Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Creating new fluorescent probes for cell biology

An Erratum to this article was published on 01 January 2003

Key Points

  • Advances in fluorescence microscopy and the engi-neering of the green fluorescent protein (GFP) from Aequorea victoria into mutants with improved properties and altered colours have provided the basic tools that allow the investigation of complex processes in live cells. Fluorescent protein-based indicators can be designed to respond to various biological events and signals, targeted to subcellular compartments and introduced into various tissues and intact organisms.

  • Recent advances in fluorescent proteins include the engineering or discovery of variants that have enhanced brightness, improved pH resistance, the ability to undergo photochemical colour conversion, or red fluorescent emission.

  • Among the alternatives to fluorescent proteins, the tetracysteine–biarsenical labelling system is one of the most promising owing to its minimal steric bulk, fast rate of labelling, the availability of several colours and its ability to provide staining that is suitable for electron microscopy.

  • The barrel-like structures of Aequorea fluorescent proteins (AFPs), which effectively shield the chromophore from the external environment, make them well suited to more 'passive' applications such as monitoring the spatio-temporal (location, translocation, accumulation and degradation) dynamics of appropriate fusion proteins.

  • In the more 'active' applications of fluorescent proteins, biochemical parameters such as metabolite concentration, enzyme activity, or protein–protein interactions can be detected by their effects on the fluorescence properties of the designed indicators. Such indicators can be divided further into molecules with single chromophores versus composites that are based on fluorescence resonance energy transfer (FRET) between two chromophores.

  • Single-fluorophore fluorescent protein-based indicators have been engineered to respond to various cellular parameters including pH, halides, free Ca2+ and redox potentials.

  • Intramolecular FRET-based indicators have been engineered to monitor intracellular Ca2+, cyclic GMP, GTPase and kinase activities. Protein–protein interactions, cyclic AMP dynamics and clustering in lipid rafts have been analysed by using intermolecular FRET-based probes.

  • The applications of fluorescent probes will continue to expand and provide exciting new insights into the biology of living cells. Particularly exciting areas include high-throughput screening, single-molecule spectroscopy and whole-body in vivo imaging.

Abstract

Fluorescent probes are one of the cornerstones of real-time imaging of live cells and a powerful tool for cell biologists. They provide high sensitivity and great versatility while minimally perturbing the cell under investigation. Genetically-encoded reporter constructs that are derived from fluorescent proteins are leading a revolution in the real-time visualization and tracking of various cellular events. Recent advances include the continued development of 'passive' markers for the measurement of biomolecule expression and localization in live cells, and 'active' indicators for monitoring more complex cellular processes such as small-molecule-messenger dynamics, enzyme activation and protein–protein interactions.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Recent advances in fluorescent proteins.
Figure 2: The biarsenical–tetracysteine system.
Figure 3: Multicolour pulse–chase biarsenical staining of gap juctions.
Figure 4: Biochemical modulation of AFP fluorescence.
Figure 5: The general design of FRET-based fluorescent probes.

Similar content being viewed by others

References

  1. Tsien, R. Y. The green fluorescent protein. Annu. Rev. Biochem. 67, 509–544 (1998).

    Article  CAS  PubMed  Google Scholar 

  2. Sawano, A. & Miyawaki, A. Directed evolution of green fluorescent protein by a new versatile PCR strategy for site-directed and semi-random mutagenesis. Nucleic Acids Res. 28, E78 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Griesbeck, O., Baird, G. S., Campbell, R. E., Zacharias, D. A. & Tsien, R. Y. Reducing the environmental sensitivity of yellow fluorescent protein. Mechanism and applications. J. Biol. Chem. 276, 29188–29194 (2001).

    CAS  PubMed  Google Scholar 

  4. Nagai, T. et al. A variant of yellow fluorescent protein with fast and efficient maturation for cell-biological applications. Nature Biotechnol. 20, 87–90 (2002).

    CAS  Google Scholar 

  5. Scholz, O., Thiel, A., Hillen, W. & Niederweis, M. Quantitative analysis of gene expression with an improved green fluorescent protein. Eur. J. Biochem. 267, 1565–1570 (2000).

    CAS  PubMed  Google Scholar 

  6. Ormö, M. et al. Crystal structure of the Aequorea victoria green fluorescent protein. Science 273, 1392–1395 (1996).

    PubMed  Google Scholar 

  7. Yokoe, H. & Meyer, T. Spatial dynamics of GFP-tagged proteins investigated by local fluorescence enhancement. Nature Biotechnol. 14, 1252–1256 (1996).

    CAS  Google Scholar 

  8. Sawin, K. E. & Nurse, P. Photoactivation of green fluorescent protein. Curr. Biol. 7, R606–R607 (1997).

    CAS  PubMed  Google Scholar 

  9. Elowitz, M. B., Surette, M. G., Wolf, P. E., Stock, J. & Leibler, S. Photoactivation turns green fluorescent protein red. Curr. Biol. 7, 809–812 (1997).

    CAS  PubMed  Google Scholar 

  10. Patterson, G. H. & Lippincott-Schwartz, J. A photoactivatable GFP for selective photolabeling of proteins and cells. Science 297, 1873–1877 (2002).

    CAS  PubMed  Google Scholar 

  11. Ando, R., Hama, H., Yamamoto-Hino, M., Mizuno, H. & Miyawaki, A. An optical marker based on the UV-induced green-to-red photoconversion of a fluorescent protein. Proc. Natl Acad. Sci. USA 99, 12651–12656 (2002). References 10 and 11 describe a pair of photoactivatable fluorescent proteins that can be used as fluorescent 'highlighters' to mark specific organelles or protein subpopulations.

