Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Phosphorescent Imaging of Living Cells Using a Cyclometalated Iridium(III) Complex

  • Dik-Lung Ma ,

    edmondma@hkbu.edu.hk

    Affiliation Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China

  • Hai-Jing Zhong,

    Affiliation State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China

  • Wai-Chung Fu,

    Affiliation Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China

  • Daniel Shiu-Hin Chan,

    Affiliation Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China

  • Hiu-Yee Kwan,

    Affiliation Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong, China

  • Wang-Fun Fong,

    Affiliation Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong, China

  • Lai-Hon Chung,

    Affiliation Department of Biology and Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong SAR, People's Republic of China

  • Chun-Yuen Wong,

    Affiliation Department of Biology and Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong SAR, People's Republic of China

  • Chung-Hang Leung

    Affiliation State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China

Abstract

A cell permeable cyclometalated iridium(III) complex has been developed as a phosphorescent probe for cell imaging. The iridium(III) solvato complex [Ir(phq)2(H2O]2)] preferentially stains the cytoplasm of both live and dead cells with a bright luminescence.

Introduction

Luminescence imaging of biological specimens using non-invasive probes is a basic technique in life and biomedical sciences for studying the morphologic characteristics of tissue at high resolution [1][5]. Since the cell is the primary structural and functional unit of all known living organisms, the morphological aberration of certain cell types can lead to various diseases such as sickle cell anemia [6], [7]. Consequently, a great deal of attention has been invested into the development of luminescent probes for live cell imaging in recent years. Currently, organic dyes constitute the majority of the most commonly-used fluorescent probes [8]. However, organic dyes can be subject to various drawbacks, including small Stokes shift values and short luminescence lifetimes [9][11]. In this context, luminescent transition metal complexes have arisen as viable alternatives to organic fluorophores for sensing and imaging applications due to the following advantages: [12][36] (i) tunable excitation and emission maxima over the visible region without the need for lengthy synthetic protocols; (ii) tunable emission energies by modification of the ancillary ligands; (iii) large Stokes shift for facile separation of excitation and emission wavelengths and elimination of self-quenching; (iv) relatively long phosphorescence lifetimes that can mitigate a short-lived autofluorescence background through the use of time-resolved spectroscopy which offers high selectivity; and (v) good solubility in aqueous solution (containing <0.01% organic solvent).

In eukaryotes, the cytoplasm is an aqueous fluid that primarily consists of a transparent substance termed hyaloplasm or cytosol. Numerous life processes take place within the cytoplasm, including protein synthesis, metabolic reactions, and cellular signaling. However, only a few phosphorescent metal complexes have been developed for cytoplasmic staining. For example, Coogan and co-workers have reported a series of Re(I) complexes of type fac-[Re(bisim)L(CO)3]+ containing highly lipophilic esters of 3-hydroxymethylpyridine as luminescence agents that selectively distribute in membranes and membrane structures within the cytoplasm of living cells [35]. Barton and co-workers investigated a series of phosphorescent ruthenium(II) complexes with different ancillary ligands that selectively stain the cytoplasm [37]. The groups of Li and Lo have developed a series of cationic iridium(III) complexes as phosphorescent probes for luminescence staining of the cytoplasm of living cells [29], [38][40].

Iridium(III) complexes with d6 electronic structures often possess excellent photophysical properties such as tunable excitation and emission wavelengths (from blue to red), high luminescent quantum yields, and relatively long phosphorescence lifetimes [41], [42]. Iridium complexes have received considerable attention in inorganic photochemistry [43][48], phosphorescent materials for optoelectronics [49][60], chemosensors [61][66], biolabeling[67][69], live cell imaging [29], [70][72], and in vivo tumor imaging [73]. As part of our continuous efforts, the cyclometalated iridium(III) solvato complex [Ir(ppy)2(solv)2]+ has been utilized as a selective luminescent switch-on probe for histidine/histidine-rich proteins and a dye for protein staining in sodium dodecyl sulfate polyacrylamide gels [74]. Subsequently, Li and co-workers reported iridium(III) solvato complex [Ir(ppy)2(DMSO)2]+ as a luminescence agent for imaging live cell nuclei [75]. Thus, we were interested to investigate the effect of varying the extent of conjugation of the CN co-ligand on the photophysical properties of this type of complex. We herein report the application of iridium(III) solvato complex [Ir(phq)2(solv)2]+ (1) for the detection of histidine/histidine-rich proteins and for luminescence imaging in cells. We demonstrate that the complex is successfully taken up by both living and dead cells and can function as a selective luminescent probe for cytoplasmic staining.

