1932

Abstract

Physiological dynamics in living cells and tissues are crucial for maintenance and regulation of their normal activities and functionalities. Tiny fluctuations in physiological microenvironments can leverage significant influences on cell growth, metabolism, differentiation, and apoptosis as well as disease evolution. Fluorescence imaging based on aggregation-induced emission luminogens (AIEgens) exhibits superior advantages in real-time sensing and monitoring of the physiological dynamics in living systems, including its unique properties such as high sensitivity and rapid response, flexible molecular design, and versatile nano- to mesostructural fabrication. The introduction of canonic AIEgens with long-wavelength, near-infrared, or microwave emission, persistent luminescence, and diversified excitation source (e.g., chemo- or bioluminescence) offers researchers a tool to evaluate the resulting molecules with excellent performance in response to subtle fluctuations in bioactivities with broader dimensionalities and deeper hierarchies.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-anchem-090420-101149
2021-07-27
2024-04-23
Loading full text...

Full text loading...

/deliver/fulltext/anchem/14/1/annurev-anchem-090420-101149.html?itemId=/content/journals/10.1146/annurev-anchem-090420-101149&mimeType=html&fmt=ahah

Literature Cited

  1. 1. 
    Vaupel P, Schlenger K, Knoop C, Höckel M. 1991. Oxygenation of human tumors: evaluation of tissue oxygen distribution in breast cancers by computerized O2 tension measurements. Cancer Res 51:3316–22
    [Google Scholar]
  2. 2. 
    Armstrong JS, Steinauer KK, Hornung B, Irish JM, Lecane P et al. 2002. Role of glutathione depletion and reactive oxygen species generation in apoptotic signaling in a human B lymphoma cell line. Cell Death Differ 9:252–63
    [Google Scholar]
  3. 3. 
    D'Autréaux B, Toledano MB 2007. ROS as signalling molecules: mechanisms that generate specificity in ROS homeostasis. Nat. Rev. Mol. Cell Biol. 8:813–24
    [Google Scholar]
  4. 4. 
    Lusic H, Grinstaff MW. 2012. X-ray-computed tomography contrast agents. Chem. Rev. 113:1641–66
    [Google Scholar]
  5. 5. 
    Fan W, Yung B, Huang P, Chen X. 2017. Nanotechnology for multimodal synergistic cancer therapy. Chem. Rev. 117:13566–638
    [Google Scholar]
  6. 6. 
    He X, Ma N. 2014. An overview of recent advances in quantum dots for biomedical applications. Colloid. Surf. B Biointerfaces 124:118–31
    [Google Scholar]
  7. 7. 
    He X, Xiong L-H, Zhao Z, Wang Z, Luo L et al. 2019. AIE-based theranostic systems for detection and killing of pathogens. Theranostics 9:3223–48
    [Google Scholar]
  8. 8. 
    Liu H, Xiong L-H, Kwok RTK, He X, Lam JWY et al. 2020. AIE bioconjugates for biomedical applications. Adv. Opt. Mater. 8:2000162
    [Google Scholar]
  9. 9. 
    Kobayashi H, Ogawa M, Alford R, Choyke PL, Urano Y. 2009. New strategies for fluorescent probe design in medical diagnostic imaging. Chem. Rev. 110:2620–40
    [Google Scholar]
  10. 10. 
    He X, Zeng T, Li Z, Wang G, Ma N 2016. Catalytic molecular imaging of microRNA in living cells by DNA-programmed nanoparticle disassembly. Angew. Chem. Int. Ed. 55:3073–76
    [Google Scholar]
  11. 11. 
    Wei W, He X, Ma N. 2014. DNA-templated assembly of a heterobivalent quantum dot nanoprobe for extra- and intracellular dual-targeting and imaging of live cancer cells. Angew. Chem. Int. Ed. 53:5573–77
    [Google Scholar]
  12. 12. 
    Li Z, He X, Luo X, Wang L, Ma N 2016. DNA-programmed quantum dot polymerization for ultrasensitive molecular imaging of cancer cells. Anal. Chem. 88:9355–58
    [Google Scholar]
  13. 13. 