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Lippincott-Schwartz, J., Snapp, E. & Kenworthy, A. Studying protein dynamics in living cells. Nature Rev. Mol. Cell Biol. 2, 444–456 (2001).

    CAS  Google Scholar 

  13. Zacharias, D. A., Violin, J. D., Newton, A. C. & Tsien, R. Y. Partitioning of lipid-modified monomeric GFPs into membrane microdomains of live cells. Science 296, 913–916 (2002).

    CAS  PubMed  Google Scholar 

  14. Ward, W. W., Prentice, H. J., Roth, A. F., Cody, C. W. & Reeves, S. C. Spectral perturbations of the Aequorea green-fluorescent protein. Photochem. Photobiol. 35, 803–808 (1982).

    CAS  Google Scholar 

  15. Yang, F., Moss, L. G. & Phillips, G. N. Jr. The molecular structure of green fluorescent protein. Nature Biotechnol. 14, 1246–1251 (1996).

    CAS  Google Scholar 

  16. Matz, M. V. et al. Fluorescent proteins from nonbioluminescent Anthozoa species. Nature Biotechnol. 17, 969–973 (1999).

    CAS  Google Scholar 

  17. Tsien, R. Y. Rosy dawn for fluorescent proteins. Nature Biotechnol. 17, 956–957 (1999).

    CAS  Google Scholar 

  18. Zacharias, D. A. Sticky caveats in an otherwise glowing report: oligomerizing fluorescent proteins and their use in cell biology. Sci. STKE. 2002, PE23 (2002).

    PubMed  Google Scholar 

  19. Lauf, U., Lopez, P. & Falk, M. M. Expression of fluorescently tagged connexins: a novel approach to rescue function of oligomeric DsRed-tagged proteins. FEBS Lett. 498, 11–15 (2001).

    CAS  PubMed  Google Scholar 

  20. Soling, A., Simm, A. & Rainov, N. Intracellular localization of Herpes simplex virus type 1 thymidine kinase fused to different fluorescent proteins depends on choice of fluorescent tag. FEBS Lett. 527, 153 (2002).

    CAS  PubMed  Google Scholar 

  21. Yanushevich, Y. G. et al. A strategy for the generation of non-aggregating mutants of Anthozoa fluorescent proteins. FEBS Lett. 511, 11–14 (2002).

    CAS  PubMed  Google Scholar 

  22. Terskikh, A. V., Fradkov, A. F., Zaraisky, A. G., Kajava, A. V. & Angres, B. Analysis of DsRed mutants: space around the fluorophore accelerates fluorescence development. J. Biol. Chem. 277, 7633–7636 (2002).

    CAS  PubMed  Google Scholar 

  23. Bevis, B. J. & Glick, B. S. Rapidly maturing variants of the Discosoma red fluorescent protein (DsRed). Nature Biotechnol. 20, 83–87 (2002).

    CAS  Google Scholar 

  24. Campbell, R. E. et al. A monomeric red fluorescent protein. Proc. Natl Acad. Sci. USA 99, 7877–7882 (2002). This paper describes the directed evolution of a monomeric RFP that overcomes the problems associated with tetrameric DsRed.

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Knop, M., Barr, F., Riedel, C. G., Heckel, T. & Reichel, C. Improved version of the red fluorescent protein (drFP583/DsRed/RFP). BioTechniques 33, 592, 594, 596–598 (2002).

    Google Scholar 

  26. CLONTECH Laboratories Inc. Living Colors User Manual Vol. II: Red Fluorescent Protein 4 (Becton, Dickinson and Company, USA, 2002).

  27. Fradkov, A. F. et al. Far-red fluorescent tag for protein labeling. Biochem. J. 368, 17–21 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Labas, Y. A. et al. Diversity and evolution of the green fluorescent protein family. Proc. Natl Acad. Sci. USA 99, 4256–4261 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Matz, M. V., Lukyanov, K. A. & Lukyanov, S. A. Family of the green fluorescent protein: journey to the end of the rainbow. Bioessays 24, 953–959 (2002).

    CAS  PubMed  Google Scholar 

  30. Peelle, B., Gururaja, T. L., Payan, D. G. & Anderson, D. C. Characterization and use of green fluorescent proteins from Renilla mulleri and Ptilosarcus guernyi for the human cell display of functional peptides. J. Protein Chem. 20, 507–519 (2001).

    CAS  PubMed  Google Scholar 

  31. Lukyanov, K. A. et al. Natural animal coloration can be determined by a non-fluorescent GFP homolog. J. Biol. Chem. 275, 25879–25882 (2000).

    CAS  PubMed  Google Scholar 

  32. Fradkov, A. F. et al. Novel fluorescent protein from Discosoma coral and its mutants possesses a unique far-red fluorescence. FEBS Lett. 479, 127–130 (2000).

    CAS  PubMed  Google Scholar 

  33. Gurskaya, N. G. et al. GFP-like chromoproteins as a source of far-red fluorescent proteins. FEBS Lett. 507, 16–20 (2001).

    CAS  PubMed  Google Scholar 

  34. Wiedenmann, J. et al. A far-red fluorescent protein with fast maturation and reduced oligomerization tendency from Entacmaea quadricolor (Anthozoa, Actinaria). Proc. Natl Acad. Sci. USA 99, 11646–11651 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Griffin, B. A., Adams, S. R. & Tsien, R. Y. Specific covalent labeling of recombinant protein molecules inside live cells. Science 281, 269–272 (1998).