The luminescence response of complex 1 to various natural amino acids was investigated (Figure 3). Complex 1 is non-emissive in aqueous buffered solution in the absence of analyte. In the presence of histidine, complex 1 exhibits an intense phosphorescence emission at λmax = 598 nm. No significant change in the emission of the complex 1 was observed upon the addition of other natural amino acids (Figure 3). This result indicates that complex 1 displays a high degree of selectivity for histidine over other amino acids. Furthermore, the emission maxima of 1 falls on the boundary of the near-infrared (NIR) “optical window” (600–900 nm), which is a region where the absorbance of photons by biological tissues decreases to a minimum [66]. This suggests that complex 1 may be potentially developed for in vivo imaging applications. By comparison, the previously reported iridium(III) complex [Ir(ppy)2(solv)2]+ (3) utilized for cellular staining emits green phosphorescence at a shorter wavelength of 505 nm in the presence of histidine, which is outside the optical window [74], [75].

We next studied the luminescence response of complex 1 with bovine serum albumin (BSA) and calf-thymus DNA (ct DNA). Complex 1 displayed an intense luminescence upon interaction with the histidine-rich BSA, but was only weakly emissive in the presence of ct DNA (Figure 4). Furthermore, the change in luminescence intensity of complex 1 upon the addition of various amounts (12.5−100 µM) of histidine or BSA was investigated. The results showed that BSA or histidine were able to induce significant luminescence enhancements in complex 1 (Figure 5). In combination with previously published reports [74], [76], we propose that the labile solvato co-ligands of complex 1 are displaced by the imidazole N-donor moieties of histidine residues via coordinative bond formation. This shelters the metal center within the hydrophobic environment of the protein, reducing solvent-mediated non-radiative decay of the excited state and thereby enhancing the phosphorescence of complex 1.

Results and Discussion

The cyclometalated iridium(III) solvent complexes 1–3 (Figure 1) were synthesized according to previously reported methods (see Materials and Methods). To examine the effect of varying the extent of the conjugation of the CN co-ligand on the emissive color of the complexes, we first obtained luminescence photographs of the complexes in dimethyl sulfoxide (DMSO) (Figure 2A). Interestingly, complex 1 emits an intense orange luminescence in DMSO under UV-transillumination and was thus considered as a promising candidate for further cell imaging studies. On the other hand, luminescence of 1 was significantly suppressed in Tris buffer (Figure 2B). We rationalize that the reduced luminescence intensity of 1 in aqueous solution is due to non-radiative decay of the excited state of complex 1 by complex-solvent interactions. Presumably, this effect is less pronounced in DMSO, leading to a higher luminescence signal.

thumbnail
Figure 1. Chemical structures of iridium(III) solvato complexes 1–3 bearing different CN ligands.

https://doi.org/10.1371/journal.pone.0055751.g001

thumbnail
Figure 2. Luminescence photographs (upper panel) of (left) [Ir(phq)2(H2O)2)]OTf (1), (middle) [Ir(ppy)2(H2O)2]OTf (3), and (right) [Ir(bzq)2(H2O)2]OTf (2) at 1 mM concentration in DMSO solution under UV-transillumination.

Emission spectra (lower panel) of complex 1 (50 µM) in 20 mM Tris buffer (pH 7.4) and DMSO.

https://doi.org/10.1371/journal.pone.0055751.g002

thumbnail
Figure 3. Emission spectra of complex 1 (50 µM) in 20 mM Tris buffer (pH 7.4) with various natural amino acids (200 µM).

