline, and should be modified according to your specific needs.
    1. Prepare a 2 to 5 mM stock solution of Cal-520® AM, Cal-590™ AM or Cal-630™ AM esters in high-quality, anhydrous DMSO.
    2. On the day of the experiment, either dissolve Cal-520® AM, Cal-590™ AM or Cal-630™ AM in DMSO or thaw an aliquot of the indicator stock solution to room temperature. Prepare a dye working solution of 10 to 20 µM in Hanks and Hepes buffer (HHBS) or the buffer of your choice with 0.04% Pluronic® F-127. The exact concentration of the indicator required for cell loading must be determined empirically.
      Note: The nonionic detergent Pluronic® F-127 is sometimes used to increase the aqueous solubility of Cal-520® AM, Cal-590™ AM or Cal-630™ AM esters. A variety of Pluronic® F-127 solutions can be purchased from AAT Bioquest.
    3. If your cells (such as CHO cells) contain organic anion-transports, than probenecid (1-2 mM) may be added to the dye working solution (final in well concentration will be 0.5-1 mM) to reduce leakage of the de-esterified indicators.
      Note: A variety of ReadiUse™ probenecid including water soluble sodium salt and stabilized solution can be purchased from AAT Bioquest.
    4. Add equal volume of the dye working solution (from Step b or c) into your cell plate.
    5. Incubate the dye-loading plate in a cell incubator for 60 to 90 minutes, and then incubate the plate at room temperature for another 30 minutes.
      Note: Incubating the dye longer than 2 hours gives better signal intensity for some cell lines.
    6. Replace the dye working solution with HHBS or if applicable, a buffer of your choice that contains an anion transporter inhibitor, such as 1 mM probenecid, to remove excess probes.
    7. Run the experiments at Ex/Em = 490/525 nm (for Cal-520® AM), 540/590 nm (for Cal-590™ AM) or 600/640 nm (for Cal-630™ AM).
  2. Measure Intracellular Calcium Responses:

Response of endogenous P2Y

Response of endogenous P2Y receptor to ATP in CHO-M1 cells without probenecid. CHO-M1 cells were seeded overnight at 40,000 cells per 100 µL per well in a 96-well black wall/clear bottom costar plate. 100 µl of 4 µM Fluo-3 AM, Fluo-4 AM or Cal 520® AM in HHBS were added into the wells, and the cells were incubated at 37°C for 2 hour. The dye loading medium were replaced with 100 µl HHBS, 50 µl of 300 µM ATP were added, and then imaged with a fluorescence microscope (Olympus IX71) using FITC channel.


ATP-stimulated calcium response

ATP-stimulated calcium response of endogenous P2Y receptor in CHO-K1 cells measured with Cal-520® or Fluo-4 AM. CHO-K1cells were seeded overnight in 50,000 cells per 100 µL per well in a 96-well black wall/clear bottom costar plate. 100 µL of 5 µM Fluo-4 AM or the Cal-520® AM with (A) or without (B) 2.5 mM probenecid was added into the cells, and the cells were incubated at 37oC for 2 hours. ATP (50 µL/well) was added by FlexStation (Molecular Devices) to achieve the final indicated concentrations.


Control
ATP

Response of endogenous P2Y receptor to ATP in CHO-K cells. CHO-K cells were seeded overnight at 40,000 cells per 100 µL per well in a 96-well black wall/clear bottom costar plate. 100 µl of 4 µM Cal 590 ™ AM or Cal 630™ AM in HHBS with 1 mM probenecid were added into the wells, and the cells were incubated at 37°C for 2 hour. The dye loading mediums were replaced with 100 µl HHBS and 1 mM probenecid , then imaged with a fluorescence microscope (Olympus IX71) using TRITC channel before and after adding 50 µl of 300 µM ATP.

 

Use of Cal-520® , Cal-590™, or Cal-630™ Salts


Calcium calibration can be carried out by measuring the fluorescence intensity of the salt form (25 to 50 µM in fluorescence microplate readers) of the indicators in solutions with precisely known free Ca2+ concentrations. Calibration solutions can be used based on 30 mM MOPS EGTA Ca2+ buffer. In general, water contains trace amount of calcium ion. It is highly recommended to use 30 mM MOPS + 100 mM KCl, pH 7.2 as buffer system. One can simply make a 0 and 39 µM calcium stock solutions as listed below, and these 2 solutions are used to make a serial solution of different Ca2+ concentrations. To determine either the free calcium concentration of a solution or the Kd of a single-wavelength calcium indicator, the following equation is used:
  1. 0 µM calcium: 30 mM MOPS + 100 mM KCl, pH 7.2 buffer + 10 mM EGTA
  2. 39 µM calcium: 30 mM MOPS + 100 mM KCl, pH 7.2 buffer + 10 mM EGTA + 10 mM CaCl2
To determine either the free calcium concentration of a solution or the Kd of a single-wavelength calcium indicator, the following equation is used:

[Ca]free = Kd[F · Fmin]/[Fmax · F]


Where F is the fluorescence intensity of the indicator at a specific experimental calcium level, Fmin is the fluorescence intensity in the absence of calcium and Fmax is the fluorescence intensity of the calciumsaturated probe.