    CAS  PubMed  Google Scholar 

  36. Nakanishi, J. et al. Imaging of conformational changes of proteins with a new environment- sensitive fluorescent probe designed for site-specific labeling of recombinant proteins in live cells. Anal. Chem. 73, 2920–2928 (2001).

    CAS  PubMed  Google Scholar 

  37. Adams, S. R. et al. New biarsenical ligands and tetracysteine motifs for protein labeling in vitro and in vivo: synthesis and biological applications. J. Am. Chem. Soc. 124, 6063–6076 (2002).

    CAS  PubMed  Google Scholar 

  38. Gaietta, G. et al. Multicolor and electron microscopic imaging of connexin trafficking. Science 296, 503–507 (2002). This paper describes the application of the tetracysteine–biarsenical system in pulse–chase labelling and electron microscopy.

    CAS  PubMed  Google Scholar 

  39. Lauf, U. et al. Dynamic trafficking and delivery of connexons to the plasma membrane and accretion to gap junctions in living cells. Proc. Natl Acad. Sci. USA 99, 10446–10451 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Farinas, J. & Verkman, A. S. Receptor-mediated targeting of fluorescent probes in living cells. J. Biol. Chem. 274, 7603–7606 (1999).

    CAS  PubMed  Google Scholar 

  41. Wu, M. M. et al. Organelle pH studies using targeted avidin and fluorescein-biotin. Chem. Biol. 7, 197–209 (2000).

    CAS  PubMed  Google Scholar 

  42. Wu, M. M. et al. Studying organelle physiology using targeted avidin and fluorescein-biotin. Methods Enzymol. 327, 546–564 (2000).

    CAS  PubMed  Google Scholar 

  43. Wu, M. M. et al. Mechanisms of pH regulation in the regulated secretory pathway. J. Biol. Chem. 276, 33027–33035 (2001).

    CAS  PubMed  Google Scholar 

  44. Sano, T. & Cantor, C. R. Expression of a cloned streptavidin gene in Escherichia coli. Proc. Natl Acad. Sci. USA 87, 142–146 (1990).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Gambetta, G. A. & Lagarias, J. C. Genetic engineering of phytochrome biosynthesis in bacteria. Proc. Natl Acad. Sci. USA 98, 10566–10571 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Tooley, A. J., Cai, Y. A. & Glazer, A. N. Biosynthesis of a fluorescent cyanobacterial C-phycocyanin holo-α subunit in a heterologous host. Proc. Natl Acad. Sci. USA 98, 10560–10565 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Tooley, A. J. & Glazer, A. N. Biosynthesis of the cyanobacterial light-harvesting polypeptide phycoerythrocyanin holo-α subunit in a heterologous host. J. Bacteriol. 184, 4666–4671 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Murphy, J. T. & Lagarias, J. C. The phytofluors: a new class of fluorescent protein probes. Curr. Biol. 7, 870–876 (1997).

    CAS  PubMed  Google Scholar 

  49. Wildt, S. & Deuschle, U. cobA, a red fluorescent transcriptional reporter for Escherichia coli, yeast, and mammalian cells. Nature Biotechnol. 17, 1175–1178 (1999).

    CAS  Google Scholar 

  50. Cubitt, A. B. et al. Understanding, using and improving green fluorescent protein. Trends Biochem. Sci. 20, 448–455 (1995).

    CAS  PubMed  Google Scholar 

  51. Gonzalez, C. & Bejarano, L. A. Protein traps: using intracellular localization for cloning. Trends Cell Biol. 10, 162–165 (2000).

    CAS  PubMed  Google Scholar 

  52. Waterman-Storer, C. M., Desai, A., Bulinski, J. C. & Salmon, E. D. Fluorescent speckle microscopy, a method to visualize the dynamics of protein assemblies in living cells. Curr. Biol. 8, 1227–1230 (1998).

    CAS  PubMed  Google Scholar 

  53. Watanabe, N. & Mitchison, T. J. Single-molecule speckle analysis of actin filament turnover in lamellipodia. Science 295, 1083–1086 (2002).

    CAS  PubMed  Google Scholar 

  54. Bulinski, J. C., Odde, D. J., Howell, B. J., Salmon, T. D. & Waterman-Storer, C. M. Rapid dynamics of the microtubule binding of ensconsin in vivo. J. Cell Sci. 114, 3885–3897 (2001).

    CAS  PubMed  Google Scholar 

  55. Watton, S. J. & Downward, J. Akt/PKB localisation and 3′ phosphoinositide generation at sites of epithelial cell–matrix and cell–cell interaction. Curr. Biol. 9, 433–436 (1999).

    CAS  PubMed  Google Scholar 

  56. Oatey, P. B. et al. Confocal imaging of the subcellular distribution of phosphatidylinositol 3,4,5-trisphosphate in insulin- and PDGF-stimulated 3T3-L1 adipocytes. Biochem. J. 344, 511–518 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Gray, A., Van Der, K. J. & Downes, C. P. The pleckstrin homology domains of protein kinase B and GRP1 (general receptor for phosphoinositides-1) are sensitive and selective probes for the cellular detection of phosphatidylinositol 3,4-bisphosphate and/or phosphatidylinositol 3,4,5-trisphosphate in vivo. Biochem. J. 344, 929–936 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Servant, G. et al. Polarization of chemoattractant receptor signaling during neutrophil chemotaxis. Science 287, 1037–1040 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Meili, R. et al. Chemoattractant-mediated transient activation and membrane localization of Akt/PKB is required for efficient chemotaxis to cAMP in Dictyostelium. EMBO J. 18, 2092–2105 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Stauffer, T. P., Ahn, S. & Meyer, T. Receptor-induced transient reduction in plasma membrane PtdIns(4,5)P2 concentration monitored in living cells. Curr. Biol. 8, 343–346 (1998).