    Yong K-T, Law W-C, Hu R, Ye L, Liu L et al. 2013. Nanotoxicity assessment of quantum dots: from cellular to primate studies. Chem. Soc. Rev. 42:1236–50
    [Google Scholar]
  14. 14. 
    He X, Li Z, Chen M, Ma N 2014. DNA-programmed dynamic assembly of quantum dots for molecular computation. Angew. Chem. Int. Ed. 53:14447–50
    [Google Scholar]
  15. 15. 
    Mei J, Leung NLC, Kwok RTK, Lam JWY, Tang BZ. 2015. Aggregation-induced emission: together we shine, united we soar!. Chem. Rev. 115:11718–940
    [Google Scholar]
  16. 16. 
    Kwok RTK, Leung CWT, Lam JWY, Tang BZ. 2015. Biosensing by luminogens with aggregation-induced emission characteristics. Chem. Soc. Rev. 44:4228–38
    [Google Scholar]
  17. 17. 
    Gao Y, He Z, He X, Zhang H, Weng J et al. 2019. Dual-color emissive AIEgen for specific and label-free double-stranded DNA recognition and single-nucleotide polymorphisms detection. J. Am. Chem. Soc. 141:20097–106
    [Google Scholar]
  18. 18. 
    Xiong L-H, He X, Zhao Z, Kwok RTK, Xiong Y et al. 2018. Ultrasensitive virion immunoassay platform with dual-modality based on a multifunctional aggregation-induced emission luminogen. ACS Nano 12:9549–57
    [Google Scholar]
  19. 19. 
    Niu G, Zheng X, Zhao Z, Zhang H, Wang J et al. 2019. AIE-active functionalized acrylonitriles: structure tuning by simple reaction-condition variation, efficient red emission and two-photon bioimaging. J. Am. Chem. Soc. 141:15111–20
    [Google Scholar]
  20. 20. 
    He X, Zhao Z, Xiong L-H, Gao PF, Peng C et al. 2018. Redox-active AIEgen-derived plasmonic and fluorescent core@shell nanoparticles for multimodality bioimaging. J. Am. Chem. Soc. 140:6904–11
    [Google Scholar]
  21. 21. 
    Qin W, Alifu N, Cai Y, Lam JWY, He X et al. 2019. Synthesis of an efficient far-red/near-infrared luminogen with AIE characteristics for in vivo bioimaging applications. Chem. Commun. 55:5615–18
    [Google Scholar]
  22. 22. 
    He X, Xiong L-H, Huang Y, Zhao Z, Wang Z et al. 2020. AIE-based energy transfer systems for biosensing, imaging, and therapeutics. TrAC Trends Anal. Chem. 122:115743
    [Google Scholar]
  23. 23. 
    Wang Y, Zhang Y, Wang J, Liang X-J. 2019. Aggregation-induced emission (AIE) fluorophores as imaging tools to trace the biological fate of nano-based drug delivery systems. Adv. Drug Deliv. Rev. 143:161–76
    [Google Scholar]
  24. 24. 
    He X, Yin F, Wang D, Xiong L-H, Kwok RTK et al. 2019. AIE featured inorganic-organic core@shell nanoparticle for high-efficiency siRNA delivery and real-time monitoring. Nano Lett 19:2272–79
    [Google Scholar]
  25. 25. 
    Cheng Y, Sun C, Liu R, Yang J, Dai J et al. 2019. A multifunctional peptide-conjugated AIEgen for efficient and sequential targeted gene delivery into the nucleus. Angew. Chem. Int. Ed. 58:5049–53
    [Google Scholar]
  26. 26. 
    He X, Yang Y, Guo Y, Lu S, Du Y et al. 2020. Phage-guided targeting, discriminative imaging, and synergistic killing of bacteria by AIE bioconjugates. J. Am. Chem. Soc. 142:3959–69
    [Google Scholar]
  27. 27. 
    Gao Y, Wang X, He X, He Z, Yang X et al. 2019. A dual-functional photosensitizer for ultraefficient photodynamic therapy and synchronous anticancer efficacy monitoring. Adv. Funct. Mater. 29:1902673
    [Google Scholar]
  28. 28. 