Group 1: L-alanine, L-arginine, L-asparagine, L-glutamine, L-threonine; Group 2: L-glycine, L-isoleucine, L-lysine, L-phenylalanine, L-proline, L-serine; Group 3: L-tryptophan, L-tyrosine, L-valine, L-glutamic acid, L-cysteine, L-methionine.

https://doi.org/10.1371/journal.pone.0055751.g003

thumbnail
Figure 4. Photograph image of complex 1 (50 µM) in absence (left) or presence of BSA (50 µM, middle) or ct DNA (50 µM, right) under UV-transillumination.

https://doi.org/10.1371/journal.pone.0055751.g004

thumbnail
Figure 5. Luminescence intensity changes of complex 1 (50 µM) in 20 mM Tris buffer (pH 7.4) with various amounts of BSA or histidine (0, 12.5, 25, 50 and 100 µM).

https://doi.org/10.1371/journal.pone.0055751.g005

We also investigated the application of iridium(III) complex 1 for staining fixed cells. HeLa cells fixed with 4% paraformaldehyde exhibited strong intracellular luminescence in the cytoplasm upon incubation with complex 1 (Figure 8b). Similar to the results with live cells, only weak luminescence was observed in the nucleus of the fixed cells (Figure 8c,d). These results suggest that complex 1 is an effective luminescent cytoplasmic stain for both living and dead cells.

The practical application of complex 1 as a luminescent probe in living cells was investigated using confocal laser scanning microscopy (Figure 6). HeLa cells showed negligible background fluorescence. After incubation with 10 µM of 1 in DMSO/PBS (pH 7.4, 1∶99, v/v) for 10 min at 37°C, an intense intracellular luminescence was observed particularly in the cytoplasm of the cells, suggesting that the iridium(III) complex is cytoplasmic permeable. No cell death was observed under the staining and imaging conditions used (Figure 7). Overlay images revealed that the luminescence pattern of complex 1 differed considerably from that of DNA-binding dye Hoechst 33258 (Figure 6d). Furthermore, a large signal ratio was observed between the nuclei and cytoplasm, indicating that complex 1 prefers to stain the cytoplasmic regions of the cells. We presume that the observed luminescence enhancement of complex 1 is due to its interactions with histidine or histidine-rich proteins in the cellular cytoplasm. These results indicate that complex 1 acts as a luminescent imaging agent for live cells without requiring prior membrane permeabilization.

thumbnail
Figure 6. Brightfield images of live HeLa cells (top left).

Luminescence images of cells stained with complex 1 (10 µM) in DMSO/PBS (pH 7.4, 1∶99 v/v) for 10 min at 37°C (top right) and then with Hoechst 33258 for a further 20 min (bottom left). Overlay of images in (b) and (c) (bottom right).

https://doi.org/10.1371/journal.pone.0055751.g006

thumbnail
Figure 7. Cytotoxicity of complex 1 (concentration of 1 = 10 µM; incubation time  = 10 min).

https://doi.org/10.1371/journal.pone.0055751.g007

thumbnail
Figure 8. Brightfield images of fixed HeLa cells (top left).

Luminescence images of cells stained with complex 1 (10 µM) in DMSO/PBS (pH 7.4, 1∶99 v/v) for 10 min at 37°C (top right) and then with Hoechst 33258 for a further 20 min (bottom left). Overlay of images in (b) and (c) (bottom right).

https://doi.org/10.1371/journal.pone.0055751.g008

Conclusions

In conclusion, we have presented the cytoplasmic permeable iridium(III) complex 1 as a phosphorescent dye for live and fixed cell imaging. Complex 1 shows a bright phosphorescence in living cells, and effectively enters and stains the cytoplasm. Given that the emission properties of metal complexes can be fine-tuned through modifications of auxiliary ligands, we envision that further improvements can be achieved in the application of luminescent iridium(III) complexes as cellular imaging probes.

Materials and Methods

Materials

Iridium chloride hydrate (IrCl3.xH2O) was purchased from Precious Metals Online. DMSO, L-alanine, L-arginine, L-asparagine, L-glutamine, L-glycine, L-isoleucine, L-lysine, L-phenylalanine, L-proline, L-serine, L-threonine, L-tryptophan, L-tyrosine, L-valine, L-glutamic acid, L-cysteine, L-methionine, L-histidine, bovine serum albumin, and calf thymus DNA were obtained from Sigma (St. Louis, MO). Hoechst 33258 and cell culture reagents were purchased from Invitrogen (Carlsbad, CA).

Synthesis of [Ir(CN)2(H2O)2]OTf (1–3, where OTf = trifluoromethanesulfonate)

The following complexes were prepared using literature methods: [Ir2(ppy)4Cl2] [77], [Ir2(bzq)4Cl2] [77], [Ir2(phq)4Cl2] [78], [Ir(ppy)2(H2O)2]OTf [79], [Ir(phq)2(H2O)2]OTf [76], and [Ir(bzq)2(H2O)2]OTf [76].