    CAS  PubMed  Google Scholar 

  61. Hirose, K., Kadowaki, S., Tanabe, M., Takeshima, H. & Lino, M. Spatiotemporal dynamics of inositol 1,4,5-trisphosphate that underlies complex Ca2+ mobilization patterns. Science 284, 1527–1530 (1999).

    CAS  PubMed  Google Scholar 

  62. Oancea, E., Teruel, M. N., Quest, A. F. G. & Meyer, T. Green fluorescent protein (GFP)-tagged cysteine-rich domains from protein kinase C as fluorescent indicators for diacylglycerol signaling in living cells. J. Cell Biol. 140, 485–498 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Teruel, M. N. & Meyer, T. Parallel single-cell monitoring of receptor-triggered membrane translocation of a calcium-sensing protein module. Science 295, 1910–1912 (2002).

    CAS  PubMed  Google Scholar 

  64. Oancea, E. & Meyer, T. Protein kinase C as a molecular machine for decoding calcium and diacylglycerol signals. Cell 95, 307–318 (1998). Reference 64 compares the kinetics of translocation of full-length PKC versus individual DAG-binding C1 domain and Ca2+-binding C2 domain, which led to a model for a sequential activation of PKC through a temporal coordination between Ca2+ and DAG signals.

    CAS  PubMed  Google Scholar 

  65. Rizzo, M. A., Shome, K., Watkins, S. C. & Romero, G. The recruitment of Raf-1 to membranes is mediated by direct interaction with phosphatidic acid and is independent of association with Ras. J. Biol. Chem. 275, 23911–23918 (2000).

    CAS  PubMed  Google Scholar 

  66. Dirks, R. W., Molenaar, C. & Tanke, H. J. Methods for visualizing RNA processing and transport pathways in living cells. Histochem. Cell Biol. 115, 3–11 (2001).

    CAS  PubMed  Google Scholar 

  67. Bassell, G. J., Oleynikov, Y. & Singer, R. H. The travels of mRNAs through all cells large and small. FASEB J. 13, 447–454 (1999).

    CAS  PubMed  Google Scholar 

  68. Bertrand, E. et al. Localization of ASH1 mRNA particles in living yeast. Mol. Cell 2, 437–445 (1998).

    CAS  PubMed  Google Scholar 

  69. Theurkauf, W. E. & Hazelrigg, T. I. In vivo analyses of cytoplasmic transport and cytoskeletal organization during Drosophila oogenesis: characterization of a multi-step anterior localization pathway. Development 125, 3655–3666 (1998).

    CAS  PubMed  Google Scholar 

  70. Zhang, H. L. et al. Neurotrophin-induced transport of a β-actin mRNP complex increases β-actin levels and stimulates growth cone motility. Neuron 31, 261–275 (2001).

    CAS  PubMed  Google Scholar 

  71. Muller, W. G., Walker, D., Hager, G. L. & McNally, J. G. Large-scale chromatin decondensation and recondensation regulated by transcription from a natural promoter. J. Cell Biol. 154, 33–48 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Tsukamoto, T. et al. Visualization of gene activity in living cells. Nature Cell Biol. 2, 871–878 (2000).

    CAS  PubMed  Google Scholar 

  73. Straight, A. F., Marshall, W. F., Sedat, J. W. & Murray, A. W. Mitosis in living budding yeast: anaphase A but no metaphase plate. Science 277, 574–578 (1997).

    CAS  PubMed  Google Scholar 

  74. Robinett, C. C. et al. In vivo localization of DNA sequences and visualization of large-scale chromatin organization using lac operator/repressor recognition. J. Cell Biol. 135, 1685–1700 (1996).

    CAS  PubMed  Google Scholar 

  75. Hadjantonakis, A. K. & Nagy, A. The color of mice: in the light of GFP-variant reporters. Histochem. Cell Biol. 115, 49–58 (2001).

    CAS  PubMed  Google Scholar 

  76. Zlokarnik, G. et al. Quantitation of transcription and clonal selection of single living cells using β-lactamase as reporter. Science 279, 84–88 (1998).

    CAS  PubMed  Google Scholar 

  77. Li, X. et al. Generation of destabilized green fluorescent protein as a transcription reporter. J. Biol. Chem. 273, 34970–34975 (1998).

    CAS  PubMed  Google Scholar 

  78. Terskikh, A. et al. 'Fluorescent timer': protein that changes color with time. Science 290, 1585–1588 (2000). This paper describes a DsRed-derived transcriptional reporter that changes colour from green to red over a period of 24 h, thereby preserving the temporal history of promoter activation.

    CAS  PubMed  Google Scholar 

  79. Dantuma, N. P., Lindsten, K., Glas, R., Jellne, M. & Masucci, M. G. Short-lived green fluorescent proteins for quantifying ubiquitin/proteasome-dependent proteolysis in living cells. Nature Biotechnol. 18, 538–543 (2000).

    CAS  Google Scholar 

  80. Kaether, C. & Gerdes, H. H. Visualization of protein transport along the secretory pathway using green fluorescent protein. FEBS Lett. 369, 267–271 (1995).