    He X, Peng C, Qiang S, Xiong L-H, Zhao Z et al. 2020. Less is more: silver-AIE core@shell nanoparticles for multimodality cancer imaging and synergistic therapy. Biomaterials 238:119834
    [Google Scholar]
  29. 29. 
    Casey JR, Grinstein S, Orlowski J. 2010. Sensors and regulators of intracellular pH. Nat. Rev. Mol. Cell Biol. 11:50–61
    [Google Scholar]
  30. 30. 
    Srivastava J, Barber DL, Jacobson MP. 2007. Intracellular pH sensors: design principles and functional significance. Physiology 22:30–39
    [Google Scholar]
  31. 31. 
    Chen S, Hong Y, Liu Y, Liu J, Leung CWT et al. 2013. Full-range intracellular pH sensing by an aggregation-induced emission-active two-channel ratiometric fluorogen. J. Am. Chem. Soc. 135:4926–29
    [Google Scholar]
  32. 32. 
    Chao J, Wang H, Zhang Y, Yin C, Huo F et al. 2018. A novel pyrene-based dual multifunctional fluorescent probe for differential sensing of pH and HSO3 and their bioimaging in live cells. New J. Chem. 42:3322–33
    [Google Scholar]
  33. 33. 
    Yin H, Zhao B, Kan W, Ding L, Wang L et al. 2020. A phenanthro[9, 10-d]imidazole-based optical sensor for dual-responsive turn-on detection of acidic pH and Cu2+ in chicken blood and living cells. Dyes Pigm 173:107916
    [Google Scholar]
  34. 34. 
    Song P, Chen X, Xiang Y, Huang L, Zhou Z et al. 2011. A ratiometric fluorescent pH probe based on aggregation-induced emission enhancement and its application in live-cell imaging. J. Mater. Chem. 21:13470–75
    [Google Scholar]
  35. 35. 
    Bai Y, Liu D, Han Z, Chen Y, Chen Z et al. 2018. BODIPY-derived ratiometric fluorescent sensors: pH-regulated aggregation-induced emission and imaging application in cellular acidification triggered by crystalline silica exposure. Sci. China Chem. 61:1413–22
    [Google Scholar]
  36. 36. 
    Yu Y, Yu C, Wu Q, Wang H, Jiao L et al. 2019. Pure E/Z isomers of N-methylpyrrole-benzohydrazide-based BF2 complexes: remarkable aggregation-, crystallization-induced emission switching properties and application in sensing intracellular pH microenvironment. J. Mater. Chem. C 7:4533–42
    [Google Scholar]
  37. 37. 
    Dou Y, Kenry Liu J, Zhang F, Cai C et al. 2019. 2-Styrylquinoline-based two-photon AIEgens for dual monitoring of pH and viscosity in living cells. J. Mater. Chem. B 7:7771–75
    [Google Scholar]
  38. 38. 
    Li K, Feng Q, Niu G, Zhang W, Li Y et al. 2018. Benzothiazole-based AIEgen with tunable excited-state intramolecular proton transfer and restricted intramolecular rotation processes for highly sensitive physiological pH sensing. ACS Sens 3:920–28
    [Google Scholar]
  39. 39. 
    Zhao X, Chen Y, Niu G, Gu D, Wang J et al. 2019. Photostable pH-sensitive near-infrared aggregation-induced emission luminogen for long-term mitochondrial tracking. ACS Appl. Mater. Interfaces 11:13134–39
    [Google Scholar]
  40. 40. 
    Lin N, Chen X, Yan S, Wang H, Lu Z et al. 2016. An aggregation-induced emission-based pH-sensitive fluorescent probe for intracellular acidity sensing. RSC Adv 6:25416–19
    [Google Scholar]
  41. 41. 
    Shi X, Yan N, Niu G, Sung SHP, Liu Z et al. 2020. In vivo monitoring of tissue regeneration using a ratiometric lysosomal AIE probe. Chem. Sci. 11:3152–63
    [Google Scholar]
  42. 42. 
    Qiu J, Jiang S, Guo H, Yang F 2018. An AIE and FRET-based BODIPY sensor with large stoke shift: novel pH probe exhibiting application in CO32− detection and living cell imaging. Dyes Pigm 157:351–58
    [Google Scholar]
  43. 43. 