The dissociation constant (Kd) is a measure of the affinity of the probe for calcium. The calcium-binding and spectroscopic properties of fluorescent indicators vary quite significantly in cellular environments compared to calibration solutions. In situ response calibrations of intracellular indicators typically yield Kd values significantly higher than in vitro determinations. In situ calibrations are performed by exposing loaded cells to controlled Ca2+ buffers in the presence of ionophores such as A-23187, 4-bromo A-23187 and ionomycin. Alternatively, cell permeabilization agents such as digitonin or Triton® X-100 can be used to expose the indicator to the controlled Ca2+ levels of the extracellular medium.

 

Use of Cal-520® , Cal-590™, or Cal-630™ Dextran Conjugate


Compared to the free ion indicator, dextran conjugates of these same indicators exhibit both reduced compartmentalization and much lower rates of dye leakage. Since the molecular weight of the dextran, net charge, degree of labeling, and nature of the dye may affect the experiment, researchers are advised to consult the primary literature for information specific to the application of interest.

 

Conclusions


Because of the importance of Ca2+ in biology, numerous techniques/methods for analyzing the mechanisms of cellular and/or subcellular Ca2+ activity have been established. Although each method for analyzing Ca2+ activity has certain advantages over the others, each also suffers from drawbacks. With the outstanding properties described above, we believe that Cal-520® , Cal-590™ and Cal-630™ calcium detection reagents provide a new powerful tool for intracellular calcium analysis and monitor in a variety of biological systems.

As might have been predicted, the interests of many researchers in Ca2+ analysis shifted from the cellular level to the subcellular level. It has been found that Ca2+ is not even distributed throughout the whole cell and that intracellular heterogeneity of Ca2+ (such as Ca2+ waves and Ca2+ sparks) is observed in a variety of cells (e.g., oocyte, heart muscle cell, hepatocyte, and exocrine cell). With the advent of the confocal laser scanning microscope (CLSM) in the 1980s and advanced microplate readers in 2000s (such as FLIPR, FDSS and NOVOStar dedicated for intracellular Ca2+ detections), the measurement of intracellular Ca2+ has accelerated significantly. Confocal laser scanning microscopy and more recently multiphoton microscopy allow the precise spatial and temporal analysis of intracellular Ca2+ signaling at the subcellular level in addition to the measurement of its concentration.

 

Product Ordering Information


 

Table 2. Ordering Information For Cal-520®, Cal-590™, and Cal-630™

Cat No.
Product Name
Unit Size
20605Cal-520®-Biotin Conjugate5x50 µg
20606Cal-520®-Biocytin Conjugate5x50 µg
20609Cal-520® NHS Ester100 µg
20610Cal-520® maleimide100 µg
21130Cal-520®, AM10x50 µg
21131Cal-520®, AM1 mg
21135Cal-520®, sodium salt10x50 µg
21136Cal-520®, sodium salt1 mg
21140Cal-520®, potassium salt10x50 μg
21141Cal-520®, potassium salt1 mg
21142Cal-520FF™, AM1 mg
21143Cal-520FF™, AM10x50 μg
21144Cal-520FF™, potassium salt10x50 μg
20600Cal-520®-Dextran Conjugate *MW 3,000*1 mg
20601Cal-520®-Dextran Conjugate *MW 10,000*5 mg
20508Cal-590™-Dextran Conjugate *MW 3,000*1 mg
20509Cal-590™-Dextran Conjugate *MW 10,000*1 mg
20510Cal-590™ AM5x50 μg
20511Cal-590™ AM10x50 μg
20512Cal-590™ AM1 mg
20530Cal-630™ AM5x50 μg
20515Cal-590™, sodium salt5x50 μg
20518Cal-590™, potassium salt5x50 μg
20531Cal-630™ AM10x50 μg
20532Cal-630™ AM1 mg
20535Cal-630™, sodium salt5x50 μg
20538Cal-630™, potassium salt5x50 μg
20546Cal-630™-Dextran Conjugate *MW 10,000*1 mg
20588Cal Red™ R525/650 potassium salt5x50 μg
20590Cal Red™ R525/650 AM1 mg
20591Cal Red™ R525/650 AM10x50 ug