    CAS  PubMed  Google Scholar 

  81. Rudolf, R., Salm, T., Rustom, A. & Gerdes, H. H. Dynamics of immature secretory granules: role of cytoskeletal elements during transport, cortical restriction, and F-actin-dependent tethering. Mol. Biol. Cell 12, 1353–1365 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Kneen, M., Farinas, J., Li, Y. & Verkman, A. S. Green fluorescent protein as a noninvasive intracellular pH indicator. Biophys. J. 74, 1591–1599 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Llopis, J., McCaffery, J. M., Miyawaki, A., Farquhar, M. G. & Tsien, R. Y. Measurement of cytosolic, mitochondrial and Golgi pH in single living cells with green fluorescent protein. Proc. Natl Acad. Sci. USA 95, 6803–6808 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Miesenböck, G., De Angelis, D. A. & Rothman, J. E. Visualizing secretion and synaptic transmission with pH-sensitive green fluorescent proteins. Nature 394, 192–195 (1998). Reference 84 describes 'synapto-pHluorins' that report synaptic neurotransmitter secretion by detecting the abrupt pH change that occurs when the acidic interior of the vesicle (pH 5) is exposed to the outside of the cell (pH 7) on fusion to the plasma membrane.

    PubMed  Google Scholar 

  85. Matsuyama, S., Llopis, J., Deveraux, Q. L., Tsien, R. Y. & Reed, J. C. Changes in intramitochondrial and cytosolic pH: early events that modulate caspase activation during apoptosis. Nature Cell Biol. 2, 318–325 (2000).

    CAS  PubMed  Google Scholar 

  86. Wachter, R. M. & Remington, S. J. Sensitivity of the yellow variant of green fluorescent protein to halides and nitrate. Curr. Biol. 9, R628–R629 (1999).

    CAS  PubMed  Google Scholar 

  87. Wachter, R. M., Yarbrough, D., Kallio, K. & Remington, S. J. Crystallographic and energetic analysis of binding of selected anions to the yellow variants of green fluorescent protein. J. Mol. Biol. 301, 157–171 (2000).

    CAS  PubMed  Google Scholar 

  88. Jayaraman, S., Haggie, P., Wachter, R. M., Remington, S. J. & Verkman, A. S. Mechanism and cellular applications of a green fluorescent protein-based halide sensor. J. Biol. Chem. 275, 6047–6050 (2000).

    CAS  PubMed  Google Scholar 

  89. Elsliger, M. -A., Wachter, R. M., Hanson, G. T., Kallio, K. & Remington, S. J. Structural and spectral response of green fluorescent protein variants to changes in pH. Biochemistry 38, 5296–5301 (1999).

    CAS  PubMed  Google Scholar 

  90. Kuner, T. & Augustine, G. J. A genetically encoded ratiometric neurotechnique indicator for chloride: capturing chloride transients in cultured hippocampal neurons. Neuron 27, 447–459 (2000).

    CAS  PubMed  Google Scholar 

  91. Barondeau, D. P., Kassmann, C. J., Tainer, J. A. & Getzoff, E. D. Structural chemistry of a green fluorescent protein Zn biosensor. J. Am. Chem. Soc. 124, 3522–3524 (2002).

    CAS  PubMed  Google Scholar 

  92. Ostergaard, H., Henriksen, A., Hansen, F. G. & Winther, J. R. Shedding light on disulfide bond formation: engineering a redox switch in green fluorescent protein. EMBO J. 20, 5853–5862 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Baird, G. S., Zacharias, D. A. & Tsien, R. Y. Circular permutation and receptor insertion within green fluorescent proteins. Proc. Natl Acad. Sci. USA 96, 11241–11246 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Rapizzi, E. et al. Recombinant expression of the voltage-dependent anion channel enhances the transfer of Ca2+ microdomains to mitochondria. J. Cell Biol. (in the press).

  95. Yu, D., Baird, G., Tsien, R. Y. & Davis, R. L. Detection of calcium transient in Drosophila mushroom body neurons with camgaroo. J. Neurosci. (in the press).

  96. Siegel, M. S. & Isacoff, E. Y. A genetically encoded optical probe of membrane voltage. Neuron 19, 735–741 (1997).

    CAS  PubMed  Google Scholar 

  97. Ataka, K. & Pieribone, V. A. A genetically targetable fluorescent probe of channel gating with rapid kinetics. Biophys. J. 82, 509–516 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Nakai, J., Ohkura, M. & Imoto, K. A high signal-to-noise Ca2+ probe composed of a single green fluorescent protein. Nature Biotechnol. 19, 137–141 (2001).

    CAS  Google Scholar 

  99. Nagai, T., Sawano, A., Park, E. & Miyawaki, A. Circularly permuted green fluorescent proteins engineered to sense Ca2+. Proc. Natl Acad. Sci. USA 98, 3197–3202 (2001). References 98 and 99 describe the insertion of circularly permuted GFP between calmodulin and M13 to yield Ca2+ indicators that have been dubbed 'GCaMP' and 'pericams', respectively.

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Heim, R. & Tsien, R. Y. Engineering green fluorescent protein for improved brightness, longer wavelengths and fluorescence energy transfer. Curr. Biol. 6, 178–182 (1996).

    CAS  PubMed  Google Scholar 

  101. Mitra, R. D., Silva, C. M. & Youvan, D. C. Fluorescence resonance energy transfer between blue-emitting and red-shifted excitation derivatives of the green fluorescent protein. Gene 173, 13–17 (1996).

    CAS  PubMed  Google Scholar 

  102. Mahajan, N. P., Harrison-Shostak, D. C., Michaux, J. & Herman, B. Novel mutant green fluorescent protein protease substrates reveal the activation of specific caspases during apoptosis. Chem. Biol. 6, 401–409 (1999).