Emission Measurement

A stock solution of the complex [Ir(phq)2(H2O)2)]OTf was diluted (50 µM, final concentration) into Tris buffer (20 mM, pH 7.4) with the corresponding concentrations of histidine (200 µM), groups of other amino acids (200 µM), ct DNA (50 µM) or BSA (50 µM). The emission spectra were recorded in the 555–730 nm range, after equilibration at 25°C for 5 min. Excitation wavelength = 365 nm.

Cell Culture

HeLa cells were maintained in minimum essential medium (MEM) supplemented with fetal bovine serum (10%), penicillin (100 U mL−1), streptomycin (100 µg mL−1) at 37°C under a humidified atmosphere with 5% CO2.

Luminescence Imaging

For colocalization imaging of living cells. The cells were washed with PBS, then incubated with 10 µM of iridium complex in DMSO/PBS (pH 7.4, 1∶99, v/v) for 10 min at 37°C, and then further incubated with Hoechst 33258 for another 20 min before imaging.

For colocalization imaging of fixed cells. The cells were detached from the culture and were fixed with 4% paraformaldehyde at room temperature for 20 min. After washing with PBS, the fixed cells were incubated with 10 µM of iridium complex in DMSO/PBS (pH 7.4, 1∶99, v/v) for 10 min at 37°C, and then further stained with Hoechst 33258 for another 20 min. After washing with PBS, the coverslips were separated from the chamber, and the cells were mounted with 10% glycerol and sealed with nail varnish on a glass substrate.

Author Contributions

Directed the research: DLM CHL. Conceived and designed the experiments: DLM CHL. Performed the experiments: HJZ WCF HYK. Analyzed the data: DSHC HJZ LHC. Contributed reagents/materials/analysis tools: DLM CHL WFF CYW. Wrote the paper: DSHC WCF.