 

References


  1. J.T. Lock, I. Parker, I.F. Smith, A comparison of fluorescent Ca2+ indicators for imaging local Ca2+ signals in cultured cells, Cell Calcium (2015) October, http://dx.doi.org/10.1016/j.ceca.2015.10.003
  2. Carsten Tischbirek, Antje Birkner, Hongbo Jia, Bert Sakmann, and Arthur Konnerth. Deep two-photon brain imaging with a red-shifted fluorometric Ca2+ indicator. PNAS. 2015; 112:11377-11382. doi: 10.1073/pnas.1514209112
  3. Søren Grubb, Gary L. Aistrup, Jussi T. Koivumäki, Tobias Speerschneider, Lisa A. Gottlieb, Nancy A. M. Mutsaers, Søren-Peter Olesen, Kirstine Calloe, Morten B.Thomsen . Preservation of cardiac function by prolonged action potentials in mice deficient of KChIP2 American Journal of Physiology - Heart and Circulatory Physiology Published 1 August 2015 Vol. 309 no. 3, H481-H489 DOI: 10.1152/ajpheart.00166.2015
  4. Emery Smith, Peter Chase, Colleen M. Niswender, Thomas J. Utley, Douglas J. Sheffler, Meredith J. Noetzel, Atin Lamsal, Michael R. Wood, P. Jeffrey Conn, Craig W. Lindsley, Franck Madoux, Mary Acosta, Louis Scampavia, Timothy Spicer, and Peter Hodder. Application of Parallel Multiparametric Cell-Based FLIPR Detection Assays for the Identification of Modulators of the Muscarinic Acetylcholine Receptor 4 (M4). J Biomol Screen. 2015; 20:858- 868. doi:10.1177/1087057115581770.
  5. Wenxiang Hu, Binlong Qiu, Wuqiang Guan, Qinying Wang, Min Wang, Wei Li, Longfei Gao, Lu Shen,Yin Huang, Gangcai Xie, Hanzhi Zhao, Ying Jin, Beisha Tang, Yongchun Yu, Jian Zhao, and Gang Pei Direct Conversion of Normal and Alzheimer's Disease Human Fibroblasts into Neuronal Cells by Small Molecules. Cell Stem Cell 17, 204–212, August 6, 2015. http://dx.doi.org/10.1016/j.stem.2015.07.006
  6. Carsten Tischbirek, Antje Birkner, Hongbo Jia, Bert Sakmann, and Arthur Konnerth. Deep two-photon brain imaging with a red-shifted fluorometric Ca2+ indicator. PNAS. 2015; 112:11377-11382. doi: 10.1073/pnas.1514209112
  7. Songqing Tang, Taoyong Chen, Mingjin Yang, Lei Wang, Zhou Yu, Bin Xie,Cheng Qian, Sheng Xu, Nan Li, Xuetao Cao and Jianli Wang. Extracellular calcium elicits feedforward regulation of the Toll-like receptor-triggered innate immune response. Cellular & Molecular Immunology, (17 August 2015) | doi:10.1038/cmi.2015.59.
  8. Mayumi Tada, Atsuya Takeuchi, Miki Hashizume, Kazuo Kitamura, Masanobu Kano.Article. A highly sensitive fluorescent indicator dye for calcium imaging of neural activity in vitro and in vivo. European Journal of Neuroscience 9 JAN 2014. DOI: 10.1111/ejn.12476.
  9. Daisuke Kodama, Akifumi Togari. Store-operated calcium entry induced by activation of Gq-coupled alpha1B adrenergic receptor in human osteoblast Biochemical and Biophysical Research Communications June (2013) doi: 10.1016/j.bbrc.2013.06.047.
  10. Rie Yamamoto, Shigeharu Ueki, Yuki Moritoki, Yoshiki Kobayashi, Hajime Oyamada, Yasunori Konno, Mami Tamaki, Masamichi Itoga, Masahide Takeda, Wataru Ito, and Junichi Chihara. Adiponectin attenuates human eosinophil adhesion and chemotaxis: implications in allergic inflammation. Journal of Asthma 2013. Posted online on July 17, 2013. (doi:10.3109/02770903.2013.816725).
  11. Carsten Tischbirek, Antje Birkner, Hongbo Jia, Bert Sakmann, and Arthur Konnerth. Deep two-photon brain imaging with a red-shifted fluorometric Ca2+ indicator. PNAS. 2015; 112:11377-11382. doi: 10.1073/pnas.1514209112


Original created on November 25, 2019, last updated on November 25, 2019
Tagged under: Calcium GPCR Analysis, Calcium Imaging, Calcium Mobilization