    CAS  PubMed  Google Scholar 

  103. Luo, K. Q., Yu, V. C., Pu, Y. & Chang, D. C. Application of the fluorescence resonance energy transfer method for studying the dynamics of caspase-3 activation during UV-induced apoptosis in living HeLa cells. Biochem. Biophys. Res. Commun. 283, 1054–1060 (2001).

    CAS  PubMed  Google Scholar 

  104. Rehm, M. et al. Single-cell fluorescence resonance energy transfer analysis demonstrates that caspase activation during apoptosis is a rapid process. Role of caspase-3. J Biol. Chem. 277, 24506–24514 (2002).

    CAS  PubMed  Google Scholar 

  105. Miyawaki, A. et al. Fluorescent indicators for Ca2+ based on green fluorescent proteins and calmodulin. Nature 388, 882–887 (1997).

    CAS  PubMed  Google Scholar 

  106. Miyawaki, A., Griesbeck, O., Heim, R. & Tsien, R. Y. Dynamic and quantitative Ca2+ measurements using improved cameleons. Proc. Natl Acad. Sci. USA 96, 2135–2140 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Miyawaki, A. & Tsien, R. Y. Monitoring protein conformations and interactions by fluorescence resonance energy transfer between mutants of green fluorescent protein. Methods Enzymol. 327, 472–500 (2000).

    CAS  PubMed  Google Scholar 

  108. Truong, K. et al. FRET-based in vivo Ca2+ imaging by a new calmodulin–GFP fusion molecule. Nature Struct. Biol. 8, 1069–1073 (2001).

    CAS  PubMed  Google Scholar 

  109. Romoser, V. A., Hinkle, P. M. & Persechini, A. Detection in living cells of Ca2+-dependent changes in the fluorescence emission of an indicator composed of two green fluorescent protein variants linked by a calmodulin-binding sequence. J. Biol. Chem. 272, 13270–13274 (1997). References 105 and 109 describe the first genetically encoded indicators to be composed of conformationally responsive elements sandwiched between two fluorescent proteins that can undergo FRET — a design that has since been extended to many other systems.

    CAS  PubMed  Google Scholar 

  110. Persechini, A. & Cronk, B. The relationship between the free concentrations of Ca2+ and Ca2+–calmodulin in intact cells. J. Biol. Chem. 274, 6827–6830 (1999).

    CAS  PubMed  Google Scholar 

  111. Sato, M., Hida, N., Ozawa, T. & Umezawa, Y. Fluorescent indicators for cyclic GMP based on cyclic GMP-dependent protein kinase I α and green fluorescent proteins. Anal. Chem. 72, 5918–5924 (2000).

    CAS  PubMed  Google Scholar 

  112. Honda, A. et al. Spatiotemporal dynamics of guanosine 3′,5′-cyclic monophosphate revealed by a genetically encoded, fluorescent indicator. Proc. Natl Acad. Sci. USA 98, 2437–2442 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Mochizuki, N. et al. Spatio-temporal images of growth-factor-induced activation of Ras and Rap1. Nature 411, 1065–1068 (2001).

    CAS  PubMed  Google Scholar 

  114. Kalab, P., Weis, K. & Heald, R. Visualization of a Ran-GTP gradient in interphase and mitotic Xenopus egg extracts. Science 295, 2452–2456 (2002).

    CAS  PubMed  Google Scholar 

  115. Kurokawa, K. et al. A pair of FRET–based probes for tyrosine phosphorylation of the CrkII adaptor protein in vivo. J. Biol. Chem. 276, 31305–31310 (2001).

    CAS  PubMed  Google Scholar 

  116. Ting, A. Y., Kain, K. H., Klemke, R. L. & Tsien, R. Y. Genetically encoded fluorescent reporters of protein tyrosine kinase activities in living cells. Proc. Natl Acad. Sci. USA 98, 15003–15008 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Sato, M., Ozawa, T., Inukai, K., Asano, T. & Umezawa, Y. Fluorescent indicators for imaging protein phosphorylation in single living cells. Nature Biotechnol. 20, 287–294 (2002).

    CAS  Google Scholar 

  118. Zhang, J., Ma, Y., Taylor, S. S. & Tsien, R. Y. Genetically encoded reporters of protein kinase A activity reveal impact of substrate tethering. Proc. Natl Acad. Sci. USA 98, 14997–15002 (2001). References 116 and 118 describe a general strategy for non-destructively imaging kinase activities in living cells using FRET-based reporters.

    CAS  PubMed  PubMed Central  Google Scholar 

  119. Sakai, R., Repunte-Canonigo, V., Raj, C. D. & Knopfel, T. Design and characterization of a DNA-encoded, voltage-sensitive fluorescent protein. Eur. J. Neurosci. 13, 2314–2318 (2001).

    CAS  PubMed  Google Scholar 

  120. Adams, S. R., Harootunian, A. T., Buechler, Y. J., Taylor, S. S. & Tsien, R. Y. Fluorescence ratio imaging of cyclic AMP in single cells. Nature 349, 694–697 (1991).

    CAS  PubMed  Google Scholar 

  121. Zaccolo, M. et al. A genetically encoded, fluorescent indicator for cyclic AMP in living cells. Nature Cell Biol. 2, 25–29 (2000).

    CAS  PubMed  Google Scholar 

  122. Zaccolo, M. & Pozzan, T. Discrete microdomains with high concentration of cAMP in stimulated rat neonatal cardiac myocytes. Science 295, 1711–1715 (2002). The use of a genetically encodable intermolecular FRET-based cAMP indicator in cardiac myocytes showed an unusual compartmentalization of cAMP.