    Fang M, Xia S, Bi J, Wigstrom TP, Valenzano L et al. 2018. A cyanine-based fluorescent cassette with aggregation-induced emission for sensitive detection of pH changes in live cells. Chem. Commun. 54:1133–36
    [Google Scholar]
  44. 44. 
    Wang J, Xia S, Bi J, Fang M, Mazi W et al. 2018. Ratiometric near-infrared fluorescent probes based on through-bond energy transfer and π-conjugation modulation between tetraphenylethene and hemicyanine moieties for sensitive detection of pH changes in live cells. Bioconjug. Chem. 29:1406–18
    [Google Scholar]
  45. 45. 
    Qi Q, Li Y, Yan X, Zhang F, Jiang S et al. 2016. Intracellular pH sensing using polymeric micelle containing tetraphenylethylene-oxazolidine. Polym. Chem. 7:5273–80
    [Google Scholar]
  46. 46. 
    Hu Y, Han T, Yan N, Liu J, Liu X et al. 2019. Visualization of biogenic amines and in vivo ratiometric mapping of intestinal pH by AIE-active polyheterocycles synthesized by metal-free multicomponent polymerizations. Adv. Funct. Mater. 29:1902240
    [Google Scholar]
  47. 47. 
    Lowell BB, Spiegelman BM. 2000. Towards a molecular understanding of adaptive thermogenesis. Nature 404:652–60
    [Google Scholar]
  48. 48. 
    Gao H, Kam C, Chou TY, Wu M-Y, Zhao X et al. 2020. A simple yet effective AIE-based fluorescent nano-thermometer for temperature mapping in living cells using fluorescence lifetime imaging microscopy. Nanoscale Horiz 5:488–94
    [Google Scholar]
  49. 49. 
    Wang Z, He X, Yong T, Miao Y, Zhang C et al. 2020. Multicolor tunable polymeric nanoparticle from the tetraphenylethylene cage for temperature sensing in living cells. J. Am. Chem. Soc. 142:512–19
    [Google Scholar]
  50. 50. 
    Luby-Phelps K. 1999. Cytoarchitecture and physical properties of cytoplasm: volume, viscosity, diffusion, intracellular surface area. Int. Rev. Cytol 192:189–221
    [Google Scholar]
  51. 51. 
    Mecocci P, Beal MF, Cecchetti R, Polidori MC, Cherubini A et al. 1997. Mitochondrial membrane fluidity and oxidative damage to mitochondrial DNA in aged and AD human brain. Mol. Chem. Neuropathol. 31:53–64
    [Google Scholar]
  52. 52. 
    Chen S, Hong Y, Zeng Y, Sun Q, Liu Y et al. 2015. Mapping live cell viscosity with an aggregation-induced emission fluorogen by means of two-photon fluorescence lifetime imaging. Chem. Eur. J. 21:4315–20
    [Google Scholar]
  53. 53. 
    Chen W, Gao C, Liu X, Liu F, Wang F et al. 2018. Engineering organelle-specific molecular viscosimeters using aggregation-induced emission luminogens for live cell imaging. Anal. Chem. 90:8736–41
    [Google Scholar]
  54. 54. 
    Wodarz A, Näthke I. 2007. Cell polarity in development and cancer. Nat. Cell Biol. 9:1016–24
    [Google Scholar]
  55. 55. 
    Owyong TC, Subedi P, Deng J, Hinde E, Paxman JJ et al. 2020. A molecular chameleon for mapping subcellular polarity in an unfolded proteome environment. Angew. Chem. Int. Ed. 59:10129–35
    [Google Scholar]
  56. 56. 
    Gandre-Babbe S, van der Bliek AM. 2008. The novel tail-anchored membrane protein Mff controls mitochondrial and peroxisomal fission in mammalian cells. Mol. Biol. Cell 19:2402–12
    [Google Scholar]
  57. 57. 
    Ow Y-LP, Green DR, Hao Z, Mak TW. 2008. Cytochrome c: functions beyond respiration. Nat. Rev. Mol. Cell Biol. 9:532–42
    [Google Scholar]
  58. 58. 
    Li J, Kwon N, Jeong Y, Lee S, Kim G et al. 2017. Aggregation-induced fluorescence probe for monitoring membrane potential changes in mitochondria. ACS Appl. Mater. Interfaces 10:12150–54
    [Google Scholar]
  59. 59. 