References

  1. 1. Evans CL, Potma EO, Puoris'haag M, Cote D, Lin CP, et al. (2005) Chemical imaging of tissue in vivo with video-rate coherent anti-Stokes Raman scattering microscopy. Proc Natl Acad Sci U S A 102: 16807–16812.
  2. 2. Huisken J, Swoger J, Del BF, Wittbrodt J, Stelzer EHK (2004) Optical Sectioning Deep Inside Live Embryos by Selective Plane Illumination Microscopy. Science 305: 1007–1010.
  3. 3. Rust MJ, Bates M, Zhuang X (2006) Sub-diffraction-limit imaging by stochastic optical reconstruction microscopy (STORM). Nat Methods 3: 793–796.
  4. 4. Stephens DJ, Allan VJ (2003) Light microscopy techniques for live cell imaging. Science 300: 82–86.
  5. 5. Yu M, Li F, Chen Z, Hu H, Zhan C, et al. (2009) Laser Scanning Up-Conversion Luminescence Microscopy for Imaging Cells Labeled with Rare-Earth Nanophosphors. Anal Chem 81: 930–935.
  6. 6. Ballas SK, Mohandas N (2004) Sickle Red Cell Microrheology and Sickle Blood Rheology. Microcirculation 11: 209–225.
  7. 7. Horne MK (1981) Sickle cell anemia as a rheologic disease. Am J Med 70: 288–298.
  8. 8. Yang Y, Zhao Q, Feng W, Li F (2012) Luminescent Chemodosimeters for Bioimaging. Chem Rev dx.doi.org/10.1021/cr2004103.
  9. 9. Kuil J, Steunenberg P, Chin PTK, Oldenburg J, Jalink K, et al. (2011) Peptide-Functionalized Luminescent Iridium Complexes for Lifetime Imaging of CXCR4 Expression. ChemBioChem 12: 1897–1903.
  10. 10. Martin RM, Leonhardt H, Cardoso MC (2005) DNA labeling in living cells. Cytometry, Part A 67A: 45–52.
  11. 11. Pfeifer GP, You Y-H, Besaratinia A (2005) Mutations induced by ultraviolet light. Mutat Res 571: 19–31.
  12. 12. Guerchais V, Fillaut J-L (2011) Sensory luminescent iridium(III) and platinum(II) complexes for cation recognition. Coord Chem Rev 255: 2448–2457.
  13. 13. You Y, Lee S, Kim T, Ohkubo K, Chae W-S, et al. (2011) Phosphorescent Sensor for Biological Mobile Zinc. J Am Chem Soc 133: 18328–18342.
  14. 14. Lee P-K, Law WH-T, Liu H-W, Lo KK-W (2011) Luminescent Cyclometalated Iridium(III) Polypyridine Di-2-picolylamine Complexes: Synthesis, Photophysics, Electrochemistry, Cation Binding, Cellular Internalization, and Cytotoxic Activity. Inorg Chem 50: 8570–8579.
  15. 15. Louie M-W, Liu H-W, Lam MH-C, Lau T-C, Lo KK-W (2009) Novel Luminescent Tricarbonylrhenium(I) Polypyridine Tyramine-Derived Dipicolylamine Complexes as Sensors for Zinc(II) and Cadmium(II) Ions. Organometallics 28: 4297–4307.
  16. 16. Fernandez-Moreira V, Thorp-Greenwood FL, Coogan MP (2010) Application of d6 transition metal complexes in fluorescence cell imaging. Chem Commun 46: 186–202.
  17. 17. Zhao Q, Huang C, Li F (2011) Phosphorescent heavy-metal complexes for bioimaging. Chem Soc Rev 40: 2508–2524.
  18. 18. Lo KK-W, Choi AW-T, Law WH-T (2012) Applications of luminescent inorganic and organometallic transition metal complexes as biomolecular and cellular probes. Dalton Trans 41: 6021–6047.
  19. 19. Lo KK-W, Hui W-K, Chung C-K, Tsang KH-K, Lee TK-M, et al. (2006) Luminescent transition metal complex biotin conjugates. Coord Chem Rev 250: 1724–1736.
  20. 20. Yam VW-W, Wong KM-C (2011) Luminescent metal complexes of d6, d8 and d10 transition metal centres. Chem Commun 47: 11579–11592.
  21. 21. Lo KK-W, Hui W-K, Chung C-K, Tsang KH-K, Ng DC-M, et al. (2005) Biological labelling reagents and probes derived from luminescent transition metal polypyridine complexes. Coord Chem Rev 249: 1434–1450.
  22. 22. Baggaley E, Weinstein JA, Williams JAG (2012) Lighting the way to see inside the live cell with luminescent transition metal complexes. Coord Chem Rev 256: 1762–1785.
  23. 23. Zhang R, Ye Z, Yin Y, Wang G, Jin D, et al. (2012) Developing Red-Emissive Ruthenium(II) Complex-Based Luminescent Probes for Cellular Imaging. Bioconjug Chem 23: 725–733.
  24. 24. Wu P, Wong EL-M, Ma D-L, Tong GS-M, Ng K-M, et al. (2009) Cyclometalated Platinum(II) Complexes as Highly Sensitive Luminescent Switch-On Probes for Practical Application in Protein Staining and Cell Imaging. Chem Eur J 15: 3652–3656.
  25. 25. Koo C-K, So LKY, Wong K-L, Ho Y-M, Lam Y-W, et al. (2010) Inside Cover: A Triphenylphosphonium-Functionalised Cyclometalated Platinum(II) Complex as a Nucleolus-Specific Two-Photon Molecular Dye (Chem. Eur. J. 13/2010). Chem Eur J 16: 3868–3868.
  26. 26. Yang T, Xia A, Liu Q, Shi M, Wu H, et al. (2011) Polymer nanoparticles with an embedded phosphorescent osmium(ii) complex for cell imaging. J Mater Chem 21: 5360–5367.
  27. 27. Botchway SW, Charnley M, Haycock JW, Parker AW, Rochester DL, et al. (2008) Time-resolved and two-photon emission imaging microscopy of live cells with inert platinum complexes. Proc Natl Acad Sci U S A 105: 16071–16076.
  28. 28. Tian X, Gill MR, Cantón I, Thomas JA, Battaglia G (2011) Live Cell Luminescence Imaging As a Function of Delivery Mechanism. ChemBioChem 12: 548–551.
  29. 29. Zhao Q, Yu M, Shi L, Liu S, Li C, et al. (2010) Cationic Iridium(III) Complexes with Tunable Emission Color as Phosphorescent Dyes for Live Cell Imaging. Organometallics 29: 1085–1091.
  30. 30. Neugebauer U, Pellegrin Y, Devocelle M, Forster RJ, Signac W, et al.. (2008) Ruthenium polypyridyl peptide conjugates: membrane permeable probes for cellular imaging. Chem Commun: 5307–5309.
  31. 31. Gill MR, Thomas JA (2012) Ruthenium(ii) polypyridyl complexes and DNA-from structural probes to cellular imaging and therapeutics. Chem Soc Rev 41: 3179–3192.
  32. 32. Xiong L, Zhao Q, Chen H, Wu Y, Dong Z, et al. (2010) Phosphorescence Imaging of Homocysteine and Cysteine in Living Cells Based on a Cationic Iridium(III) Complex. Inorg Chem 49: 6402–6408.
  33. 33. O'Connor NA, Stevens N, Samaroo D, Solomon MR, Marti AA, et al.. (2009) A covalently linked phenanthridine-ruthenium(ii) complex as a RNA probe. Chem Commun: 2640–2642.
  34. 34. Lau JS-Y, Lee P-K, Tsang KH-K, Ng CH-C, Lam Y-W, et al. (2008) Luminescent Cyclometalated Iridium(III) Polypyridine Indole Complexes Synthesis, Photophysics, Electrochemistry, Protein-Binding Properties, Cytotoxicity, and Cellular Uptake. Inorg Chem 48: 708–718.
  35. 35. Amoroso AJ, Coogan MP, Dunne JE, Fernandez-Moreira V, Hess JB, et al.. (2007) Rhenium fac tricarbonyl bisimine complexes: Biologically useful fluorochromes for cell imaging applications. Chem Commun: 3066–3068.
  36. 36. You Y, Han Y, Lee Y-M, Park SY, Nam W, et al. (2011) Phosphorescent Sensor for Robust Quantification of Copper(II) Ion. J Am Chem Soc 133: 11488–11491.
  37. 37. Puckett CA, Barton JK (2007) Methods to Explore Cellular Uptake of Ruthenium Complexes. J Am Chem Soc 129: 46–47.
  38. 38. Yu M, Zhao Q, Shi L, Li F, Zhou Z, et al.. (2008) Cationic iridium(III) complexes for phosphorescence staining in the cytoplasm of living cells. Chem Commun: 2115–2117.
  39. 39. Liu WH-W, Zhang KY, Law H-T, Lo KK-W (2010) Cyclometalated Iridium(III) Bipyridine Complexes Functionalized with an N-Methylamino-oxy Group as Novel Phosphorescent Labeling Reagents for Reducing Sugars. Organometallics 29: 3474–3476.
  40. 40. Lee P-K, Liu H-W, Yiu S-M, Louie M-W, Lo KK-W (2011) Luminescent cyclometallated iridium(iii) bis(quinolylbenzaldehyde) diimine complexes-synthesis, photophysics, electrochemistry, protein cross-linking properties, cytotoxicity and cellular uptake. Dalton Trans 40: 2180–2189.
  41. 41. Murphy L, Congreve A, Palsson L-O, Williams JAG (2010) The time domain in co-stained cell imaging: time-resolved emission imaging microscopy using a protonatable luminescent iridium complex. Chem Commun 46: 8743–8745.
  42. 42. Steunenberg P, Ruggi A, van dBNS, Buckle T, Kuil J, et al. (2012) Phosphorescence Imaging of Living Cells with Amino Acid-Functionalized Tris(2-phenylpyridine)iridium(III) Complexes. Inorg Chem 51: 2105–2114.
  43. 43. Ikai M, Tokito S, Sakamoto Y, Suzuki T, Taga Y (2001) Highly efficient phosphorescence from organic light-emitting devices with an exciton-block layer. Phys Rev Lett 79: 156–158.
  44. 44. Baldo MA, Lamansky S, Burrows PE, Thompson ME, Forrest SR (1999) Very high-efficiency green organic light-emitting devices based on electrophosphorescence. Phys Rev Lett 75: 4–6.
  45. 45. Tamayo AB, Alleyne BD, Djurovich PI, Lamansky S, Tsyba I, et al. (2003) Synthesis and Characterization of Facial and Meridional Tris-cyclometalated Iridium(III) Complexes. J Am Chem Soc 125: 7377–7387.
  46. 46. Coppo P, Plummer EA, De Cola L (2004) Tuning iridium(iii) phenylpyridine complexes in the “almost blue” region. Chem Commun: 1774–1775.
  47. 47. Tsuboyama A, Takiguchi T, Okada S, Osawa M, Hoshino M, et al.. (2004) A novel dinuclear cyclometalated iridium complex bridged with 1,4-bis[pyridine-2-yl]benzene: its structure and photophysical properties. Dalton Trans: 1115–1116.
  48. 48. Lau M-K, Cheung K-M, Zhang Q-F, Song Y, Wong W-T, et al. (2004) Iridium(III) and rhodium(III) cyclometalated complexes containing sulfur and selenium donor ligands. J Organomet Chem 689: 2401–2410.
  49. 49. Baldo MA, Thompson ME, Forrest SR (2000) High-efficiency fluorescent organic light-emitting devices using a phosphorescent sensitizer. Nature 403: 750–753.
  50. 50. Grushin VV, Herron N, LeCloux DD, Marshall WJ, Petrov VA, et al.. (2001) New, efficient electroluminescent materials based on organometallic Ir complexes. Chem Commun: 1494–1495.
  51. 51. Chen F-C, Yang Y, Thompson ME, Kido J (2002) High-performance polymer light-emitting diodes doped with a red phosphorescent iridium complex. Phys Rev Lett 80: 2308–2310.
  52. 52. Nazeeruddin MK, Humphry-Baker R, Berner D, Rivier S, Zuppiroli L, et al. (2003) Highly Phosphorescence Iridium Complexes and Their Application in Organic Light-Emitting Devices. J Am Chem Soc 125: 8790–8797.
  53. 53. Beeby A, Bettington S, Samuel IDW, Wang Z (2003) Tuning the emission of cyclometalated iridium complexes by simple ligand modification. J Mater Chem 13: 80–83.
  54. 54. Slinker JD, Gorodetsky AA, Lowry MS, Wang J, Parker S, et al. (2004) Efficient Yellow Electroluminescence from a Single Layer of a Cyclometalated Iridium Complex. J Am Chem Soc 126: 2763–2767.
  55. 55. Sandee AJ, Williams CK, Evans NR, Davies JE, Boothby CE, et al. (2004) Solution-Processible Conjugated Electrophosphorescent Polymers. J Am Chem Soc 126: 7041–7048.
  56. 56. Kim JI, Shin I-S, Kim H, Lee J-K (2005) Efficient Electrogenerated Chemiluminescence from Cyclometalated Iridium(III) Complexes. J Am Chem Soc 127: 1614–1615.
  57. 57. Finkenzeller WJ, Hofbeck T, Thompson ME, Yersin H (2007) Triplet State Properties of the OLED Emitter Ir(btp)2(acac): Characterization by Site-Selective Spectroscopy and Application of High Magnetic Fields. Inorg Chem 46: 5076–5083.
  58. 58. Hanson K, Tamayo A, Diev VV, Whited MT, Djurovich PI, et al. (2010) Efficient Dipyrrin-Centered Phosphorescence at Room Temperature from Bis-Cyclometalated Iridium(III) Dipyrrinato Complexes. Inorg Chem 49: 6077–6084.
  59. 59. Wu C, Chen H-F, Wong K-T, Thompson ME (2010) Study of Ion-Paired Iridium Complexes (Soft Salts) and Their Application in Organic Light Emitting Diodes. J Am Chem Soc 132: 3133–3139.
  60. 60. Rausch AF, Thompson ME, Yersin H (2009) Blue Light Emitting Ir(III) Compounds for OLEDs - New Insights into Ancillary Ligand Effects on the Emitting Triplet State. J Phys Chem A 113: 5927–5932.
  61. 61. Liu J, Liu Y, Liu Q, Li C, Sun L, et al. (2011) Iridium(III) Complex-Coated Nanosystem for Ratiometric Upconversion Luminescence Bioimaging of Cyanide Anions. J Am Chem Soc 133: 15276–15279.
  62. 62. Zhao Q, Li F, Huang C (2010) Phosphorescent chemosensors based on heavy-metal complexes. Chem Soc Rev 39: 3007–3030.
  63. 63. Schäferling M (2012) The Art of Fluorescence Imaging with Chemical Sensors. Angew Chem Int Ed Engl 51: 3532–3554.
  64. 64. Ho M-L, Hwang F-M, Chen P-N, Hu Y-H, Cheng Y-M, et al. (2006) Design and synthesis of iridium(iii) azacrown complex: application as a highly sensitive metal cation phosphorescence sensor. Org Biomol Chem 4: 98–103.
  65. 65. Wu Y, Jing H, Dong Z, Zhao Q, Wu H, et al. (2011) Ratiometric Phosphorescence Imaging of Hg(II) in Living Cells Based on a Neutral Iridium(III) Complex. Inorg Chem 50: 7412–7420.
  66. 66. Ntziachristos V, Bremer C, Weissleder R (2003) Fluorescence imaging with near-infrared light: new technological advances that enable in vivo molecular imaging. Eur Radiol 13: 195–208.
  67. 67. Ulbricht C, Beyer B, Friebe C, Winter A, Schubert US (2009) Recent Developments in the Application of Phosphorescent Iridium(III) Complex Systems. Adv Mater 21: 4418–4441.
  68. 68. Lo KK-W, Chung C-K, Zhu N (2006) Nucleic Acid Intercalators and Avidin Probes Derived from Luminescent Cyclometalated Iridium(III)–Dipyridoquinoxaline and –Dipyridophenazine Complexes. Chem Eur J 12: 1500–1512.
  69. 69. Lo KK-W, Zhang KY, Chung C-K, Kwok KY (2007) Synthesis, Photophysical and Electrochemical Properties, and Protein-Binding Studies of Luminescent Cyclometalated Iridium(III) Bipyridine Estradiol Conjugates. Chem Eur J 13: 7110–7120.
  70. 70. Zhang KY, Li SP-Y, Zhu N, Or IW-S, Cheung MS-H, et al. (2010) Structure, Photophysical and Electrochemical Properties, Biomolecular Interactions, and Intracellular Uptake of Luminescent Cyclometalated Iridium(III) Dipyridoquinoxaline Complexes. Inorg Chem 49: 2530–2540.
  71. 71. Wu H, Yang T, Zhao Q, Zhou J, Li C, et al. (2011) A cyclometalated iridium(iii) complex with enhanced phosphorescence emission in the solid state (EPESS): synthesis, characterization and its application in bioimaging. Dalton Trans 40: 1969–1976.
  72. 72. Tan W, Zhou J, Li F, Yi T, Tian H (2011) Visible Light-Triggered Photoswitchable Diarylethene-Based Iridium(III) Complexes for Imaging Living Cells. Chem Asian J 6: 1263–1268.
  73. 73. Zhang S-J, Hosaka M, Yoshihara T, Negishi K, Iida Y, et al. (2010) Phosphorescent Light-Emitting Iridium Complexes Serve as a Hypoxia-Sensing Probe for Tumor Imaging in Living Animals. Cancer Res 70: 4490–4498.
  74. 74. Ma D-L, Wong W-L, Chung W-H, Chan F-Y, So P-K, et al. (2008) A Highly Selective Luminescent Switch-On Probe for Histidine/Histidine-Rich Proteins and Its Application in Protein Staining. Angew Chem Int Ed Engl 47: 3735–3739.
  75. 75. Li C, Yu M, Sun Y, Wu Y, Huang C, et al. (2011) A Nonemissive Iridium(III) Complex That Specifically Lights-Up the Nuclei of Living Cells. J Am Chem Soc 133: 11231–11239.
  76. 76. Man BY-W, Chan H-M, Leung C-H, Chan DS-H, Bai L-P, et al. (2011) Group 9 metal-based inhibitors of [small beta]-amyloid (1–40) fibrillation as potential therapeutic agents for Alzheimer's disease. Chem Sci 2: 917–921.
  77. 77. Lowry MS, Hudson WR, Pascal RA Jr, Bernhard S (2004) Accelerated luminophor discovery through combinatorial synthesis. J Am Chem Soc 126: 14129–14135.
  78. 78. Kwon T-H, Oh YH, Shin I-S, Hong J-I (2009) New approach toward fast response light-emitting electrochemical cells based on neutral iridium complexes via cation transport. Adv Funct Mater 19: 711–717.
  79. 79. McDaniel ND, Coughlin FJ, Tinker LL, Bernhard S (2008) Cyclometalated Iridium(III) Aquo Complexes: Efficient and Tunable Catalysts for the Homogeneous Oxidation of Water. J Am Chem Soc 130: 210–217.