    CAS  PubMed  Google Scholar 

  123. Bacskai, B. J. et al. Spatially resolved dynamics of cAMP and protein kinase A subunits in Aplysia sensory neurons. Science 260, 222–226 (1993).

    CAS  PubMed  Google Scholar 

  124. Hempel, C. M., Vincent, P., Adams, S. R., Tsien, R. Y. & Selverston, A. I. Spatio-temporal dynamics of cAMP signals in an intact neural circuit. Nature 384, 166–169 (1996).

    CAS  PubMed  Google Scholar 

  125. Periasamy, A., Kay, S. A. & Day, R. N. in Functional Imaging and Optical Manipulation of Living Cells. SPIE Proc. Vol. 2983 (eds Farkas, D. L. & Tromberg, B. J.) (The International Society for Optical Engineers, USA, 1997).

    Google Scholar 

  126. Llopis, J. et al. Ligand-dependent interactions of coactivators SRC-1 and PBP with nuclear hormone receptors can be imaged in live cells and are required for transcription. Proc. Natl Acad. Sci. USA 97, 4363–4368 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Janetopoulos, C., Jin, T. & Devreotes, P. Receptor-mediated activation of heterotrimeric G proteins in living cells. Science 291, 2408–2411 (2001).

    CAS  PubMed  Google Scholar 

  128. Damelin, M. & Silver, P. A. Mapping interactions between nuclear transport factors in living cells reveals pathways through the nuclear pore complex. Mol. Cell 5, 133–140 (2000).

    CAS  PubMed  Google Scholar 

  129. Ruehr, M. L., Zakhary, D. R., Damron, D. S. & Bond, M. Cyclic AMP-dependent protein kinase binding to A-kinase anchoring proteins in living cells by fluorescence resonance energy transfer of green fluorescent protein fusion proteins. J. Biol. Chem. 274, 33092–33096 (1999).

    CAS  PubMed  Google Scholar 

  130. Siegel, R. M. et al. Fas preassociation required for apoptosis signaling and dominant inhibition by pathogenic mutations. Science 288, 2354–2357 (2000).

    CAS  PubMed  Google Scholar 

  131. Xia, Z., Zhou, Q., Lin, J. & Liu, Y. Stable SNARE complex prior to evoked synaptic vesicle fusion revealed by fluorescence resonance energy transfer. J. Biol. Chem. 276, 1766–1771 (2001).

    CAS  PubMed  Google Scholar 

  132. Li, H. Y. et al. Protein–protein interaction of FHL3 with FHL2 and visualization of their interaction by green fluorescent proteins (GFP) two-fusion fluorescence resonance energy transfer (FRET). J. Cell. Biochem. 80, 293–303 (2001).

    CAS  PubMed  Google Scholar 

  133. Mas, P., Devlin, P. F., Panda, S. & Kay, S. A. Functional interaction of phytochrome B and cryptochrome 2. Nature 408, 207–211 (2000).

    CAS  PubMed  Google Scholar 

  134. Tertoolen, L. G. et al. Dimerization of receptor protein-tyrosine phosphatase α in living cells. B.M.C. Cell Biol. 2, 8 (2001).

    CAS  Google Scholar 

  135. Ghosh, I., Hamilton, A. D. & Regan, L. Antiparallel leucine zipper-directed protein reassembly: Application to the green fluorescent protein. J. Am. Chem. Soc. 122, 5658–5659 (2000).

    CAS  Google Scholar 

  136. Hu, C. -D., Chinenov, Y. & Kerppola, T. K. Visualization of interactions among bZIP and Rel family proteins in living cells using bimolecular fluorescence complementation. Mol. Cell 9, 789–798 (2002).

    CAS  PubMed  Google Scholar 

  137. Rossi, F., Charlton, C. A. & Blau, H. M. Monitoring protein-protein interactions in intact eukaryotic cells by β-galactosidase complementation. Proc. Natl Acad. Sci. USA 94, 8405–8410 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  138. Michnick, S. W., Remy, I., Campbell-Valois, F. X., Vallee-Belisle, A. & Pelletier, J. N. Detection of protein–protein interactions by protein fragment complementation strategies. Methods Enzymol. 328, 208–230 (2000).

    CAS  PubMed  Google Scholar 

  139. Wehrman, T., Kleaveland, B., Her, J. H., Balint, R. F. & Blau, H. M. Protein–protein interactions monitored in mammalian cells via complementation of β-lactamase enzyme fragments. Proc. Natl Acad. Sci. USA 99, 3469–3474 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  140. Galarneau, A., Primeau, M., Trudeau, L. E. & Michnick, S. W. β-Lactamase protein fragment complementation assays as in vivo and in vitro sensors of protein–protein interactions. Nature Biotechnol. 20, 619–622 (2002).

    CAS  Google Scholar 

  141. Gordon, G. W., Berry, G., Liang, X. H., Levine, B. & Herman, B. Quantitative fluorescence resonance energy transfer measurements using fluorescence microscopy. Biophys. J. 74, 2702–2713 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Sorkin, A., McClure, M., Huang, F. & Carter, R. Interaction of EGF receptor and grb2 in living cells visualized by fluorescence resonance energy transfer (FRET) microscopy. Curr. Biol. 10, 1395–1398 (2000).

    CAS  PubMed  Google Scholar 

  143. Erickson, M. G., Alseikhan, B. A., Peterson, B. Z. & Yue, D. T. Preassociation of calmodulin with voltage-gated Ca(2+) channels revealed by FRET in single living cells. Neuron 31, 973–985 (2001).