    Zhao N, Chen S, Hong Y, Tang BZ. 2015. A red emitting mitochondria-targeted AIE probe as an indicator for membrane potential and mouse sperm activity. Chem. Commun. 51:13599–602
    [Google Scholar]
  60. 60. 
    Zhang L, Liu W, Huang X, Zhang G, Wang X et al. 2015. Old is new again: a chemical probe for targeting mitochondria and monitoring mitochondrial membrane potential in cells. Analyst 140:5849–54
    [Google Scholar]
  61. 61. 
    Zeng Z, Ren X, Yin T, Gao X, Tsai M et al. 2019. Multiplexed detection and the establishment of a novel high-throughput method for human germ cell quality screening based on aggregation-induced emission. Am. J. Transl. Res. 11:6907
    [Google Scholar]
  62. 62. 
    Situ B, Chen S, Zhao E, Leung CWT, Chen Y et al. 2016. Real-time imaging of cell behaviors in living organisms by a mitochondria-targeting AIE fluorogen. Adv. Funct. Mater. 26:7132–38
    [Google Scholar]
  63. 63. 
    Xue T, Jia X, Wang J, Xiang J, Wang W et al. 2019.. “ Turn-on” activatable AIE dots for tumor hypoxia imaging. Chem. Eur. J. 25:9634–38
    [Google Scholar]
  64. 64. 
    Xue T, Shao K, Xiang J, Pan X, Zhu Z et al. 2020. In situ construction of a self-assembled AIE probe for tumor hypoxia imaging. Nanoscale 12:7509–13
    [Google Scholar]
  65. 65. 
    Xu C, Zou H, Zhao Z, Zhang P, Kwok RT et al. 2019. A new strategy toward “simple” water-soluble AIE probes for hypoxia detection. Adv. Funct. Mater. 29:1903278
    [Google Scholar]
  66. 66. 
    You X, Li L, Li X, Ma H, Zhang G et al. 2016. A new tetraphenylethylene-derived fluorescent probe for nitroreductase detection and hypoxic-tumor-cell imaging. Chem. Asian J. 11:2918–23
    [Google Scholar]
  67. 67. 
    Lin Y, Sun L, Zeng F, Wu S. 2019. An unsymmetrical squaraine-based activatable probe for imaging lymphatic metastasis by responding to tumor hypoxia with MSOT and aggregation-enhanced fluorescent imaging. Chem. Eur. J. 25:16740–47
    [Google Scholar]
  68. 68. 
    Weerapana E, Wang C, Simon GM, Richter F, Khare S et al. 2010. Quantitative reactivity profiling predicts functional cysteines in proteomes. Nature 468:790–95
    [Google Scholar]
  69. 69. 
    Liu Y, Yu Y, Lam JW, Hong Y, Faisal M et al. 2010. Simple biosensor with high selectivity and sensitivity: thiol-specific biomolecular probing and intracellular imaging by AIE fluorogen on a TLC plate through a thiol–ene click mechanism. Chem. Eur. J. 16:8433–38
    [Google Scholar]
  70. 70. 
    Zhou X, Guo D, Jiang Y, Gong D-M, Zhao X-J et al. 2017. A novel AIEE and ESIPT fluorescent probe for selective detection of cysteine. Tetrahedron Lett 58:3214–18
    [Google Scholar]
  71. 71. 
    Cui L, Baek Y, Lee S, Kwon N, Yoon J. 2016. An AIE and ESIPT based kinetically resolved fluorescent probe for biothiols. J. Mater. Chem. C 4:2909–14
    [Google Scholar]
  72. 72. 
    Sheng H, Hu Y, Zhou Y, Fan S, Cao Y et al. 2018. A hydroxyphenylquinazolinone-based fluorescent probe for turn-on detection of cysteine with a large Stokes shift and its application in living cells. J. Photochem. Photobiol. A 364:750–57
    [Google Scholar]
  73. 73. 
    Chu Y, Xie Z, Yue Y, Yue Y, Kong X et al. 2019. New fast, highly selective probe with both aggregation-induced emission enhancement and intramolecular charge-transfer characteristics for homocysteine detection. ACS Omega 4:5367–73
    [Google Scholar]
  74. 74. 
    Chu Y, Xie Z, Zhuang D, Yue Y, Yue Y et al. 2019. An intramolecular charge transfer and aggregation induced emission enhancement fluorescent probe based on 2-phenyl-1,2,3-triazole for highly selective and sensitive detection of homocysteine and its application in living cells. Chin. J. Chem. 37:1216–22
    [Google Scholar]
  75. 75. 
    Townsend DM, Tew KD, Tapiero H. 2003. The importance of glutathione in human disease. Biomed. Pharmacother. 57:145–55
    [Google Scholar]
  76. 76. 
    Lou X, Hong Y, Chen S, Leung CWT, Zhao N et al. 2014. A selective glutathione probe based on AIE fluorogen and its application in enzymatic activity assay. Sci. Rep. 4:4272
    [Google Scholar]
  77. 77. 
    Wang B, Li C, Yang L, Zhang C, Liu L-J et al. 2019. Tetraphenylethene decorated with disulfide-functionalized hyperbranched poly (amido amine)s as metal/organic solvent-free turn-on AIE probes for biothiol determination. J. Mater. Chem. B 7:3846–55
    [Google Scholar]
  78. 78. 
    Han W, Zhang S, Qian J, Zhang J, Wang X et al. 2019. Redox-responsive fluorescent nanoparticles based on diselenide-containing AIEgens for cell imaging and selective cancer therapy. Chem. Asian J. 14:1745–53
    [Google Scholar]
  79. 79. 
    Reiffenstein R, Hulbert WC, Roth SH. 1992. Toxicology of hydrogen sulfide. Annu. Rev. Pharmacol. Toxicol. 32:109–34
    [Google Scholar]
  80. 80. 
    Luo Y, Zhu C, Du D, Lin Y. 2019. A review of optical probes based on nanomaterials for the detection of hydrogen sulfide in biosystems. Anal. Chim. Acta 106:1–12
    [Google Scholar]
  81. 81. 
    Chen M, Chen R, Shi Y, Wang J, Cheng Y et al. 2018. Malonitrile-functionalized tetraphenylpyrazine: aggregation-induced emission, ratiometric detection of hydrogen sulfide, and mechanochromism. Adv. Funct. Mater. 28:1704689
    [Google Scholar]
  82. 82. 
    Pramanik S, Bhalla V, Kim HM, Singh H, Lee HW et al. 2015. A hexaphenylbenzene based AIEE active two photon probe for the detection of hydrogen sulfide with tunable self-assembly in aqueous media and application in live cell imaging. Chem. Commun. 51:15570–3
    [Google Scholar]
  83. 83. 
    Zhao B, Yang B, Hu X, Liu B. 2018. Two colorimetric and ratiometric fluorescence probes for hydrogen sulfide based on AIE strategy of α–cyanostilbenes. Spectrochim. Acta A 199:117–22
    [Google Scholar]
  84. 84. 
    Ma Y, Wang H, Su S, Chen Y, Li Y et al. 2019. A red mitochondria-targeted AIEgen for visualizing H2S in living cells and tumours. Analyst 144:3381–88
    [Google Scholar]
  85. 85. 
    Zhang P, Hong Y, Wang H, Yu M, Gao Y et al. 2017. Selective visualization of endogenous hydrogen sulfide in lysosomes using aggregation induced emission dots. Polym. Chem. 8:7271–78
    [Google Scholar]
  86. 86. 
    Zhang P, Nie X, Gao M, Zeng F, Qin A et al. 2017. A highly selective fluorescent nanoprobe based on AIE and ESIPT for imaging hydrogen sulfide in live cells and zebrafish. Mater. Chem. Front. 1:838–45
    [Google Scholar]
  87. 87. 
    Kontos HA, Wei EP. 1986. Superoxide production in experimental brain injury. J. Neurosurg. 64:803–7
    [Google Scholar]
  88. 88. 
    Yang J, Liu X, Wang H, Tan H, Xie X et al. 2018. A turn-on near-infrared fluorescence probe with aggregation-induced emission based on dibenzo[a,c]phenazine for detection of superoxide anions and its application in cell imaging. Analyst 143:1242–49
    [Google Scholar]
  89. 89. 
    Niu J, Fan J, Wang X, Xiao Y, Xie X et al. 2017. Simultaneous fluorescence and chemiluminescence turned on by aggregation-induced emission for real-time monitoring of endogenous superoxide anion in live cells. Anal. Chem. 89:7210–15
    [Google Scholar]
  90. 90. 
    Alderton WK, Cooper CE, Knowles RG. 2001. Nitric oxide synthases: structure, function and inhibition. Biochem. J. 357:593–615
    [Google Scholar]
  91. 91. 
    Liu L, Zhang F, Xu B, Tian W. 2017. Silica nanoparticles based on an AIE-active molecule for ratiometric detection of RNS in vitro. J. Mater. Chem. B 5:9197–203
    [Google Scholar]
  92. 92. 
    Song Z, Mao D, Sung SH, Kwok RT, Lam JW et al. 2016. Activatable fluorescent nanoprobe with aggregation-induced emission characteristics for selective in vivo imaging of elevated peroxynitrite generation. Adv. Mater. 28:7249–56
    [Google Scholar]
  93. 93. 
    Wu W, Mao D, Cai X, Duan Y, Hu F et al. 2018. ONOO and ClO responsive organic nanoparticles for specific in vivo image-guided photodynamic bacterial ablation. Chem. Mater. 30:3867–73
    [Google Scholar]
  94. 94. 
    Davies MJ. 2010. Myeloperoxidase-derived oxidation: mechanisms of biological damage and its prevention. J. Clin. Biochem. Nutr. 48:8–19
    [Google Scholar]
  95. 95. 
    Yadav AK, Bracher A, Doran SF, Leustik M, Squadrito GL et al. 2010. Mechanisms and modification of chlorine-induced lung injury in animals. Proc. Am. Thorac. Soc. 7:278–83
    [Google Scholar]
  96. 96. 
    Wang L, Chen X, Xia Q, Liu R, Qu J. 2018. Deep-red AIE-active fluorophore for hypochlorite detection and bioimaging in live cells. Ind. Eng. Chem. Res. 57:7735–41
    [Google Scholar]
  97. 97. 
    Dong X, Zhang G, Shi J, Wang Y, Wang M et al. 2017. A highly selective fluorescence turn-on detection of ClO with 1-methyl-1, 2-dihydropyridine-2-thione unit modified tetraphenylethylene. Chem. Commun. 53:11654–57
    [Google Scholar]
  98. 98. 
    Gu J, Li X, Zhou Z, Liao R, Gao J et al. 2019. Synergistic regulation of effective detection for hypochlorite based on a dual-mode probe by employing aggregation induced emission (AIE) and intramolecular charge transfer (ICT) effects. Chem. Eng. J. 368:157–64
    [Google Scholar]
  99. 99. 
    Huang Y, Zhang P, Gao M, Zeng F, Qin A et al. 2016. Ratiometric detection and imaging of endogenous hypochlorite in live cells and in vivo achieved by using an aggregation induced emission (AIE)-based nanoprobe. Chem. Commun. 52:7288–91
    [Google Scholar]
  100. 100. 
    Yu H-X, Zhi J, Shen T, Ding W, Zhang X et al. 2019. Donor–acceptor type aggregation-induced emission luminophores based on the 1,1-dicyanomethylene-3-indanone unit for bridge-dependent reversible mechanochromism and light-up biosensing of hypochlorites. J. Mater. Chem. C 7:8888–97
    [Google Scholar]
  101. 101. 
    Zhu Y, Wang K, Wu X, Sun Y, Gong X et al. 2020. A highly sensitive turn-on fluorescent probe for real-time detecting hypochlorite and its application in living cells. Talanta 209:120548
    [Google Scholar]
  102. 102. 
    Xie Y, Yan L, Tang Y, Tang M, Wang S et al. 2019. A smart fluorescent probe based on salicylaldehyde Schiff's base with AIE and ESIPT characteristics for the detections of N2H4 and ClO. J. Fluoresc. 29:399–406
    [Google Scholar]
  103. 103. 
    Duan Q, Zheng G, Li Z, Cheng K, Zhang J et al. 2019. An ultra-sensitive ratiometric fluorescent probe for hypochlorous acid detection by the synergistic effect of AIE and TBET and its application of detecting exogenous/endogenous HOCl in living cells. J. Mater. Chem. B 7:5125–31
    [Google Scholar]
  104. 104. 
    Reuter S, Gupta SC, Chaturvedi MM, Aggarwal BB. 2010. Oxidative stress, inflammation, and cancer: How are they linked? Free Radic. . Biol. Med. 49:1603–16
    [Google Scholar]
  105. 105. 
    Zhang W, Liu W, Li P, Huang F, Wang H et al. 2015. Rapid-response fluorescent probe for hydrogen peroxide in living cells based on increased polarity of C–B bonds. Anal. Chem. 87:9825–28
    [Google Scholar]
  106. 106. 
    Selvaraj M, Rajalakshmi K, Nam Y-S, Lee Y, Kim BC et al. 2019. Rapid-response and highly sensitive boronate derivative-based fluorescence probe for detecting H2O2 in living cells. J. Anal. Methods Chem. 2019.5174764
    [Google Scholar]
  107. 107. 
    Cheng Y, Dai J, Sun C, Liu R, Zhai T et al. 2018. An intracellular H2O2-responsive AIEgen for the peroxidase-mediated selective imaging and inhibition of inflammatory cells. Angew. Chem. Int. Ed. 57:3123–27
    [Google Scholar]
  108. 108. 
    Mao D, Wu W, Ji S, Chen C, Hu F et al. 2017. Chemiluminescence-guided cancer therapy using a chemiexcited photosensitizer. Chemistry 3:991–1007
    [Google Scholar]
  109. 109. 
    Zhang Y, Yan C, Wang C, Guo Z, Liu X et al. 2020. A sequential dual-lock strategy for photoactivatable chemiluminescent probes enabling bright duplex optical imaging. Angew. Chem. Int. Ed. 59:9059–66
    [Google Scholar]
  110. 110. 
    Xiong Y, Zhao Z, Zhao W, Ma H, Peng Q et al. 2018. Designing efficient and ultralong pure organic room-temperature phosphorescent materials by structural isomerism. Angew. Chem. Int. Ed. 57:7997–8001
    [Google Scholar]
  111. 111. 
    Zhang X, Du L, Zhao W, Zhao Z, Xiong Y et al. 2019. Ultralong UV/mechano-excited room temperature phosphorescence from purely organic cluster excitons. Nat. Commun. 10:5161
    [Google Scholar]
  112. 112. 
    Wang N. 2017. Review of cellular mechanotransduction. J. Phys. D Appl. Phys. 50:233002
    [Google Scholar]
  113. 113. 
    Tajik A, Zhang Y, Wei F, Sun J, Jia Q et al. 2016. Transcription upregulation via force-induced direct stretching of chromatin. Nat. Mater. 15:1287–96
    [Google Scholar]
  114. 114. 
    Liu J, Li F, Wang Y, Pan L, Lin P et al. 2020. A sensitive and specific nanosensor for monitoring extracellular potassium levels in the brain. Nat. Nanotechnol. 15:321–30
    [Google Scholar]
  115. 115. 
    Liu J, Pan L, Shang C, Lu B, Wu R et al. 2020. A highly sensitive and selective nanosensor for near-infrared potassium imaging. Sci. Adv. 6:eaax9757
    [Google Scholar]
  116. 116. 
    Feng X, Li Y, He X, Liu H, Zhao Z et al. 2018. A substitution-dependent light-up fluorescence probe for selectively detecting Fe3+ ions and its cell imaging application. Adv. Funct. Mater. 28:1802833
    [Google Scholar]
  117. 117. 
    Gao M, Li Y, Chen X, Li S, Ren L et al. 2018. Aggregation-induced emission probe for light-up and in situ detection of calcium ions at high concentration. ACS Appl. Mater. Interfaces 10:14410–17
    [Google Scholar]
/content/journals/10.1146/annurev-anchem-090420-101149
Loading
/content/journals/10.1146/annurev-anchem-090420-101149
Loading

Data & Media loading...

Supplemental Material

Supplementary Data

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error