    CAS  PubMed  Google Scholar 

  144. Chan, F. K. et al. Fluorescence resonance energy transfer analysis of cell surface receptor interactions and signaling using spectral variants of the green fluorescent protein. Cytometry 44, 361–368 (2001).

    CAS  PubMed  Google Scholar 

  145. Ng, T. et al. Imaging protein kinase Cα activation in cells. Science 283, 2085–2089 (1999).

    CAS  PubMed  Google Scholar 

  146. Verveer, P. J., Wouters, F. S., Reynolds, A. R. & Bastiaens, P. I. Quantitative imaging of lateral ErbB1 receptor signal propagation in the plasma membrane. Science 290, 1567–1570 (2000).

    CAS  PubMed  Google Scholar 

  147. Harpur, A. G., Wouters, F. S. & Bastiaens, P. I. Imaging FRET between spectrally similar GFP molecules in single cells. Nature Biotechnol. 19, 167–169 (2001).

    CAS  Google Scholar 

  148. Weiss, S. Fluorescence spectroscopy of single biomolecules. Science 283, 1676–1683 (1999).

    CAS  PubMed  Google Scholar 

  149. Sako, Y., Minoguchi, S. & Yanagida, T. Single-molecule imaging of EGFR signalling on the surface of living cells. Nature Cell Biol. 2, 168–172 (2000).

    CAS  PubMed  Google Scholar 

  150. Sonnleitner, A., Mannuzzu, L. M., Terakawa, S. & Isacoff, E. Y. Structural rearrangements in single ion channels detected optically in living cells. Proc. Natl Acad. Sci. USA 99, 12759–12764 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  151. Harms, G. S. et al. Single-molecule imaging of l-type Ca(2+) channels in live cells. Biophys. J. 81, 2639–2646 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  152. Ito, S. et al. Real-time observation of micrometastasis formation in the living mouse liver using a green fluorescent protein gene-tagged rat tongue carcinoma cell line. Int. J. Cancer 93, 212–217 (2001).

    CAS  PubMed  Google Scholar 

  153. Brown, E. B. et al. In vivo measurement of gene expression, angiogenesis and physiological function in tumors using multiphoton laser scanning microscopy. Nature Med. 7, 864–868 (2001).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank S. R. Adams and J. Babendure for their comments on the manuscript. This work was supported by a National Institutes of Health grant to R.Y.T. and postdoctoral fellowship to A.Y.T, a grant from the Alliance for Cellular Signaling (to J.Z. and R.Y.T.) and the Howard Hughes Medical Institute. J.Z. is supported in part by a postdoctoral fellowship from La Jolla Interfaces in Science and Burroughs Wellcome Fund, and R.E.C. is supported in part by a postdoctoral fellowship from the Canadian Institutes of Health Research.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Roger Y. Tsien.

Related links

Related links

DATABASES

Entrez

Kaede

Swiss-Prot

CFTR

EGF

GFP

ZBP

FURTHER INFORMATION

Roger Y. Tsien's laboratory

Alice Y. Ting's laboratory

BD Biosciences Clontech (EGFP, YFP-S65G/V68L/S72A/T203Y, CFP, DsRed2, HcRed, Fluorescent Timer, and DsRed-Express)

NanoLight Technology (Renilla mullerei GFP)

PanVera (FlAsH–EDT2 and ReAsH–EDT2 labelling kits)

Glossary

PHOTOBLEACHING

The irreversible destruction, by any one of a number of different mechanisms, of a fluorophore that is under illumination.

FLUORESCENCE RESONANCE ENERGY TRANSFER

(FRET).The non-radiative transfer of energy from a donor fluorophore to an acceptor fluorophore that is typically < 80 Å away. FRET will only occur between fluorophores in which the emission spectrum of the donor has a significant overlap with the excitation of the acceptor.

QUANTUM YIELD

The probability of luminescence occurring in given conditions — expressed by the ratio of the number of photons that are emitted by the luminescing species to the number of photons that are absorbed.

pKa

The pH at which a molecule, or a particular site within a molecule, carries an ionizable H+ 50% of the time.

PHYCOBILIPROTEINS

Proteins from blue-green algae and red algae that exhibit intense fluorescence owing to the presence of multiple bilin chromophores that are covalently attached to the protein.

CHROMOPHORE

The core portion of a molecule that is directly responsible for absorbing photons. Chromophores usually contain alternating single and double bonds.

FLUOROPHORE

A chromophore that can re-emit photons.

MUSHROOM BODIES

Two prominent bilaterally symmetrical structures in the fly brain that are crucial for olfactory learning and memory.

FLUORESCENCE-LIFETIME IMAGING MICROSCOPY

(FLIM). An imaging technique in which the lifetime, rather than the intensity, of the fluorescent signal is measured. This approach can be used to measure FRET.

FLUORESCENCE-ACTIVATED CELL SORTING

(FACS). A flow cytometry application in which live fluorescent cells are excited at a specific wavelength and then sorted into physically separated subpopulations on the basis of their fluorescence emission.

POSITRON EMISSION TOMOGRAPHY

(PET). Positron emission tomography is an imaging technique that is used to detect decaying nuclides, such as 15O, 13N, 11C, 18F, 124I and 94mTc.

MAGNETIC RESONANCE IMAGING

The use of radio waves in the presence of a magnetic field to extract information from certain atomic nuclei (most commonly hydrogen, for example, in water). This technique is used to show certain types of tissue damage and the presence of tumours.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Zhang, J., Campbell, R., Ting, A. et al. Creating new fluorescent probes for cell biology. Nat Rev Mol Cell Biol 3, 906–918 (2002). https://doi.org/10.1038/nrm976

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrm976

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing