Engineered Luciferase Reporter from a Deep Sea Shrimp Utilizing a Novel Imidazopyrazinone SubstrateClick to copy article linkArticle link copied!
- Mary P. Hall
- James Unch
- Brock F. Binkowski
- Michael P. Valley
- Braeden L. Butler
- Monika G. Wood
- Paul Otto
- Kristopher Zimmerman
- Gediminas Vidugiris
- Thomas Machleidt
- Matthew B. Robers
- Hélène A. Benink
- Christopher T. Eggers
- Michael R. Slater
- Poncho L. Meisenheimer
- Dieter H. Klaubert
- Frank Fan
- Lance P. Encell
- Keith V. Wood
Abstract
Bioluminescence methodologies have been extraordinarily useful due to their high sensitivity, broad dynamic range, and operational simplicity. These capabilities have been realized largely through incremental adaptations of native enzymes and substrates, originating from luminous organisms of diverse evolutionary lineages. We engineered both an enzyme and substrate in combination to create a novel bioluminescence system capable of more efficient light emission with superior biochemical and physical characteristics. Using a small luciferase subunit (19 kDa) from the deep sea shrimp Oplophorus gracilirostris, we have improved luminescence expression in mammalian cells ∼2.5 million-fold by merging optimization of protein structure with development of a novel imidazopyrazinone substrate (furimazine). The new luciferase, NanoLuc, produces glow-type luminescence (signal half-life >2 h) with a specific activity ∼150-fold greater than that of either firefly (Photinus pyralis) or Renilla luciferases similarly configured for glow-type assays. In mammalian cells, NanoLuc shows no evidence of post-translational modifications or subcellular partitioning. The enzyme exhibits high physical stability, retaining activity with incubation up to 55 °C or in culture medium for >15 h at 37 °C. As a genetic reporter, NanoLuc may be configured for high sensitivity or for response dynamics by appending a degradation sequence to reduce intracellular accumulation. Appending a signal sequence allows NanoLuc to be exported to the culture medium, where reporter expression can be measured without cell lysis. Fusion onto other proteins allows luminescent assays of their metabolism or localization within cells. Reporter quantitation is achievable even at very low expression levels to facilitate more reliable coupling with endogenous cellular processes.
Results and Discussion
Prediction of Stabilizing Amino Acids
Structural Optimization and Screening of Novel Coelenterazine Substrates
luciferase | coelenterazine signal intensitya,b,c | furimazine signal intensitya,b,d | ||
---|---|---|---|---|
Oluc-N166R | 0.000089 ± 0.000007 | (3) | 0.0023 ± 0.0002 | (78) |
C1A4E | 2.6 ± 0.2 | (88,000) | 16 ± 1 | (540,000) |
Nluc | 2.4 ± 0.3 | (81,000) | 75 ± 9 | (2,500,000) |
Rluc | 0.51 ± 0.02 | (17,000) | 0.00045 ± 0.00003 | (15) |
N = 4.
Normalized to Fluc/ONE-Glo.
10 μM coelenterazine.
50 μM furimazine. Values normalized to Oluc-19/coelenterazine are shown in parentheses.
luciferase | enzyme stabilitya,b (37 °C; t1/2, min) | signal durationa,b (22 °C; t1/2, min) |
---|---|---|
Oluc-N166R | 5.1 ± 0.4 | NDc |
C1A4E | 330 ± 17(5.5 h) | 92 ± 5 |
Nluc | 11,000 ± 220(7.7 days) | 160 ± 18 |
Rluc | 99 ± 2(1.7 h) | 86 ± 5 |
Fluc | 7.3 ± 0.3 | 62 ± 5 |
N = 4.
Oluc-N166R, C1A4E, and Nluc measured using assay buffer/50 μM furimazine; Rluc measured using Renilla-Glo buffer/10 μM coelenterazine; Fluc measured using ONE-Glo.
ND = not determined.
NanoLuc Characterization and Comparison to Fluc and Rluc
Performance as a Genetic Reporter
Performance as a Fusion Reporter
Summary
Methods
Synthesis of Coelenterazine Analogues
Variant Enzyme Screening
NanoLuc Assay buffer
Supporting Information
This material is available free of charge via the Internet at http://pubs.acs.org
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgment
We thank M. Scurria, R. Arbit, H. Wang, L. Bernad, D. Simpson, R. Hurst, S. Saveliev, A. Niles, M. O’Brien, E. Strauss, J. Wilkinson, and T. Lubben for technical expertise and insightful discussions. We also thank G. Colwell at Gene Dynamics, LLC for help with vector constructions and J. Bujnicki at the IIMCB in Warsaw, Poland for assistance with fold-recognition analysis.
References
This article references 38 other publications.
- 1Widder, E. A. (2010) Bioluminescence in the ocean: origins of biological, chemical, and ecological diversity Science 328, 704– 708Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXlsFCitbg%253D&md5=c07748c8f13482b1ae8362cec0ffe5e1Bioluminescence in the ocean: Origins of biological, chemical, and ecological diversityWidder, E. A.Science (Washington, DC, United States) (2010), 328 (5979), 704-708CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)A review. From bacteria to fish, a remarkable variety of marine life depends on bioluminescence (the chem. generation of light) for finding food, attracting mates, and evading predators. Disparate biochem. systems and diverse phylogenetic distribution patterns of light-emitting organisms highlight the ecol. benefits of bioluminescence, with biochem. and genetic analyses providing new insights into the mechanisms of its evolution. The origins and functions of some bioluminescent systems, however, remain obscure. Here, the author reviews recent advances in understanding bioluminescence in the ocean and highlights future research efforts that will unite mol. details with ecol. and evolutionary relations.
- 2Melnick, J. S., Janes, J., Kim, S., Chang, J. Y., Sipes, D. G., Gunderson, D., Jarnes, L., Matzen, J. T., Garcia, M. E., Hood, T. L., Beigi, R., Xia, G., Harig, R. A., Asatryan, H., Yan, S. F., Zhou, Y., Gu, X. J., Saadat, A., Zhou, V., King, F. J., Shaw, C. M., Su, A. I., Downs, R., Gray, N. S., Schultz, P. G., Warmuth, M., and Caldwell, J. S. (2006) An efficient rapid system for profiling the cellular activities of molecular libraries Proc. Natl. Acad. Sci. U.S.A. 103, 3153– 3158Google ScholarThere is no corresponding record for this reference.
- 3Doshi, U. and Li, A. P. (2011) Luciferin IPA-based higher throughput human hepatocyte screening assays for CYP3A4 inhibition and induction J. Biomol. Screen. 16, 903– 909Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXht1yitrnM&md5=2533a5fda46939de0805661c6b61a090Luciferin IPA-based higher throughput human hepatocyte screening assays for CYP3A4 inhibition and inductionDoshi, Utkarsh; Li, Albert P.Journal of Biomolecular Screening (2011), 16 (8), 903-909CODEN: JBISF3; ISSN:1087-0571. (Sage Publications)The authors report here higher throughput screening (HTS) assays for the evaluation of CYP3A4 inhibition and CYP3A4 induction in human hepatocytes using a novel CYP3A4 substrate, luciferin IPA (LIPA). Using human recombinant CYP450 isoforms, LIPA was found to be metabolized extensively by CYP3A4 but not by CYP1A2, CYP2C9, CYP2C19, CYP2D6, or CYP2E1. In the 384-well plate CYP3A4 inhibition assay, the known inhibitors 1-aminobenzotriazole, erythromycin, ketoconazole, and verapamil were found to cause extensive (max. inhibition of >80%), dose-dependent, statistically significant inhibition of LIPA metab. The non-CYP3A4 inhibitors diethyldithiocarbamate, quercetin, quinidine, sulfaphenazole, ticlopidine, and tranylcypromine were found to have substantially lower (max. inhibition of <50%) or no apparent inhibitory effects in the HTS assay. In the 96-well plate induction assay, the CYP3A4 inducers rifampin, phenobarbital, carbamazepine, phenytoin, troglitazone, rosiglitazone, and pioglitazone yielded dose-dependent induction of LIPA metab., whereas the CYP1A2 inducers omeprazole and 3-methylcholanthrene did not display any induction in the CYP3A4 activity. The high sensitivity and specificity of the assays, the relative ease of execution, and reduced cost, time, and test material requirements suggest that the HTS assays may be applied routinely for screening a large no. of chems. in the drug discovery phase for CYP3A4 inhibitory and inducing potential.
- 4Smirnova, N. A., Haskew-Layton, R. E., Basso, M., Hushpulian, D. M., Payappilly, J. B., Speer, R. E., Ahn, Y. H., Rakhman, I., Cole, P. A., Pinto, J. T., Ratan, R. R., and Gazaryan, I. G. (2011) Development of Neh2-luciferase reporter and its application for high throughput screening and real-time monitoring of Nrf2 activators Chem. Biol. 18, 752– 765Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXotVejtbc%253D&md5=3919f582cce67396e5b869ce7b9e0325Development of Neh2-Luciferase Reporter and Its Application for High Throughput Screening and Real-Time Monitoring of Nrf2 ActivatorsSmirnova, Natalya A.; Haskew-Layton, Renee E.; Basso, Manuela; Hushpulian, Dmitry M.; Payappilly, Jimmy B.; Speer, Rachel E.; Ahn, Young-Hoon; Rakhman, Ilay; Cole, Philip A.; Pinto, John T.; Ratan, Rajiv R.; Gazaryan, Irina G.Chemistry & Biology (Cambridge, MA, United States) (2011), 18 (6), 752-765CODEN: CBOLE2; ISSN:1074-5521. (Cell Press)Summary: The NF-E2-related factor 2 (Nrf2) is a key transcriptional regulator of antioxidant defense and detoxification. To directly monitor stabilization of Nrf2, we fused its Neh2 domain, responsible for the interaction with its nucleocytoplasmic regulator, Keap1, to firefly luciferase (Neh2-luciferase). We show that Neh2 domain is sufficient for recognition, ubiquitination, and proteasomal degrdn. of Neh2-luciferase fusion protein. The Neh2-luc reporter system allows direct monitoring of the adaptive response to redox stress and classification of drugs based on the time course of reporter activation. The reporter was used to screen the Spectrum library of 2000 biol. active compds. to identify activators of Nrf2. The most robust and yet nontoxic Nrf2 activators found-nordihydroguaiaretic acid, fisetin, and gedunin-induced astrocyte-dependent neuroprotection from oxidative stress via an Nrf2-dependent mechanism.
- 5Perroy, J., Pontier, S., Charest, P. G., Aubry, M., and Bouvier, M. (2004) Real-time monitoring of ubiquitination in living cells by BRET Nat. Methods 1, 203– 208Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXisVGhs74%253D&md5=ae20928d2b7c8208099f526431f89154Real-time monitoring of ubiquitination in living cells by BRETPerroy, Julie; Pontier, Stephanie; Charest, Pascale G.; Aubry, Muriel; Bouvier, MichelNature Methods (2004), 1 (3), 203-208CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)Ubiquitin has emerged as an important regulator of protein stability and function in organisms ranging from yeast to mammals. The ability to detect in situ changes in protein ubiquitination without perturbing the physiol. environment of cells would be a major step forward in understanding the ubiquitination process and its consequences. Here, we describe a new method to study this dynamic post-translational modification in intact human embryonic kidney cells. Using bioLuminescence resonance energy transfer (BRET), we measured the ubiquitination of β-arrestin 2, a regulatory protein implicated in the modulation of G protein-coupled receptors. In addn. to allowing the detection of basal and GPCR-regulated ubiquitination of p-arrestin 2 in living cells, real-time BRET measurements permitted the recording of distinct ubiquitination kinetics that are dictated by the identity of the activated receptor. The ubiquitination BRET assay should prove to be a useful tool for studying the dynamic ubiquitination of proteins and for understanding which cellular functions are regulated by this post-translational event.
- 6Fan, F., Binkowski, B. F., Butler, B. L., Stecha, P. F., Lewis, M. K., and Wood, K. V. (2008) Novel genetically encoded biosensors using firefly luciferase ACS Chem. Biol. 3, 346– 351Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXntlKrsrY%253D&md5=59474ab5133cadb6752eed96e89fe96cNovel Genetically Encoded Biosensors Using Firefly LuciferaseFan, Frank; Binkowski, Brock F.; Butler, Braeden L.; Stecha, Peter F.; Lewis, Martin K.; Wood, Keith V.ACS Chemical Biology (2008), 3 (6), 346-351CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)Genetically encoded biosensors have proven valuable for real-time monitoring of intracellular phenomena, particularly FRET-based sensors incorporating variants of green fluorescent protein. To increase detection sensitivity and response dynamics, the authors genetically engineered firefly luciferase to detect specific intermol. interactions through modulation of its luminescence activity. This concept has been applied in covalent, noncovalent, and allosteric design configurations. The covalent design gives sensitive detection of protease activity through a cleavage-dependent increase in luminescence. The noncovalent and allosteric designs allow reversible detection of the small mols. rapamycin and cAMP, resp. These sensors allow detection of mol. processes within living cells following addn. of the luciferin substrate to the growth medium. For example, the cAMP sensor allows monitoring of intracellular signal transduction assocd. with G-protein coupled receptor function. These and other luminescent biosensors will be useful for the sensitive detection of cellular physiol. in research and drug discovery.
- 7Remy, I. and Michnick, S. W. (2006) A highly sensitive protein-protein interaction assay based on Gaussia luciferase Nat. Methods 3, 977– 979Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xht1Wqu77I&md5=6d80a3eded2f378506dff827c5af3e04A highly sensitive protein-protein interaction assay based on Gaussia luciferaseRemy, Ingrid; Michnick, Stephen W.Nature Methods (2006), 3 (12), 977-979CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)Protein-fragment complementation assays (PCAs) provide a general strategy to study the dynamics of protein-protein interactions in vivo and in vitro. The full potential of PCA requires assays that are fully reversible and sensitive at subendogenous protein expression levels. We describe a new assay that meets these criteria, based on the Gaussia princeps luciferase enzyme, demonstrating chem. reversal, and induction and inhibition of a key interaction linking insulin and TGFβ signaling.
- 8Tannous, B. A., Kim, D. E., Fernandez, J. L., Weissleder, R., and Breakefield, X. O. (2005) Codon-optimized Gaussia luciferase cDNA for mammalian gene expression in culture and in vivo Mol. Ther. 11, 435– 443Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXhsVOju7w%253D&md5=bc4ba7b78ea19d52184ebf6415cc410bCodon-optimized Gaussia luciferase cDNA for mammalian gene expression in culture and in vivoTannous, Bakhos A.; Kim, Dong-Eog; Fernandez, Juliet L.; Weissleder, Ralph; Breakefield, Xandra O.Molecular Therapy (2005), 11 (3), 435-443CODEN: MTOHCK; ISSN:1525-0016. (Elsevier)Photoproteins have played a major role in advancing our understanding of biol. processes. A broader array of biocompatible, nontoxic, and novel reporters can serve to expand this potential. Here we describe the properties of a luciferase from the copepod marine organism Gaussia princeps. It is a monomeric protein composed of 185 aa (19.9 kDa) with a short coding sequence (555 bp) making it suitable for viral vectors. The humanized form of Gaussia luciferase (hGLuc) was efficiently expressed in mammalian cells following delivery by HSV-1 amplicon vectors. It was found to be nontoxic and naturally secreted, with flash bioluminescence characteristics similar to those of other coelenterazine luciferases. HGLuc generated over 1000-fold higher bioluminescent signal intensity from live cells together with their immediate environment and over 100-fold higher intensity from viable cells alone (not including secreted luciferase) or cell lysates, compared to humanized forms of firefly (hFLuc) and Renilla (hRLuc) luciferases expressed under similar conditions. Furthermore, hGLuc showed 200-fold higher signal intensity than hRLuc and intensity comparable to that of hFLuc in vivo under std. imaging conditions. Gaussia luciferase provides a sensitive means of imaging gene delivery and other events in living cells in culture and in vivo, with a unique combination of features including high signal intensity, secretion, and ATP independence, thus being able to report from the cells and their environment in real time.
- 9Markova, S. V., Golz, S., Frank, L. A., Kalthof, B., and Vysotski, E. S. (2004) Cloning and expression of cDNA for a luciferase from the marine copepod Metridia longa. A novel secreted bioluminescent reporter enzyme J. Biol. Chem. 279, 3212– 3217Google ScholarThere is no corresponding record for this reference.
- 10Nakajima, Y., Kobayashi, K., Yamagishi, K., Enomoto, T., and Ohmiya, Y. (2004) cDNA cloning and characterization of a secreted luciferase from the luminous Japanese ostracod, Cypridina noctiluca Biosci. Biotechnol. Biochem. 68, 565– 570Google ScholarThere is no corresponding record for this reference.
- 11Suzuki, C., Nakajima, Y., Akimoto, H., Wu, C., and Ohmiya, Y. (2005) A new additional reporter enzyme, dinoflagellate luciferase, for monitoring of gene expression in mammalian cells Gene 344, 61– 66Google ScholarThere is no corresponding record for this reference.
- 12Inouye, S., Watanabe, K., Nakamura, H., and Shimomura, O. (2000) Secretional luciferase of the luminous shrimp Oplophorus gracilirostris: cDNA cloning of a novel imidazopyrazinone luciferase FEBS Lett. 481, 19– 25Google ScholarThere is no corresponding record for this reference.
- 13Wurdinger, T., Badr, C., Pike, L., de Kleine, R., Weissleder, R., Breakefield, X. O., and Tannous, B. A. (2008) A secreted luciferase for ex vivo monitoring of in vivo processes Nat. Methods 5, 171– 173Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhsV2ksLw%253D&md5=e0cc4226e91256ae0e481f84ab4f3a77A secreted luciferase for ex vivo monitoring of in vivo processesWurdinger, Thomas; Badr, Christian; Pike, Lisa; de Kleine, Ruben; Weissleder, Ralph; Breakefield, Xandra O.; Tannous, Bakhos A.Nature Methods (2008), 5 (2), 171-173CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)Luciferases are widely used to monitor biol. processes. Here we describe the naturally secreted Gaussia princeps luciferase (Gluc) as a highly sensitive reporter for quant. assessment of cells in vivo by measuring its concn. in blood. The Gluc blood assay complements in vivo bioluminescence imaging, which has the ability to localize the signal and provides a multifaceted assessment of cell viability, proliferation and location in exptl. disease and therapy models.
- 14Andreu, N., Zelmer, A., Fletcher, T., Elkington, P. T., Ward, T. H., Ripoll, J., Parish, T., Bancroft, G. J., Schaible, U., Robertson, B. D., and Wiles, S. (2010) Optimisation of bioluminescent reporters for use with mycobacteria PLoS One 5, e10777Google ScholarThere is no corresponding record for this reference.
- 15Shimomura, O., Masugi, T., Johnson, F. H., and Haneda, Y. (1978) Properties and reaction mechanism of the bioluminescence system of the deep-sea shrimp Oplophorus gracilirostris Biochemistry 17, 994– 998Google ScholarThere is no corresponding record for this reference.
- 16Kurowski, M. A. and Bujnicki, J. M. (2003) GeneSilico protein structure prediction meta-server Nucleic Acids Res. 31, 3305– 3307Google ScholarThere is no corresponding record for this reference.
- 17Flower, D. R., North, A. C., and Sansom, C. E. (2000) The lipocalin protein family: structural and sequence overview Biochim. Biophys. Acta 1482, 9– 24Google ScholarThere is no corresponding record for this reference.
- 18Loening, A. M., Fenn, T. D., Wu, A. M., and Gambhir, S. S. (2006) Consensus guided mutagenesis of Renilla luciferase yields enhanced stability and light output Protein Eng., Des. Sel. 19, 391– 400Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xotl2ksro%253D&md5=c006a4ff7a42c04c1ccf02b80fb1c9ffConsensus guided mutagenesis of Renilla luciferase yields enhanced stability and light outputLoening, Andreas Markus; Fenn, Timothy David; Wu, Anna M.; Gambhir, Sanjiv SamProtein Engineering, Design & Selection (2006), 19 (9), 391-400CODEN: PEDSBR; ISSN:1741-0126. (Oxford University Press)Luciferases, which have seen expansive employment as reporter genes in biol. research, could also be used in applications where the protein itself is conjugated to ligands to create probes that are appropriate for use in small animal imaging. As the bioluminescence activity of commonly used luciferases is too labile in serum to permit this application, specific mutations of Renilla luciferase, selected using a consensus sequence driven strategy, were screened for their ability to confer stability of activity in serum as well as their light output. Using this information, a total of eight favorable mutations were combined to generate a mutant Renilla luciferase (RLuc8) that, compared with the parental enzyme, is 200-fold more resistant to inactivation in murine serum and exhibits a 4-fold improvement in light output. Results of the mutational anal. were also used to generate a double mutant optimized for use as a reporter gene. The double mutant had half the resistance to inactivation in serum of the native enzyme while yielding a 5-fold improvement in light output.These variants of Renilla luciferase, which exhibit significantly improved properties compared with the native enzyme, will allow enhanced sensitivity in existing luciferase-based assays as well as enable the development of novel probes labeled with the luciferase protein.
- 19Woo, J. and von Arnim, A. G. (2008) Mutational optimization of the coelenterazine-dependent luciferase from Renilla Plant Methods 4, 23Google ScholarThere is no corresponding record for this reference.
- 20Branchini, B. R., Ablamsky, D. M., Davis, A. L., Southworth, T. L., Butler, B., Fan, F., Jathoul, A. P., and Pule, M. A. (2010) Red-emitting luciferases for bioluminescence reporter and imaging applications Anal. Biochem. 396, 290– 297Google ScholarThere is no corresponding record for this reference.
- 21Inouye, S. and Shimomura, O. (1997) The use of Renilla luciferase, Oplophorus luciferase, and Apoaequorin as bioluminescent reporter protein in the presence of coelenterazine analogues as substrate Biochem. Biophys. Res. Commun. 233, 349– 353Google ScholarThere is no corresponding record for this reference.
- 22Ando, Y., Niwa, K., Yamada, N., Enomoto, T., Irie, T., Kubota, H., Ohmiya, Y., and Akiyama, H. (2008) Firefly bioluminescence quantum yield and colour change by pH-sensitive green emission Nat. Photonics 2, 44– 47Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXmtVSnuw%253D%253D&md5=00549500c02c8644684b672d7530e863Firefly bioluminescence quantum yield and colour change by pH-sensitive green emissionAndo, Yoriko; Niwa, Kazuki; Yamada, Nobuyuki; Enomoto, Toshiteru; Irie, Tsutomu; Kubota, Hidehiro; Ohmiya, Yoshihiro; Akiyama, HidefumiNature Photonics (2008), 2 (1), 44-47CODEN: NPAHBY; ISSN:1749-4885. (Nature Publishing Group)Firefly bioluminescence is the most well-known ideal photoemitter system in biophotonics, known in particular for its extremely high quantum yield, 88 ± 25% (refs. 2,3) or higher, and its magnificent pH-dependent emission-color change between yellow-green and red, modelled as the chem. equil. between two corresponding states. However, the need for re-examn. has also been discussed. In this letter we quantify quantum yields and color changes using our new total-photon-flux spectrometer. We det. the highest quantum yield to be 41.0 ± 7.4% (1 std. deviation (s.d.) est., coverage factor k = 1), and find that bioluminescence spectra are systematically decompd. into one pH-sensitive and two pH-insensitive gaussian components. There is no intensity conversion between yellow-green and red emissions through pH equil., but simple intensity variation of the pH-sensitive gaussian peak at 2.2 eV causes the changes in emission colors. This represents a paradigm shift in the concept of color detn. from long-standing interpretation based on pH equil.
- 23Davis, R. E., Zhang, Y. Q., Southall, N., Staudt, L. M., Austin, C. P., Inglese, J., and Auld, D. S. (2007) A cell-based assay for IκBα stabilization using a two-color dual luciferase-based sensor Assay Drug Dev. Technol. 5, 85– 103Google ScholarThere is no corresponding record for this reference.
- 24Almond, B., Hawkins, E., Stecha, P., Garvin, D., Paguio, A., Butler, B. L., Beck, M., Wood, M., and Wood, K. (2003) Introducing ChromaLuc technology Promega Notes 85, 11– 14Google ScholarThere is no corresponding record for this reference.
- 25Swanson, B., Fan, F., and Wood, K. V. (2007) Enhanced response dynamics for transcription analysis using new pGL4 luciferase reporter vectors Cell Notes 17, 3– 5Google ScholarThere is no corresponding record for this reference.
- 26Auld, D. S., Zhang, Y. Q., Southall, N. T., Rai, G., Landsman, M., MacLure, J., Langevin, D., Thomas, C. J., Austin, C. P., and Inglese, J. (2009) A basis for reduced chemical library inhibition of firefly luciferase obtained from directed evolution J. Med. Chem. 52, 1450– 1458Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhvVWnt7o%253D&md5=aac37056be38a2c0ed06098d2e2f142aA Basis for Reduced Chemical Library Inhibition of Firefly Luciferase Obtained from Directed EvolutionAuld, Douglas S.; Zhang, Ya-Qin; Southall, Noel T.; Rai, Ganesha; Landsman, Marc; MacLure, Jennifer; Langevin, Daniel; Thomas, Craig J.; Austin, Christopher P.; Inglese, JamesJournal of Medicinal Chemistry (2009), 52 (5), 1450-1458CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)We measured the "druggability" of the ATP-dependent luciferase derived from the firefly Photuris pennsylvanica that was optimized using directed evolution (Ultra-Glo, Promega). Quant. high-throughput screening (qHTS) was used to det. IC50s of 198899 samples against a formulation of Ultra-Glo luciferase (Kinase-Glo). We found that only 0.1% of the Kinase-Glo inhibitors showed an IC50 < 10 μM compared to 0.9% found from a previous qHTS against the firefly luciferase from Photinus pyralis (lucPpy). Further, the max. affinity identified in the lucPpy qHTS was 50 nM, while for Kinase-Glo this value increased to 600 nM. Compds. with interactions stretching outside the luciferin binding pocket were largely lost with Ultra-Glo luciferase. Therefore, Ultra-Glo luciferase will show less compd. interference when used as an ATP sensor compared to lucPpy. This study demonstrates the power of large-scale quant. anal. of structure-activity relationships (>100K compds.) in addressing important questions such as a target's druggability.
- 27Fahey, R. C., Hunt, J. S., and Windham, G. C. (1977) On the cysteine and cystine content of proteins. Differences between intracellular and extracellular proteins J. Mol. Evol. 10, 155– 160Google ScholarThere is no corresponding record for this reference.
- 28Goerke, A. R., Loening, A. M., Gambhir, S. S., and Swartz, J. R. (2008) Cell-free metabolic engineering promotes high-level production of bioactive Gaussia princeps luciferase Metab. Eng. 10, 187– 200Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXnt1ektbc%253D&md5=1f54e550163e57beeae2a131e48a513cCell-free metabolic engineering promotes high-level production of bioactive Gaussia princeps luciferaseGoerke, Aaron R.; Loening, Andreas M.; Gambhir, Sanjiv S.; Swartz, James R.Metabolic Engineering (2008), 10 (3-4), 187-200CODEN: MEENFM; ISSN:1096-7176. (Elsevier Inc.)Due to its small size and intense luminescent signal, Gaussia princeps luciferase (GLuc) is attractive as a potential imaging agent in both cell culture and small animal research models. However, recombinant GLuc prodn. using in vivo techniques has only produced small quantities of active luciferase, likely due to five disulfide bonds being required for full activity. Cell-free biol. provides the freedom to control both the catalyst and chem. compns. in biol. reactions, and we capitalized on this to produce large amts. of highly active GLuc in cell-free reactions. Active yields were improved by mutating the cell ext. source strain to reduce proteolysis, adjusting reaction conditions to enhance oxidative protein folding, further activating energy metab., and encouraging post-translational activation. This cell-free protein synthesis procedure produced 412 μg/mL of purified GLuc, relative to 5 μg/mL isolated for intracellular Escherichia coli expression. The cell-free product had a specific activity of 4.2×1024 photons/s/mol, the highest reported activity for any characterized luciferase.
- 29Inouye, S. and Sasaki, S. (2007) Overexpression, purification and characterization of the catalytic component of Oplophorus luciferase in the deep-sea shrimp, Oplophorus gracilirostris Protein Expression Purif. 56, 261– 268Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXht1anurrL&md5=0e4fd0d955d54530c24027053fb1cc18Overexpression, purification and characterization of the catalytic component of Oplophorus luciferase in the deep-sea shrimp, Oplophorus gracilirostrisInouye, Satoshi; Sasaki, SatokoProtein Expression and Purification (2007), 56 (2), 261-268CODEN: PEXPEJ; ISSN:1046-5928. (Elsevier B.V.)The luciferase secreted by the deep-sea shrimp Oplophorus consists of 19 and 35 kDa proteins. The 19-kDa protein (19kOLase), the catalytic component of luminescence reaction, was expressed in Escherichia coli using the cold-shock inducted expression system. 19KOLase, expressed as inclusion bodies, was solubilized with 6 M urea and purified by urea-nickel chelate affinity chromatog. The yield of 19kOLase was 16 mg from 400 mL of cultured cells. 19kOLase in 6 M urea could be refolded rapidly by diln. with 50 mM Tris-HCl (pH 7.8)-10 mM EDTA, and the refolded protein showed luminescence activity. The luminescence properties of refolded 19kOLase were characterized, in comparison with native Oplophorus luciferase. Luminescence intensity with bisdeoxycoelenterazine as a substrate was stimulated in the presence of org. solvents. The 19kOLase is a thermolabile protein and is 98 % inhibited by 1 μM Cu2+. The cysteine residue of 19kOLase is not essential for catalysis of the luminescence reaction.
- 30Wong, G. G., Witek-Giannotti, J., Hewick, R. M., Clark, S. C., and Ogawa, M. (1988) Interleukin 6: identification as a hematopoietic colony-stimulating factor Behring Inst. Mitt. 83, 40– 47Google Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL1MXitFSmur8%253D&md5=b253f52b9e95b28e7ca725a9ef526756Interleukin 6: identification as a hematopoietic colony-stimulating factorWong, G. G.; Witek-Giannotti, J.; Hewick, R. M.; Clark, S. C.; Ogawa, M.Behring Institute Mitteilungen (1988), 83 (), 40-7CODEN: BHIMA2; ISSN:0301-0457.A cDNA encoding a novel myeloid colony-stimulating activity from a human T cell line was identified and mol. cloned. This cytokine proved to be identical to the factor currently known as interleukin (IL)-6, thereby demonstrating effects of IL-6 with hematopoietic target cells. In addn. to its ability to support murine granulocyte-macrophage colony formation, IL-6 acted synergistically with IL-3 in both the murine and human systems in support of colony formation by the primitive blast cell colony forming cell. This multitude of biol. activities suggests that IL-6 plays a prominent role within a network of cytokines in integrating the different arms of the host response to infection.
- 31Lippincott-Schwartz, J., Roberts, T. H., and Hirschberg, K. (2000) Secretory protein trafficking and organelle dynamics in living cells Annu. Rev. Cell Dev. Biol. 16, 557– 589Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXpvFyq&md5=b5cf93a3d1fd4b750c843c43780bd567Secretory protein trafficking and organelle dynamics in living cellsLippincott-Schwartz, Jennifer; Roberts, Theresa H.; Hirschberg, KoretAnnual Review of Cell and Developmental Biology (2000), 16 (), 557-589CODEN: ARDBF8; ISSN:1081-0706. (Annual Reviews Inc.)A review with many refs. Green fluorescent protein chimerae acting as reporters for protein localization and trafficking within the secretory membrane system of living cells have been used in a wide variety of applications, including time-lapse imaging, double-labeling, energy transfer, quantitation, and photobleaching expts. Results from this work are clarifying the steps involved in the formation, translocation, and fusion of transport intermediates; the organization and biogenesis of organelles; and the mechanisms of protein retention, sorting, and recycling in the secretory pathway. In so doing, they are broadening our thinking about the temporal and spatial relationships among secretory organelles and the membrane trafficking pathways that operate between them.
- 32Simmons, S. O. (2011) Fireflies in the coalmine: luciferase technologies in next-generation toxicity testing Comb. Chem. High Throughput Screening 14, 688– 702Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtVKqtrnI&md5=1224daf1cf9b892c342b3c87d13c478bFireflies in the coalmine: luciferase technologies in next-generation toxicity testingSimmons, Steven O.Combinatorial Chemistry & High Throughput Screening (2011), 14 (8), 688-702CODEN: CCHSFU; ISSN:1386-2073. (Bentham Science Publishers Ltd.)Whole-animal studies have been the mainstay of toxicity testing for decades. These approaches are too expensive and laborious to effectively characterize all of the chems. currently in com. use. In addn., there are social and ethical pressures to reduce, refine and replace animal testing in toxicol. The National Research Council (NRC) has outlined a new strategy to transition from animal-based tests to high throughput, cell-based assays and computational modeling approaches to characterize chem. toxicants. Crit. to this vision, assays that measure toxicity pathways assocd. with adverse health effects must be developed. Bioluminescent assays are particularly well suited to the demands of next-generation toxicity testing because they measure a wide range of biol. activities in a quant. and high throughput manner. This review describes the limitations of traditional, animal-based toxicity testing and discusses the current and developing uses of bioluminescent technologies in next-generation testing based on three general assays formats: luciferase-limited assays, ATP-limited assays and luciferin-limited assays.
- 33Rehemtulla, A., Taneja, N., and Ross, B. D. (2004) Bioluminescence detection of cells having stabilized p53 in response to a genotoxic event Mol. Imaging 3, 63– 68Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXkslGjsLc%253D&md5=e3a74f1ffe9966213c680a7668f356e3Bioluminescence detection of cells having stabilized p53 in response to a genotoxic eventRehemtulla, Alnawaz; Taneja, Neelam; Ross, Brian D.Molecular Imaging (2004), 3 (1), 63-68CODEN: MIOMBP; ISSN:1535-3508. (MIT Press)Inactivation of p53 is one of the most frequent mol. events in neoplastic transformation. Approx. 60% of all human tumors have mutations in both p53 alleles. Wild-type p53 activity is regulated in large part by the proteosome-dependent degrdn. of p53, resulting in a short p53 half-life in unstressed and untransformed cells. Activation of p53 by a variety of stimuli, including DNA damage induced by genotoxic drugs or radiation, is accomplished by stabilization of wild-type p53. The stabilized and active p53 can result in either cell-cycle arrest or apoptosis. Surprisingly, the majority of tumor-assocd., inactivating p53 mutations also result in p53 accumulation. Thus, constitutive elevation of p53 levels in cells is a reliable measure of p53 inactivation, whereas transiently increased p53 levels reflect a recent genotoxic stress. In order to facilitate noninvasive imaging of p53 accumulation, the authors here describe the construction of a p53-luciferase fusion protein. Induction of DNA damage in cells expressing, the fusion protein resulted in a time-dependent accumulation of the fusion that was noninvasively detected using bioluminescence imaging and validated by Western blot anal. The p53-Luc protein retains p53 function because its expression in HCT116 cells lacking functional p53 resulted in activation of p21 expression as well as induction of apoptosis in response to a DNA damaging event. Employed in a transgenic animal model, the proposed p53-reporter fusion protein will be useful for studying p53 activation in response to exposure to DNA-damaging carcinogenic agents. It could also be used to study p53 stabilization as a result of inactivating p53 mutations. Such studies will further the understanding of p53's role as the "guardian of the genome" and its function in tumorigenesis.
- 34Horn, H. F. and Vousden, K. H. (2007) Coping with stress: multiple ways to activate p53 Oncogene 26, 1306– 1316Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXitVKhsLY%253D&md5=2d7b83d1eee3c060185f2441868ed16dCoping with stress: multiple ways to activate p53Horn, H. F.; Vousden, K. H.Oncogene (2007), 26 (9), 1306-1316CODEN: ONCNES; ISSN:0950-9232. (Nature Publishing Group)A review. Over the years, p53 has been shown to sit at the center of an increasingly complex web of incoming stress signals and outgoing effector pathways. The no. and diversity of stress signals that lead to p53 activation illustrates the breadth of p53's remit - responding to a wide variety of potentially oncogenic insults to prevent tumor development. Interestingly, different stress signals can use different and independent pathways to activate p53, and there is some evidence that different stress signals can mediate different responses. How each of the responses to p53 contributes to inhibition of malignant progression is beginning to be clarified, with the hope that identification of responses that are key to tumor suppression will allow a more focused and effective search for new therapeutic targets. In this review, we will highlight some recently identified roles for p53 in tumor suppression, and discuss some of the numerous mechanisms through which p53 can be regulated and activated.
- 35Nishizuka, Y. (1984) The role of protein kinase C in cell surface signal transduction and tumour promotion Nature 308, 693– 698Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL2cXitVCmtrk%253D&md5=33d139697753d3f7e10417573e131ff9The role of protein kinase C in cell surface signal transduction and tumor promotionNishizuka, YasutomiNature (London, United Kingdom) (1984), 308 (5961), 693-8CODEN: NATUAS; ISSN:0028-0836.A review with 80 refs. on the title role of protein kinase C [9026-43-1].
- 36Htun, H., Barsony, J., Renyi, I., Gould, D. L., and Hager, G. L. (1996) Visualization of glucocorticoid receptor translocation and intranuclear organization in living cells with a green fluorescent protein chimera Proc. Natl. Acad. Sci. U.S.A. 93, 4845– 4850Google ScholarThere is no corresponding record for this reference.
- 37Nakajima, Y., Yamazaki, T., Nishii, S., Noguchi, T., Hoshino, H., Niwa, K., Viviani, V. R., and Ohmiya, Y. (2010) Enhanced beetle luciferase for high-resolution bioluminescence imaging PLoS One 5, e10011Google ScholarThere is no corresponding record for this reference.
- 38Suzuki, T., Kondo, C., Kanamori, T., and Inouye, S. (2011) Video rate bioluminescence imaging of secretory proteins in living cells: localization, secretory frequency, and quantification Anal. Biochem. 415, 182– 189Google ScholarThere is no corresponding record for this reference.
Cited By
This article is cited by 1154 publications.
- Anneliese M. M. Gest, Ayse Z. Sahan, Yanghao Zhong, Wei Lin, Sohum Mehta, Jin Zhang. Molecular Spies in Action: Genetically Encoded Fluorescent Biosensors Light up Cellular Signals. Chemical Reviews 2024, 124
(22)
, 12573-12660. https://doi.org/10.1021/acs.chemrev.4c00293
- Giulia Tedeschi, Mariana X. Navarro, Lorenzo Scipioni, Tanvi K. Sondhi, Jennifer A. Prescher, Michelle A. Digman. Monitoring Macrophage Polarization with Gene Expression Reporters and Bioluminescence Phasor Analysis. Chemical & Biomedical Imaging 2024, 2
(11)
, 765-774. https://doi.org/10.1021/cbmi.4c00049
- Yinghui Yang, Akihito Inoue, Takanobu Yasuda, Hiroshi Ueda, Bo Zhu, Tetsuya Kitaguchi. BRET Nano Q-Body: A Nanobody-Based Ratiometric Bioluminescent Immunosensor for Point-of-Care Testing. ACS Sensors 2024, 9
(11)
, 5955-5965. https://doi.org/10.1021/acssensors.4c01800
- Paramesh K. Ramaraj, Mugdha Pol, Samuel L. Scinto, Xinqiao Jia, Joseph M. Fox. Covalent Attachment of Functional Proteins to Microfiber Surfaces via a General Strategy for Site-Selective Tetrazine Ligation. ACS Applied Materials & Interfaces 2024, 16
(46)
, 63195-63206. https://doi.org/10.1021/acsami.4c12609
- Yoo-Hong Min, Yoonseo Hong, Cheol-Hee Kim, Kyung-Ho Lee, Yong-Beom Shin, Ju-Young Byun. Split Probe-Induced Protein Translational Amplification for Nucleic Acid Detection. ACS Applied Bio Materials 2024, Article ASAP.
- Antoine Lévrier, Julien Capin, Pauline Mayonove, Ioannis-Ilie Karpathakis, Peter Voyvodic, Angelique DeVisch, Ana Zuniga, Martin Cohen-Gonsaud, Stéphanie Cabantous, Vincent Noireaux, Jerome Bonnet. Split Reporters Facilitate Monitoring of Gene Expression and Peptide Production in Linear Cell-Free Transcription–Translation Systems. ACS Synthetic Biology 2024, 13
(10)
, 3119-3127. https://doi.org/10.1021/acssynbio.4c00353
- Yanian Xiong, Yingtung Lo, Huizhu Song, Jianzhong Lu. Development of a Self-Luminescent Living Bioreactor for Enhancing Photodynamic Therapy in Breast Cancer. Bioconjugate Chemistry 2024, 35
(8)
, 1269-1282. https://doi.org/10.1021/acs.bioconjchem.4c00334
- Lingfei Wang, Hanfeng Lin, Bin Yang, Xiqian Jiang, Jianwei Chen, Sandipan Roy Chowdhury, Ninghui Cheng, Paul A. Nakata, David M. Lonard, Meng. C. Wang, Jin Wang. Development of a Novel Amplifiable System to Quantify Hydrogen Peroxide in Living Cells. Journal of the American Chemical Society 2024, 146
(32)
, 22396-22404. https://doi.org/10.1021/jacs.4c05366
- Hanfeng Lin, Kristin Riching, May Poh Lai, Dong Lu, Ran Cheng, Xiaoli Qi, Jin Wang. Lysineless HiBiT and NanoLuc Tagging Systems as Alternative Tools for Monitoring Targeted Protein Degradation. ACS Medicinal Chemistry Letters 2024, 15
(8)
, 1367-1375. https://doi.org/10.1021/acsmedchemlett.4c00271
- Brian L. Zhong, Jeandele M. Elliot, Pengli Wang, Hongquan Li, R. Nelson Hall, Bo Wang, Manu Prakash, Alexander R. Dunn. Split Luciferase Molecular Tension Sensors for Bioluminescent Readout of Mechanical Forces in Biological Systems. ACS Sensors 2024, 9
(7)
, 3489-3495. https://doi.org/10.1021/acssensors.3c02664
- Eleonora Comeo, Joëlle Goulding, Chia-Yang Lin, Marleen Groenen, Jeanette Woolard, Nicholas D. Kindon, Clare R. Harwood, Simon Platt, Stephen J. Briddon, Laura E. Kilpatrick, Peter J. Scammells, Stephen J. Hill, Barrie Kellam. Ligand-Directed Labeling of the Adenosine A1 Receptor in Living Cells. Journal of Medicinal Chemistry 2024, 67
(14)
, 12099-12117. https://doi.org/10.1021/acs.jmedchem.4c00835
- Atsushi Ogawa, Masahiro Fujikawa, Kazuki Onishi, Hajime Takahashi. Cell-Free Biosensors Based on Modular Eukaryotic Riboswitches That Function in One Pot at Ambient Temperature. ACS Synthetic Biology 2024, 13
(7)
, 2238-2245. https://doi.org/10.1021/acssynbio.4c00341
- Lara Toy, Max E. Huber, Minhee Lee, Ana Alonso Bartolomé, Natalia V. Ortiz Zacarías, Sherif Nasser, Stephan Scholl, Darius P. Zlotos, Yasmine M. Mandour, Laura H. Heitman, Martyna Szpakowska, Andy Chevigné, Matthias Schiedel. Fluorophore-Labeled Pyrrolones Targeting the Intracellular Allosteric Binding Site of the Chemokine Receptor CCR1. ACS Pharmacology & Translational Science 2024, 7
(7)
, 2080-2092. https://doi.org/10.1021/acsptsci.4c00182
- Juan Sanz García, Rosa Maskri, Alexander Mitrushchenkov, Loïc Joubert-Doriol. Optimizing Conical Intersections without Explicit Use of Non-Adiabatic Couplings. Journal of Chemical Theory and Computation 2024, 20
(13)
, 5643-5654. https://doi.org/10.1021/acs.jctc.4c00326
- Angelica Rose Galvan, Christopher M. Green, Shelby L. Hooe, Esra Oktay, Meghna Thakur, Sebastián A. Díaz, Remi Veneziano, Igor L. Medintz, Divita Mathur. Design and Characterization of a Gene-Encoding DNA Nanoparticle in a Cell-Free Transcription–Translation System. ACS Applied Nano Materials 2024, 7
(11)
, 12891-12902. https://doi.org/10.1021/acsanm.4c01456
- Georg Rueedi, Philippe Panchaud, Astrid Friedli, Jean-Luc Specklin, Christian Hubschwerlen, Anne-Catherine Blumstein, Patrick Caspers, Michel Enderlin-Paput, Loïc Jacob, Christopher Kohl, Hans H. Locher, Philippe Pfaff, Christine Schmitt, Peter Seiler, Daniel Ritz. Discovery and Structure–Activity Relationship of Cadazolid: A First-In-Class Quinoxolidinone Antibiotic for the Treatment of Clostridioides difficile Infection. Journal of Medicinal Chemistry 2024, 67
(11)
, 9465-9484. https://doi.org/10.1021/acs.jmedchem.4c00558
- Francesco Russo, Beatrice Civili, Nicolas Winssinger. Bright Red Bioluminescence from Semisynthetic NanoLuc (sNLuc). ACS Chemical Biology 2024, 19
(5)
, 1035-1039. https://doi.org/10.1021/acschembio.4c00033
- Shengyu Zhao, Ying Xiong, Ranganayakulu Sunnapu, Yiyu Zhang, Xiaodong Tian, Hui-wang Ai. Bioluminescence Imaging of Potassium Ion Using a Sensory Luciferin and an Engineered Luciferase. Journal of the American Chemical Society 2024, 146
(19)
, 13406-13416. https://doi.org/10.1021/jacs.4c02473
- Hannah Vogt, Patrick Shinkwin, Max E. Huber, Nico Staffen, Harald Hübner, Peter Gmeiner, Matthias Schiedel, Dorothee Weikert. Development of a Fluorescent Ligand for the Intracellular Allosteric Binding Site of the Neurotensin Receptor 1. ACS Pharmacology & Translational Science 2024, 7
(5)
, 1533-1545. https://doi.org/10.1021/acsptsci.4c00086
- Taylor B. Dolberg, Taylor F. Gunnels, Te Ling, Kelly A. Sarnese, John D. Crispino, Joshua N. Leonard. Building Synthetic Biosensors Using Red Blood Cell Proteins. ACS Synthetic Biology 2024, 13
(4)
, 1273-1289. https://doi.org/10.1021/acssynbio.3c00754
- Jakob Gleixner, Sergei Kopanchuk, Lukas Grätz, Maris-Johanna Tahk, Tõnis Laasfeld, Santa Veikšina, Carina Höring, Albert O. Gattor, Laura J. Humphrys, Christoph Müller, Nataliya Archipowa, Johannes Köckenberger, Markus R. Heinrich, Roger Jan Kutta, Ago Rinken, Max Keller. Illuminating Neuropeptide Y Y4 Receptor Binding: Fluorescent Cyclic Peptides with Subnanomolar Binding Affinity as Novel Molecular Tools. ACS Pharmacology & Translational Science 2024, 7
(4)
, 1142-1168. https://doi.org/10.1021/acsptsci.4c00013
- Eva A. van Aalen, Joep J. J. Lurvink, Leandra Vermeulen, Benice van Gerven, Yan Ni, Remco Arts, Maarten Merkx. Turning Antibodies into Ratiometric Bioluminescent Sensors for Competition-Based Homogeneous Immunoassays. ACS Sensors 2024, 9
(3)
, 1401-1409. https://doi.org/10.1021/acssensors.3c02478
- Ethan M. Jones, Yang Su, Chris Sander, Quincey A. Justman, Michael Springer, Pamela A. Silver. LanTERN: A Fluorescent Sensor That Specifically Responds to Lanthanides. ACS Synthetic Biology 2024, 13
(3)
, 958-962. https://doi.org/10.1021/acssynbio.3c00600
- Mark A. Klein, Sergey Lazarev, Charles Gervasi, Cristopher Cowan, Thomas Machleidt, Rachel Friedman Ohana. Luciferase Calibrants Enable Absolute Quantitation of Bioluminescence Power. ACS Measurement Science Au 2023, 3
(6)
, 496-503. https://doi.org/10.1021/acsmeasuresciau.3c00036
- Yi-Sheng Lu, Ruhan Fan, Willem Vugs, Lieuwe Biewenga, Ziming Zhou, Sanahan Vijayakumar, Maarten Merkx, Michael J. Sailor. Harnessing the Materials Chemistry of Mesoporous Silicon Nanoparticles to Prepare “Armor-Clad” Enzymes. Chemistry of Materials 2023, 35
(23)
, 10247-10257. https://doi.org/10.1021/acs.chemmater.3c02637
- Masatoshi Eguchi, Hideaki Yoshimura, Yoshibumi Ueda, Takeaki Ozawa. Split Luciferase-Fragment Reconstitution for Unveiling RNA Localization and Dynamics in Live Cells. ACS Sensors 2023, 8
(11)
, 4055-4063. https://doi.org/10.1021/acssensors.3c01080
- Philipp Kemp, Wadim Weber, Charlotte Desczyk, Marwan Kaufmann, Josefine Panthel, Theresa Wörmann, Viktor Stein. Dissecting the Permeability of the Escherichia coli Cell Envelope to a Small Molecule Using Tailored Intensiometric Fluorescent Protein Sensors. ACS Omega 2023, 8
(42)
, 39562-39569. https://doi.org/10.1021/acsomega.3c05405
- Dawson B. Ling, William Nguyen, Oliver Looker, Zahra Razook, Kirsty McCann, Alyssa E. Barry, Christian Scheurer, Sergio Wittlin, Mufuliat Toyin Famodimu, Michael J Delves, Hayley E. Bullen, Brendan S. Crabb, Brad E. Sleebs, Paul R. Gilson. A Pyridyl-Furan Series Developed from the Open Global Health Library Block Red Blood Cell Invasion and Protein Trafficking in Plasmodium falciparum through Potential Inhibition of the Parasite’s PI4KIIIB Enzyme. ACS Infectious Diseases 2023, 9
(9)
, 1695-1710. https://doi.org/10.1021/acsinfecdis.3c00138
- Whitney K. Lieberman, Zachary A. Brown, Daniel S. Kantner, Yihang Jing, Emily Megill, Nya D. Evans, McKenna C. Crawford, Isita Jhulki, Carissa Grose, Jane E. Jones, Nathaniel W. Snyder, Jordan L. Meier. Chemoproteomics Yields a Selective Molecular Host for Acetyl-CoA. Journal of the American Chemical Society 2023, 145
(30)
, 16899-16905. https://doi.org/10.1021/jacs.3c05489
- Max E. Huber, Silas Wurnig, Lara Toy, Corinna Weiler, Nicole Merten, Evi Kostenis, Finn K. Hansen, Matthias Schiedel. Fluorescent Ligands Enable Target Engagement Studies for the Intracellular Allosteric Binding Site of the Chemokine Receptor CXCR2. Journal of Medicinal Chemistry 2023, 66
(14)
, 9916-9933. https://doi.org/10.1021/acs.jmedchem.3c00769
- Vasilisa V. Krasitskaya, Maxim K. Efremov, Ludmila A. Frank. Luciferase NLuc Site-Specific Conjugation to Generate Reporters for In Vitro Assays. Bioconjugate Chemistry 2023, 34
(7)
, 1282-1289. https://doi.org/10.1021/acs.bioconjchem.3c00165
- Ruslan Gibadullin, Brian P. Cary, Samuel H. Gellman. Differential Responses of the GLP-1 and GLP-2 Receptors to N-Terminal Modification of a Dual Agonist. Journal of the American Chemical Society 2023, 145
(22)
, 12105-12114. https://doi.org/10.1021/jacs.3c01628
- Junbin Li, Na Wang, Mengyi Xiong, Min Dai, Cheng Xie, Qianqian Wang, Ke Quan, Yibo Zhou, Zhihe Qing. A Reaction-Based Ratiometric Bioluminescent Platform for Point-of-Care and Quantitative Detection Using a Smartphone. Analytical Chemistry 2023, 95
(18)
, 7142-7149. https://doi.org/10.1021/acs.analchem.2c05422
- Anhuy Pham, Shane Bassett, Wilfred Chen, Nancy A. Da Silva. Assembly of Metabolons in Yeast Using Cas6-Mediated RNA Scaffolding. ACS Synthetic Biology 2023, 12
(4)
, 1164-1174. https://doi.org/10.1021/acssynbio.2c00650
- Lai Wei, Kaijing Xiang, Hongjian Kang, Yancheng Yu, Hongjie Fan, Han Zhou, Tao Hou, Yonglin Ge, Jixia Wang, Zhimou Guo, Yang Chen, Yaopeng Zhao, Xinmiao Liang. Development and Characterization of Fluorescent Probes for the G Protein-Coupled Receptor 35. ACS Medicinal Chemistry Letters 2023, 14
(4)
, 411-416. https://doi.org/10.1021/acsmedchemlett.2c00461
- Jie Yu, Yan Zhang, Yanping Zhao, Xuxiang Zhang, Hongqiang Ren. Highly Sensitive and Selective Detection of Inorganic Phosphates in the Water Environment by Biosensors Based on Bioluminescence Resonance Energy Transfer. Analytical Chemistry 2023, 95
(11)
, 4904-4913. https://doi.org/10.1021/acs.analchem.2c04748
- Peter Braun, Rene Raab, Joachim J. Bugert, Simone Braun. Recombinant Reporter Phage rTUN1::nLuc Enables Rapid Detection and Real-Time Antibiotic Susceptibility Testing of Klebsiella pneumoniae K64 Strains. ACS Sensors 2023, 8
(2)
, 630-639. https://doi.org/10.1021/acssensors.2c01822
- Anna C. Love, Donald R. Caldwell, Bethany Kolbaba-Kartchner, Katherine M. Townsend, Lila P. Halbers, Zi Yao, Caroline K. Brennan, Joseph Ivanic, Tanya Hadjian, Jeremy H. Mills, Martin J. Schnermann, Jennifer A. Prescher. Red-Shifted Coumarin Luciferins for Improved Bioluminescence Imaging. Journal of the American Chemical Society 2023, 145
(6)
, 3335-3345. https://doi.org/10.1021/jacs.2c07220
- Lan Mi, Qikun Yu, Aruni P. K. K. Karunanayake Mudiyanselage, Rigumula Wu, Zhining Sun, Ru Zheng, Kewei Ren, Mingxu You. Genetically Encoded RNA-Based Bioluminescence Resonance Energy Transfer (BRET) Sensors. ACS Sensors 2023, 8
(1)
, 308-316. https://doi.org/10.1021/acssensors.2c02213
- Atsushi Ogawa, Honami Inoue, Yu Itoh, Hajime Takahashi. Facile Expansion of the Variety of Orthogonal Ligand/Aptamer Pairs for Artificial Riboswitches. ACS Synthetic Biology 2023, 12
(1)
, 35-42. https://doi.org/10.1021/acssynbio.2c00475
- Jingxin Liu, Chenchen Ge, Ling Zha, Ligen Lin, Rongsong Li. Simple Nano-Luciferase-Based Assay for the Rapid and High-Throughput Detection of SARS-CoV-2 3C-Like Protease. Analytical Chemistry 2023, 95
(2)
, 714-719. https://doi.org/10.1021/acs.analchem.2c02590
- Dimitra Apostolidou, Pan Zhang, Weitao Yang, Piotr E. Marszalek. Mechanical Unfolding and Refolding of NanoLuc via Single-Molecule Force Spectroscopy and Computer Simulations. Biomacromolecules 2022, 23
(12)
, 5164-5178. https://doi.org/10.1021/acs.biomac.2c00997
- Shikha S. Chauhan, Nick J. Marotta, Anna C. Karls, Emily E. Weinert. Binding of 2′,3′-Cyclic Nucleotide Monophosphates to Bacterial Ribosomes Inhibits Translation. ACS Central Science 2022, 8
(11)
, 1518-1526. https://doi.org/10.1021/acscentsci.2c00681
- Kathryn A. Hufziger, Emma L. Farquharson, Brenda G. Werner, Qingmin Chen, Julie M. Goddard, Sam R. Nugen. In Vivo Capsid Engineering of Bacteriophages for Oriented Surface Conjugation. ACS Applied Bio Materials 2022, 5
(11)
, 5104-5112. https://doi.org/10.1021/acsabm.2c00428
- Lukas Grätz, Christoph Müller, Andrea Pegoli, Lisa Schindler, Günther Bernhardt, Timo Littmann. Insertion of Nanoluc into the Extracellular Loops as a Complementary Method To Establish BRET-Based Binding Assays for GPCRs. ACS Pharmacology & Translational Science 2022, 5
(11)
, 1142-1155. https://doi.org/10.1021/acsptsci.2c00162
- Mohammad Javad Afshari, Cang Li, Jianfeng Zeng, Jiabin Cui, Shuwang Wu, Mingyuan Gao. Self-illuminating NIR-II bioluminescence imaging probe based on silver sulfide quantum dots. ACS Nano 2022, 16
(10)
, 16824-16832. https://doi.org/10.1021/acsnano.2c06667
- Maylis Boitet, Hyeju Eun, Asma Achek, Virgínia Carla de Almeida Falcão, Vincent Delorme, Regis Grailhe. Biolum’ RGB: A Low-Cost, Versatile, and Sensitive Bioluminescence Imaging Instrument for a Broad Range of Users. ACS Sensors 2022, 7
(9)
, 2556-2566. https://doi.org/10.1021/acssensors.2c00457
- Tino W. Sanchez, Michael H. Ronzetti, Ashley E. Owens, Maria Antony, Ty Voss, Eric Wallgren, Daniel Talley, Krishna Balakrishnan, Sebastian E. Leyes Porello, Ganesha Rai, Juan J. Marugan, Samuel G. Michael, Bolormaa Baljinnyam, Noel Southall, Anton Simeonov, Mark J. Henderson. Real-Time Cellular Thermal Shift Assay to Monitor Target Engagement. ACS Chemical Biology 2022, 17
(9)
, 2471-2482. https://doi.org/10.1021/acschembio.2c00334
- Anil Mathew Tharappel, Zhong Li, Yan Chun Zhu, Xiangmeng Wu, Sudha Chaturvedi, Qing-Yu Zhang, Hongmin Li. Calcimycin Inhibits Cryptococcus neoformans In Vitro and In Vivo by Targeting the Prp8 Intein Splicing. ACS Infectious Diseases 2022, 8
(9)
, 1851-1868. https://doi.org/10.1021/acsinfecdis.2c00137
- Lara Toy, Max E. Huber, Maximilian F. Schmidt, Dorothee Weikert, Matthias Schiedel. Fluorescent Ligands Targeting the Intracellular Allosteric Binding Site of the Chemokine Receptor CCR2. ACS Chemical Biology 2022, 17
(8)
, 2142-2152. https://doi.org/10.1021/acschembio.2c00263
- Virginia A. Kincaid, Hui Wang, Casey A. Sondgeroth, Emily A. Torio, Valerie T. Ressler, Connor Fitzgerald, Mary P. Hall, Robin Hurst, Monika G. Wood, Julia K. Gilden, Thomas A. Kirkland, Dan Lazar, Hsu Chia-Chang, Lance P. Encell, Thomas Machleidt, Wenhui Zhou, Melanie L. Dart. Simple, Rapid Chemical Labeling and Screening of Antibodies with Luminescent Peptides. ACS Chemical Biology 2022, 17
(8)
, 2179-2187. https://doi.org/10.1021/acschembio.2c00306
- Rebeka C. Fanti, Stanley N. S. Vasconcelos, Carolina M. C. Catta-Preta, Jaryd R. Sullivan, Gustavo P. Riboldi, Caio V. dos Reis, Priscila Z. Ramos, Aled M. Edwards, Marcel A. Behr, Rafael M. Couñago. A Target Engagement Assay to Assess Uptake, Potency, and Retention of Antibiotics in Living Bacteria. ACS Infectious Diseases 2022, 8
(8)
, 1449-1467. https://doi.org/10.1021/acsinfecdis.2c00073
- Ying Xiong, Yiyu Zhang, Zefan Li, Md Shamim Reza, Xinyu Li, Xiaodong Tian, Hui-wang Ai. Engineered Amber-Emitting Nano Luciferase and Its Use for Immunobioluminescence Imaging In Vivo. Journal of the American Chemical Society 2022, 144
(31)
, 14101-14111. https://doi.org/10.1021/jacs.2c02320
- Dan Sindhikara Jennifer Johnston . Roles of Conformations on Predictions of Peptide Properties. , 103-135. https://doi.org/10.1021/bk-2022-1417.ch004
- Alexander Gräwe, Maarten Merkx, Viktor Stein. iFLinkC-X: A Scalable Framework to Assemble Bespoke Genetically Encoded Co-polymeric Linkers of Variable Lengths and Amino Acid Composition. Bioconjugate Chemistry 2022, 33
(7)
, 1415-1421. https://doi.org/10.1021/acs.bioconjchem.2c00250
- Nikki McArthur, Carlos Cruz-Teran, Apoorva Thatavarty, Gregory T. Reeves, Balaji M. Rao. Experimental and Analytical Framework for “Mix-and-Read” Assays Based on Split Luciferase. ACS Omega 2022, 7
(28)
, 24551-24560. https://doi.org/10.1021/acsomega.2c02319
- Xuan Yang, Rebekah J. Dickmander, Armin Bayati, Sharon A. Taft-Benz, Jeffery L. Smith, Carrow I. Wells, Emily A. Madden, Jason W. Brown, Erik M. Lenarcic, Boyd L. Yount, Jr, Edcon Chang, Alison D. Axtman, Ralph S. Baric, Mark T. Heise, Peter S. McPherson, Nathaniel J. Moorman, Timothy M. Willson. Host Kinase CSNK2 is a Target for Inhibition of Pathogenic SARS-like β-Coronaviruses. ACS Chemical Biology 2022, 17
(7)
, 1937-1950. https://doi.org/10.1021/acschembio.2c00378
- Masaharu Somiya, Shun’ichi Kuroda. Engineering of Extracellular Vesicles for Small Molecule-Regulated Cargo Loading and Cytoplasmic Delivery of Bioactive Proteins. Molecular Pharmaceutics 2022, 19
(7)
, 2495-2505. https://doi.org/10.1021/acs.molpharmaceut.2c00192
- Claire M. S. Michielsen, Eva A. van Aalen, Maarten Merkx. Ratiometric Bioluminescent Zinc Sensor Proteins to Quantify Serum and Intracellular Free Zn2+. ACS Chemical Biology 2022, 17
(6)
, 1567-1576. https://doi.org/10.1021/acschembio.2c00227
- Hope Adamson, Modupe O. Ajayi, Kate E. Gilroy, Michael J. McPherson, Darren C. Tomlinson, Lars J. C. Jeuken. Rapid Quantification of C. difficile Glutamate Dehydrogenase and Toxin B (TcdB) with a NanoBiT Split-Luciferase Assay. Analytical Chemistry 2022, 94
(23)
, 8156-8163. https://doi.org/10.1021/acs.analchem.1c05206
- Yuna Nakagawa, Jan Vincent V. Arafiles, Yoshimasa Kawaguchi, Ikuhiko Nakase, Hisaaki Hirose, Shiroh Futaki. Stearylated Macropinocytosis-Inducing Peptides Facilitating the Cellular Uptake of Small Extracellular Vesicles. Bioconjugate Chemistry 2022, 33
(5)
, 869-880. https://doi.org/10.1021/acs.bioconjchem.2c00113
- Wenhua Li, Zhao Ma, Jiwei Chen, Gaopan Dong, Lupei Du, Minyong Li. Discovery of Environment-Sensitive Fluorescent Ligands of β-Adrenergic Receptors for Cell Imaging and NanoBRET Assay. Analytical Chemistry 2022, 94
(19)
, 7021-7028. https://doi.org/10.1021/acs.analchem.1c05646
- Yuki Ohmuro-Matsuyama, Tadaomi Furuta, Hayato Matsui, Masaki Kanai, Hiroshi Ueda. Miniaturization of Bright Light-Emitting Luciferase ALuc: picALuc. ACS Chemical Biology 2022, 17
(4)
, 864-872. https://doi.org/10.1021/acschembio.1c00897
- Elbegduuren Erdenee, Alice Y. Ting. A Dual-Purpose Real-Time Indicator and Transcriptional Integrator for Calcium Detection in Living Cells. ACS Synthetic Biology 2022, 11
(3)
, 1086-1095. https://doi.org/10.1021/acssynbio.1c00597
- Mariko Orioka, Masatoshi Eguchi, Yuki Mizui, Yuma Ikeda, Akihiro Sakama, Qiaojing Li, Hideaki Yoshimura, Takeaki Ozawa, Daniel Citterio, Yuki Hiruta. A Series of Furimazine Derivatives for Sustained Live-Cell Bioluminescence Imaging and Application to the Monitoring of Myogenesis at the Single-Cell Level. Bioconjugate Chemistry 2022, 33
(3)
, 496-504. https://doi.org/10.1021/acs.bioconjchem.2c00035
- Linggang Zheng, Yang Tan, Yucan Hu, Juntao Shen, Zepeng Qu, Xianbo Chen, Chun Loong Ho, Elaine Lai-Han Leung, Wei Zhao, Lei Dai. CRISPR/Cas-Based Genome Editing for Human Gut Commensal Bacteroides Species. ACS Synthetic Biology 2022, 11
(1)
, 464-472. https://doi.org/10.1021/acssynbio.1c00543
- Ssu-Tzu Tsai, Wen-Jui Cheng, Qian-Xian Zhang, Yi-Chun Yeh. Gold-Specific Biosensor for Monitoring Wastewater Using Genetically Engineered Cupriavidus metallidurans CH34. ACS Synthetic Biology 2021, 10
(12)
, 3576-3582. https://doi.org/10.1021/acssynbio.1c00520
- Mihris Ibnu Saleem Naduthodi, Christian Südfeld, Emmanouil Klimis Avitzigiannis, Nicola Trevisan, Eduard van Lith, Javier Alcaide Sancho, Sarah D’Adamo, Maria Barbosa, John van der Oost. Comprehensive Genome Engineering Toolbox for Microalgae Nannochloropsis oceanica Based on CRISPR-Cas Systems. ACS Synthetic Biology 2021, 10
(12)
, 3369-3378. https://doi.org/10.1021/acssynbio.1c00329
- Shirley Liu, Yichi Su, Michael Z. Lin, John A. Ronald. Brightening up Biology: Advances in Luciferase Systems for in Vivo Imaging. ACS Chemical Biology 2021, 16
(12)
, 2707-2718. https://doi.org/10.1021/acschembio.1c00549
- Taha Azad, Helena J. Janse van Rensburg, Jessica Morgan, Reza Rezaei, Mathieu J. F. Crupi, Rui Chen, Mina Ghahremani, Monire Jamalkhah, Nicole Forbes, Carolina Ilkow, John C. Bell. Luciferase-Based Biosensors in the Era of the COVID-19 Pandemic. ACS Nanoscience Au 2021, 1
(1)
, 15-37. https://doi.org/10.1021/acsnanoscienceau.1c00009
- Mengying Deng, Jing Yuan, Haibin Yang, Xuli Wu, Xiaoyuan Wei, Yang Du, Garry Wong, Yuyong Tao, Gang Liu, Zongwen Jin, Jun Chu. A Genetically Encoded Bioluminescent System for Fast and Highly Sensitive Detection of Antibodies with a Bright Green Fluorescent Protein. ACS Nano 2021, 15
(11)
, 17602-17612. https://doi.org/10.1021/acsnano.1c05164
- Marcel V. Alavi. OMA1 High-Throughput Screen Reveals Protease Activation by Kinase Inhibitors. ACS Chemical Biology 2021, 16
(11)
, 2202-2211. https://doi.org/10.1021/acschembio.1c00350
- Muyang Guan, Mikael V. Garabedian, Marcel Leutenegger, Benjamin S. Schuster, Matthew C. Good, Daniel A. Hammer. Incorporation and Assembly of a Light-Emitting Enzymatic Reaction into Model Protein Condensates. Biochemistry 2021, 60
(42)
, 3137-3151. https://doi.org/10.1021/acs.biochem.1c00373
- Mitsuru Hattori, Nae Sugiura, Tetsuichi Wazawa, Tomoki Matsuda, Takeharu Nagai. Ratiometric Bioluminescent Indicator for a Simple and Rapid Measurement of Thrombin Activity Using a Smartphone. Analytical Chemistry 2021, 93
(40)
, 13520-13526. https://doi.org/10.1021/acs.analchem.1c02396
- Yu Pang, Hao Zhang, Hui-wang Ai. Genetically Encoded Fluorescent Redox Indicators for Unveiling Redox Signaling and Oxidative Toxicity. Chemical Research in Toxicology 2021, 34
(8)
, 1826-1845. https://doi.org/10.1021/acs.chemrestox.1c00149
- Jonathan D. Mortison, Ivan Cornella-Taracido, Gireedhar Venkatchalam, Anthony W. Partridge, Nirodhini Siriwardana, Simon M. Bushell. Rapid Evaluation of Small Molecule Cellular Target Engagement with a Luminescent Thermal Shift Assay. ACS Medicinal Chemistry Letters 2021, 12
(8)
, 1288-1294. https://doi.org/10.1021/acsmedchemlett.1c00276
- Álvaro Luque-Uría, Riikka Peltomaa, Tarja K. Nevanen, Henri O. Arola, Kristiina Iljin, Elena Benito-Peña, María C. Moreno-Bondi. Recombinant Peptide Mimetic NanoLuc Tracer for Sensitive Immunodetection of Mycophenolic Acid. Analytical Chemistry 2021, 93
(29)
, 10358-10364. https://doi.org/10.1021/acs.analchem.1c02109
- Sim Yee Chen, Melissa Harrison, Eng Khoon Ng, Dominic Sauvageau, Anastasia Elias. Immobilized Reporter Phage on Electrospun Polymer Fibers for Improved Capture and Detection of Escherichia coli O157:H7. ACS Food Science & Technology 2021, 1
(6)
, 1085-1094. https://doi.org/10.1021/acsfoodscitech.1c00101
- Hyukjun Choi, Soomin Eom, Han-ul Kim, Yoonji Bae, Hyun Suk Jung, Sebyung Kang. Load and Display: Engineering Encapsulin as a Modular Nanoplatform for Protein-Cargo Encapsulation and Protein-Ligand Decoration Using Split Intein and SpyTag/SpyCatcher. Biomacromolecules 2021, 22
(7)
, 3028-3039. https://doi.org/10.1021/acs.biomac.1c00481
- Riho Takahashi, Takanobu Yasuda, Yuki Ohmuro-Matsuyama, Hiroshi Ueda. BRET Q-Body: A Ratiometric Quench-based Bioluminescent Immunosensor Made of Luciferase–Dye–Antibody Fusion with Enhanced Response. Analytical Chemistry 2021, 93
(21)
, 7571-7578. https://doi.org/10.1021/acs.analchem.0c05217
- Brittany R. Benlian, Pavel E. Z. Klier, Kayli N. Martinez, Marie K. Schwinn, Thomas A. Kirkland, Evan W. Miller. Small Molecule–Protein Hybrid for Voltage Imaging via Quenching of Bioluminescence. ACS Sensors 2021, 6
(5)
, 1857-1863. https://doi.org/10.1021/acssensors.1c00058
- Eleonora Comeo, Phuc Trinh, Anh T. Nguyen, Cameron J. Nowell, Nicholas D. Kindon, Mark Soave, Leigh A. Stoddart, Jonathan M. White, Stephen J. Hill, Barrie Kellam, Michelle L. Halls, Lauren T. May, Peter J. Scammells. Development and Application of Subtype-Selective Fluorescent Antagonists for the Study of the Human Adenosine A1 Receptor in Living Cells. Journal of Medicinal Chemistry 2021, 64
(10)
, 6670-6695. https://doi.org/10.1021/acs.jmedchem.0c02067
- Kwangho Kim, Plamen P. Christov, Ian Romaine, Jianhua Tian, Somnath Jana, Alexander P. Lamers, Brendan F. Dutter, Toya Scaggs, Kyouk Jeon, Benjamin Guttentag, C. David Weaver, Craig W. Lindsley, Alex G. Waterson, Gary A. Sulikowski. Ten-Year Retrospective of the Vanderbilt Institute of Chemical Biology Chemical Synthesis Core. ACS Chemical Biology 2021, 16
(5)
, 787-793. https://doi.org/10.1021/acschembio.0c00818
- Masaharu Somiya, Shun’ichi Kuroda. Real-Time Luminescence Assay for Cytoplasmic Cargo Delivery of Extracellular Vesicles. Analytical Chemistry 2021, 93
(13)
, 5612-5620. https://doi.org/10.1021/acs.analchem.1c00339
- Mary P. Hall, Virginia A. Kincaid, Emily A. Jost, Thomas P. Smith, Robin Hurst, Stuart K. Forsyth, Connor Fitzgerald, Valerie T. Ressler, Kris Zimmermann, Dan Lazar, Monika G. Wood, Keith V. Wood, Thomas A. Kirkland, Lance P. Encell, Thomas Machleidt, Melanie L. Dart. Toward a Point-of-Need Bioluminescence-Based Immunoassay Utilizing a Complete Shelf-Stable Reagent. Analytical Chemistry 2021, 93
(12)
, 5177-5184. https://doi.org/10.1021/acs.analchem.0c05074
- Yukino Itoh, Mitsuru Hattori, Tetsuichi Wazawa, Yoshiyuki Arai, Takeharu Nagai. Ratiometric Bioluminescent Indicator for Simple and Rapid Diagnosis of Bilirubin. ACS Sensors 2021, 6
(3)
, 889-895. https://doi.org/10.1021/acssensors.0c02000
- Kaitlyn Bacon, Abigail Blain, John Bowen, Matthew Burroughs, Nikki McArthur, Stefano Menegatti, Balaji M. Rao. Quantitative Yeast–Yeast Two Hybrid for the Discovery and Binding Affinity Estimation of Protein–Protein Interactions. ACS Synthetic Biology 2021, 10
(3)
, 505-514. https://doi.org/10.1021/acssynbio.0c00472
- Elisa Lázaro-Ibáñez, Farid N. Faruqu, Amer F. Saleh, Andreia M. Silva, Julie Tzu-Wen Wang, Janusz Rak, Khuloud T. Al-Jamal, Niek Dekker. Selection of Fluorescent, Bioluminescent, and Radioactive Tracers to Accurately Reflect Extracellular Vesicle Biodistribution in Vivo. ACS Nano 2021, 15
(2)
, 3212-3227. https://doi.org/10.1021/acsnano.0c09873
- Aida Shahraki, Ali Işbilir, Berna Dogan, Martin J. Lohse, Serdar Durdagi, Necla Birgul-Iyison. Structural and Functional Characterization of Allatostatin Receptor Type-C of Thaumetopoea pityocampa, a Potential Target for Next-Generation Pest Control Agents. Journal of Chemical Information and Modeling 2021, 61
(2)
, 715-728. https://doi.org/10.1021/acs.jcim.0c00985
- Andrea Peier, Lan Ge, Nicolas Boyer, John Frost, Ruchia Duggal, Kaustav Biswas, Scott Edmondson, Jeffrey D. Hermes, Lin Yan, Chad Zimprich, Ahmad Sadruddin, Hung Yi Kristal Kaan, Arun Chandramohan, Christopher J. Brown, Dawn Thean, Xue Er Lee, Tsz Ying Yuen, Fernando J. Ferrer-Gago, Charles W. Johannes, David P. Lane, Brad Sherborne, Cesear Corona, Matthew B. Robers, Tomi K. Sawyer, Anthony W. Partridge. NanoClick: A High Throughput, Target-Agnostic Peptide Cell Permeability Assay. ACS Chemical Biology 2021, 16
(2)
, 293-309. https://doi.org/10.1021/acschembio.0c00804
- Adam D. Cotton, Duy P. Nguyen, Josef A. Gramespacher, Ian B. Seiple, James A. Wells. Development of Antibody-Based PROTACs for the Degradation of the Cell-Surface Immune Checkpoint Protein PD-L1. Journal of the American Chemical Society 2021, 143
(2)
, 593-598. https://doi.org/10.1021/jacs.0c10008
- Ryo Nishihara, Kazuki Niwa, Tatsunosuke Tomita, Ryoji Kurita. Coelenterazine Analogue with Human Serum Albumin-Specific Bioluminescence. Bioconjugate Chemistry 2020, 31
(12)
, 2679-2684. https://doi.org/10.1021/acs.bioconjchem.0c00536
- Hope Adamson, Lars J. C. Jeuken. Engineering Protein Switches for Rapid Diagnostic Tests. ACS Sensors 2020, 5
(10)
, 3001-3012. https://doi.org/10.1021/acssensors.0c01831
- Emily J. Hanan, Jun Liang, Xiaojing Wang, Robert A. Blake, Nicole Blaquiere, Steven T. Staben. Monomeric Targeted Protein Degraders. Journal of Medicinal Chemistry 2020, 63
(20)
, 11330-11361. https://doi.org/10.1021/acs.jmedchem.0c00093
- Scott A. Busby, Seth Carbonneau, John Concannon, Christoph E. Dumelin, YounKyoung Lee, Shin Numao, Nicole Renaud, Thomas M. Smith, Douglas S. Auld. Advancements in Assay Technologies and Strategies to Enable Drug Discovery. ACS Chemical Biology 2020, 15
(10)
, 2636-2648. https://doi.org/10.1021/acschembio.0c00495
- Chee Ka Candice Lam, Kevin Truong. Engineering a Synthesis-Friendly Constitutive Promoter for Mammalian Cell Expression. ACS Synthetic Biology 2020, 9
(10)
, 2625-2631. https://doi.org/10.1021/acssynbio.0c00310
- Atsushi Ogawa, Yu Itoh. In Vitro Selection of RNA Aptamers Binding to Nanosized DNA for Constructing Artificial Riboswitches. ACS Synthetic Biology 2020, 9
(10)
, 2648-2655. https://doi.org/10.1021/acssynbio.0c00384
- Hannah S. Zurier, Michelle M. Duong, Julie M. Goddard, Sam R. Nugen. Engineering Biorthogonal Phage-Based Nanobots for Ultrasensitive, In Situ Bacteria Detection. ACS Applied Bio Materials 2020, 3
(9)
, 5824-5831. https://doi.org/10.1021/acsabm.0c00546
- James M. Wagstaff, Matthew Balmforth, Nick Lewis, Rachel Dods, Catherine Rowland, Katerine van Rietschoten, Liuhong Chen, Helen Harrison, Michael J. Skynner, Michael Dawson, Gabriela Ivanova-Berndt, Paul Beswick. An Assay for Periplasm Entry Advances the Development of Chimeric Peptide Antibiotics. ACS Infectious Diseases 2020, 6
(9)
, 2355-2361. https://doi.org/10.1021/acsinfecdis.0c00389
- Feng Wang, Zhen-Feng Li, Yuan-Yuan Yang, De-Bin Wan, Natalia Vasylieva, Yu-Qi Zhang, Jun Cai, Hong Wang, Yu-Dong Shen, Zhen-Lin Xu, Bruce D. Hammock. Chemiluminescent Enzyme Immunoassay and Bioluminescent Enzyme Immunoassay for Tenuazonic Acid Mycotoxin by Exploitation of Nanobody and Nanobody–Nanoluciferase Fusion. Analytical Chemistry 2020, 92
(17)
, 11935-11942. https://doi.org/10.1021/acs.analchem.0c02338
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
References
This article references 38 other publications.
- 1Widder, E. A. (2010) Bioluminescence in the ocean: origins of biological, chemical, and ecological diversity Science 328, 704– 7081https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXlsFCitbg%253D&md5=c07748c8f13482b1ae8362cec0ffe5e1Bioluminescence in the ocean: Origins of biological, chemical, and ecological diversityWidder, E. A.Science (Washington, DC, United States) (2010), 328 (5979), 704-708CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)A review. From bacteria to fish, a remarkable variety of marine life depends on bioluminescence (the chem. generation of light) for finding food, attracting mates, and evading predators. Disparate biochem. systems and diverse phylogenetic distribution patterns of light-emitting organisms highlight the ecol. benefits of bioluminescence, with biochem. and genetic analyses providing new insights into the mechanisms of its evolution. The origins and functions of some bioluminescent systems, however, remain obscure. Here, the author reviews recent advances in understanding bioluminescence in the ocean and highlights future research efforts that will unite mol. details with ecol. and evolutionary relations.
- 2Melnick, J. S., Janes, J., Kim, S., Chang, J. Y., Sipes, D. G., Gunderson, D., Jarnes, L., Matzen, J. T., Garcia, M. E., Hood, T. L., Beigi, R., Xia, G., Harig, R. A., Asatryan, H., Yan, S. F., Zhou, Y., Gu, X. J., Saadat, A., Zhou, V., King, F. J., Shaw, C. M., Su, A. I., Downs, R., Gray, N. S., Schultz, P. G., Warmuth, M., and Caldwell, J. S. (2006) An efficient rapid system for profiling the cellular activities of molecular libraries Proc. Natl. Acad. Sci. U.S.A. 103, 3153– 3158There is no corresponding record for this reference.
- 3Doshi, U. and Li, A. P. (2011) Luciferin IPA-based higher throughput human hepatocyte screening assays for CYP3A4 inhibition and induction J. Biomol. Screen. 16, 903– 9093https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXht1yitrnM&md5=2533a5fda46939de0805661c6b61a090Luciferin IPA-based higher throughput human hepatocyte screening assays for CYP3A4 inhibition and inductionDoshi, Utkarsh; Li, Albert P.Journal of Biomolecular Screening (2011), 16 (8), 903-909CODEN: JBISF3; ISSN:1087-0571. (Sage Publications)The authors report here higher throughput screening (HTS) assays for the evaluation of CYP3A4 inhibition and CYP3A4 induction in human hepatocytes using a novel CYP3A4 substrate, luciferin IPA (LIPA). Using human recombinant CYP450 isoforms, LIPA was found to be metabolized extensively by CYP3A4 but not by CYP1A2, CYP2C9, CYP2C19, CYP2D6, or CYP2E1. In the 384-well plate CYP3A4 inhibition assay, the known inhibitors 1-aminobenzotriazole, erythromycin, ketoconazole, and verapamil were found to cause extensive (max. inhibition of >80%), dose-dependent, statistically significant inhibition of LIPA metab. The non-CYP3A4 inhibitors diethyldithiocarbamate, quercetin, quinidine, sulfaphenazole, ticlopidine, and tranylcypromine were found to have substantially lower (max. inhibition of <50%) or no apparent inhibitory effects in the HTS assay. In the 96-well plate induction assay, the CYP3A4 inducers rifampin, phenobarbital, carbamazepine, phenytoin, troglitazone, rosiglitazone, and pioglitazone yielded dose-dependent induction of LIPA metab., whereas the CYP1A2 inducers omeprazole and 3-methylcholanthrene did not display any induction in the CYP3A4 activity. The high sensitivity and specificity of the assays, the relative ease of execution, and reduced cost, time, and test material requirements suggest that the HTS assays may be applied routinely for screening a large no. of chems. in the drug discovery phase for CYP3A4 inhibitory and inducing potential.
- 4Smirnova, N. A., Haskew-Layton, R. E., Basso, M., Hushpulian, D. M., Payappilly, J. B., Speer, R. E., Ahn, Y. H., Rakhman, I., Cole, P. A., Pinto, J. T., Ratan, R. R., and Gazaryan, I. G. (2011) Development of Neh2-luciferase reporter and its application for high throughput screening and real-time monitoring of Nrf2 activators Chem. Biol. 18, 752– 7654https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXotVejtbc%253D&md5=3919f582cce67396e5b869ce7b9e0325Development of Neh2-Luciferase Reporter and Its Application for High Throughput Screening and Real-Time Monitoring of Nrf2 ActivatorsSmirnova, Natalya A.; Haskew-Layton, Renee E.; Basso, Manuela; Hushpulian, Dmitry M.; Payappilly, Jimmy B.; Speer, Rachel E.; Ahn, Young-Hoon; Rakhman, Ilay; Cole, Philip A.; Pinto, John T.; Ratan, Rajiv R.; Gazaryan, Irina G.Chemistry & Biology (Cambridge, MA, United States) (2011), 18 (6), 752-765CODEN: CBOLE2; ISSN:1074-5521. (Cell Press)Summary: The NF-E2-related factor 2 (Nrf2) is a key transcriptional regulator of antioxidant defense and detoxification. To directly monitor stabilization of Nrf2, we fused its Neh2 domain, responsible for the interaction with its nucleocytoplasmic regulator, Keap1, to firefly luciferase (Neh2-luciferase). We show that Neh2 domain is sufficient for recognition, ubiquitination, and proteasomal degrdn. of Neh2-luciferase fusion protein. The Neh2-luc reporter system allows direct monitoring of the adaptive response to redox stress and classification of drugs based on the time course of reporter activation. The reporter was used to screen the Spectrum library of 2000 biol. active compds. to identify activators of Nrf2. The most robust and yet nontoxic Nrf2 activators found-nordihydroguaiaretic acid, fisetin, and gedunin-induced astrocyte-dependent neuroprotection from oxidative stress via an Nrf2-dependent mechanism.
- 5Perroy, J., Pontier, S., Charest, P. G., Aubry, M., and Bouvier, M. (2004) Real-time monitoring of ubiquitination in living cells by BRET Nat. Methods 1, 203– 2085https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXisVGhs74%253D&md5=ae20928d2b7c8208099f526431f89154Real-time monitoring of ubiquitination in living cells by BRETPerroy, Julie; Pontier, Stephanie; Charest, Pascale G.; Aubry, Muriel; Bouvier, MichelNature Methods (2004), 1 (3), 203-208CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)Ubiquitin has emerged as an important regulator of protein stability and function in organisms ranging from yeast to mammals. The ability to detect in situ changes in protein ubiquitination without perturbing the physiol. environment of cells would be a major step forward in understanding the ubiquitination process and its consequences. Here, we describe a new method to study this dynamic post-translational modification in intact human embryonic kidney cells. Using bioLuminescence resonance energy transfer (BRET), we measured the ubiquitination of β-arrestin 2, a regulatory protein implicated in the modulation of G protein-coupled receptors. In addn. to allowing the detection of basal and GPCR-regulated ubiquitination of p-arrestin 2 in living cells, real-time BRET measurements permitted the recording of distinct ubiquitination kinetics that are dictated by the identity of the activated receptor. The ubiquitination BRET assay should prove to be a useful tool for studying the dynamic ubiquitination of proteins and for understanding which cellular functions are regulated by this post-translational event.
- 6Fan, F., Binkowski, B. F., Butler, B. L., Stecha, P. F., Lewis, M. K., and Wood, K. V. (2008) Novel genetically encoded biosensors using firefly luciferase ACS Chem. Biol. 3, 346– 3516https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXntlKrsrY%253D&md5=59474ab5133cadb6752eed96e89fe96cNovel Genetically Encoded Biosensors Using Firefly LuciferaseFan, Frank; Binkowski, Brock F.; Butler, Braeden L.; Stecha, Peter F.; Lewis, Martin K.; Wood, Keith V.ACS Chemical Biology (2008), 3 (6), 346-351CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)Genetically encoded biosensors have proven valuable for real-time monitoring of intracellular phenomena, particularly FRET-based sensors incorporating variants of green fluorescent protein. To increase detection sensitivity and response dynamics, the authors genetically engineered firefly luciferase to detect specific intermol. interactions through modulation of its luminescence activity. This concept has been applied in covalent, noncovalent, and allosteric design configurations. The covalent design gives sensitive detection of protease activity through a cleavage-dependent increase in luminescence. The noncovalent and allosteric designs allow reversible detection of the small mols. rapamycin and cAMP, resp. These sensors allow detection of mol. processes within living cells following addn. of the luciferin substrate to the growth medium. For example, the cAMP sensor allows monitoring of intracellular signal transduction assocd. with G-protein coupled receptor function. These and other luminescent biosensors will be useful for the sensitive detection of cellular physiol. in research and drug discovery.
- 7Remy, I. and Michnick, S. W. (2006) A highly sensitive protein-protein interaction assay based on Gaussia luciferase Nat. Methods 3, 977– 9797https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xht1Wqu77I&md5=6d80a3eded2f378506dff827c5af3e04A highly sensitive protein-protein interaction assay based on Gaussia luciferaseRemy, Ingrid; Michnick, Stephen W.Nature Methods (2006), 3 (12), 977-979CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)Protein-fragment complementation assays (PCAs) provide a general strategy to study the dynamics of protein-protein interactions in vivo and in vitro. The full potential of PCA requires assays that are fully reversible and sensitive at subendogenous protein expression levels. We describe a new assay that meets these criteria, based on the Gaussia princeps luciferase enzyme, demonstrating chem. reversal, and induction and inhibition of a key interaction linking insulin and TGFβ signaling.
- 8Tannous, B. A., Kim, D. E., Fernandez, J. L., Weissleder, R., and Breakefield, X. O. (2005) Codon-optimized Gaussia luciferase cDNA for mammalian gene expression in culture and in vivo Mol. Ther. 11, 435– 4438https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXhsVOju7w%253D&md5=bc4ba7b78ea19d52184ebf6415cc410bCodon-optimized Gaussia luciferase cDNA for mammalian gene expression in culture and in vivoTannous, Bakhos A.; Kim, Dong-Eog; Fernandez, Juliet L.; Weissleder, Ralph; Breakefield, Xandra O.Molecular Therapy (2005), 11 (3), 435-443CODEN: MTOHCK; ISSN:1525-0016. (Elsevier)Photoproteins have played a major role in advancing our understanding of biol. processes. A broader array of biocompatible, nontoxic, and novel reporters can serve to expand this potential. Here we describe the properties of a luciferase from the copepod marine organism Gaussia princeps. It is a monomeric protein composed of 185 aa (19.9 kDa) with a short coding sequence (555 bp) making it suitable for viral vectors. The humanized form of Gaussia luciferase (hGLuc) was efficiently expressed in mammalian cells following delivery by HSV-1 amplicon vectors. It was found to be nontoxic and naturally secreted, with flash bioluminescence characteristics similar to those of other coelenterazine luciferases. HGLuc generated over 1000-fold higher bioluminescent signal intensity from live cells together with their immediate environment and over 100-fold higher intensity from viable cells alone (not including secreted luciferase) or cell lysates, compared to humanized forms of firefly (hFLuc) and Renilla (hRLuc) luciferases expressed under similar conditions. Furthermore, hGLuc showed 200-fold higher signal intensity than hRLuc and intensity comparable to that of hFLuc in vivo under std. imaging conditions. Gaussia luciferase provides a sensitive means of imaging gene delivery and other events in living cells in culture and in vivo, with a unique combination of features including high signal intensity, secretion, and ATP independence, thus being able to report from the cells and their environment in real time.
- 9Markova, S. V., Golz, S., Frank, L. A., Kalthof, B., and Vysotski, E. S. (2004) Cloning and expression of cDNA for a luciferase from the marine copepod Metridia longa. A novel secreted bioluminescent reporter enzyme J. Biol. Chem. 279, 3212– 3217There is no corresponding record for this reference.
- 10Nakajima, Y., Kobayashi, K., Yamagishi, K., Enomoto, T., and Ohmiya, Y. (2004) cDNA cloning and characterization of a secreted luciferase from the luminous Japanese ostracod, Cypridina noctiluca Biosci. Biotechnol. Biochem. 68, 565– 570There is no corresponding record for this reference.
- 11Suzuki, C., Nakajima, Y., Akimoto, H., Wu, C., and Ohmiya, Y. (2005) A new additional reporter enzyme, dinoflagellate luciferase, for monitoring of gene expression in mammalian cells Gene 344, 61– 66There is no corresponding record for this reference.
- 12Inouye, S., Watanabe, K., Nakamura, H., and Shimomura, O. (2000) Secretional luciferase of the luminous shrimp Oplophorus gracilirostris: cDNA cloning of a novel imidazopyrazinone luciferase FEBS Lett. 481, 19– 25There is no corresponding record for this reference.
- 13Wurdinger, T., Badr, C., Pike, L., de Kleine, R., Weissleder, R., Breakefield, X. O., and Tannous, B. A. (2008) A secreted luciferase for ex vivo monitoring of in vivo processes Nat. Methods 5, 171– 17313https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhsV2ksLw%253D&md5=e0cc4226e91256ae0e481f84ab4f3a77A secreted luciferase for ex vivo monitoring of in vivo processesWurdinger, Thomas; Badr, Christian; Pike, Lisa; de Kleine, Ruben; Weissleder, Ralph; Breakefield, Xandra O.; Tannous, Bakhos A.Nature Methods (2008), 5 (2), 171-173CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)Luciferases are widely used to monitor biol. processes. Here we describe the naturally secreted Gaussia princeps luciferase (Gluc) as a highly sensitive reporter for quant. assessment of cells in vivo by measuring its concn. in blood. The Gluc blood assay complements in vivo bioluminescence imaging, which has the ability to localize the signal and provides a multifaceted assessment of cell viability, proliferation and location in exptl. disease and therapy models.
- 14Andreu, N., Zelmer, A., Fletcher, T., Elkington, P. T., Ward, T. H., Ripoll, J., Parish, T., Bancroft, G. J., Schaible, U., Robertson, B. D., and Wiles, S. (2010) Optimisation of bioluminescent reporters for use with mycobacteria PLoS One 5, e10777There is no corresponding record for this reference.
- 15Shimomura, O., Masugi, T., Johnson, F. H., and Haneda, Y. (1978) Properties and reaction mechanism of the bioluminescence system of the deep-sea shrimp Oplophorus gracilirostris Biochemistry 17, 994– 998There is no corresponding record for this reference.
- 16Kurowski, M. A. and Bujnicki, J. M. (2003) GeneSilico protein structure prediction meta-server Nucleic Acids Res. 31, 3305– 3307There is no corresponding record for this reference.
- 17Flower, D. R., North, A. C., and Sansom, C. E. (2000) The lipocalin protein family: structural and sequence overview Biochim. Biophys. Acta 1482, 9– 24There is no corresponding record for this reference.
- 18Loening, A. M., Fenn, T. D., Wu, A. M., and Gambhir, S. S. (2006) Consensus guided mutagenesis of Renilla luciferase yields enhanced stability and light output Protein Eng., Des. Sel. 19, 391– 40018https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xotl2ksro%253D&md5=c006a4ff7a42c04c1ccf02b80fb1c9ffConsensus guided mutagenesis of Renilla luciferase yields enhanced stability and light outputLoening, Andreas Markus; Fenn, Timothy David; Wu, Anna M.; Gambhir, Sanjiv SamProtein Engineering, Design & Selection (2006), 19 (9), 391-400CODEN: PEDSBR; ISSN:1741-0126. (Oxford University Press)Luciferases, which have seen expansive employment as reporter genes in biol. research, could also be used in applications where the protein itself is conjugated to ligands to create probes that are appropriate for use in small animal imaging. As the bioluminescence activity of commonly used luciferases is too labile in serum to permit this application, specific mutations of Renilla luciferase, selected using a consensus sequence driven strategy, were screened for their ability to confer stability of activity in serum as well as their light output. Using this information, a total of eight favorable mutations were combined to generate a mutant Renilla luciferase (RLuc8) that, compared with the parental enzyme, is 200-fold more resistant to inactivation in murine serum and exhibits a 4-fold improvement in light output. Results of the mutational anal. were also used to generate a double mutant optimized for use as a reporter gene. The double mutant had half the resistance to inactivation in serum of the native enzyme while yielding a 5-fold improvement in light output.These variants of Renilla luciferase, which exhibit significantly improved properties compared with the native enzyme, will allow enhanced sensitivity in existing luciferase-based assays as well as enable the development of novel probes labeled with the luciferase protein.
- 19Woo, J. and von Arnim, A. G. (2008) Mutational optimization of the coelenterazine-dependent luciferase from Renilla Plant Methods 4, 23There is no corresponding record for this reference.
- 20Branchini, B. R., Ablamsky, D. M., Davis, A. L., Southworth, T. L., Butler, B., Fan, F., Jathoul, A. P., and Pule, M. A. (2010) Red-emitting luciferases for bioluminescence reporter and imaging applications Anal. Biochem. 396, 290– 297There is no corresponding record for this reference.
- 21Inouye, S. and Shimomura, O. (1997) The use of Renilla luciferase, Oplophorus luciferase, and Apoaequorin as bioluminescent reporter protein in the presence of coelenterazine analogues as substrate Biochem. Biophys. Res. Commun. 233, 349– 353There is no corresponding record for this reference.
- 22Ando, Y., Niwa, K., Yamada, N., Enomoto, T., Irie, T., Kubota, H., Ohmiya, Y., and Akiyama, H. (2008) Firefly bioluminescence quantum yield and colour change by pH-sensitive green emission Nat. Photonics 2, 44– 4722https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXmtVSnuw%253D%253D&md5=00549500c02c8644684b672d7530e863Firefly bioluminescence quantum yield and colour change by pH-sensitive green emissionAndo, Yoriko; Niwa, Kazuki; Yamada, Nobuyuki; Enomoto, Toshiteru; Irie, Tsutomu; Kubota, Hidehiro; Ohmiya, Yoshihiro; Akiyama, HidefumiNature Photonics (2008), 2 (1), 44-47CODEN: NPAHBY; ISSN:1749-4885. (Nature Publishing Group)Firefly bioluminescence is the most well-known ideal photoemitter system in biophotonics, known in particular for its extremely high quantum yield, 88 ± 25% (refs. 2,3) or higher, and its magnificent pH-dependent emission-color change between yellow-green and red, modelled as the chem. equil. between two corresponding states. However, the need for re-examn. has also been discussed. In this letter we quantify quantum yields and color changes using our new total-photon-flux spectrometer. We det. the highest quantum yield to be 41.0 ± 7.4% (1 std. deviation (s.d.) est., coverage factor k = 1), and find that bioluminescence spectra are systematically decompd. into one pH-sensitive and two pH-insensitive gaussian components. There is no intensity conversion between yellow-green and red emissions through pH equil., but simple intensity variation of the pH-sensitive gaussian peak at 2.2 eV causes the changes in emission colors. This represents a paradigm shift in the concept of color detn. from long-standing interpretation based on pH equil.
- 23Davis, R. E., Zhang, Y. Q., Southall, N., Staudt, L. M., Austin, C. P., Inglese, J., and Auld, D. S. (2007) A cell-based assay for IκBα stabilization using a two-color dual luciferase-based sensor Assay Drug Dev. Technol. 5, 85– 103There is no corresponding record for this reference.
- 24Almond, B., Hawkins, E., Stecha, P., Garvin, D., Paguio, A., Butler, B. L., Beck, M., Wood, M., and Wood, K. (2003) Introducing ChromaLuc technology Promega Notes 85, 11– 14There is no corresponding record for this reference.
- 25Swanson, B., Fan, F., and Wood, K. V. (2007) Enhanced response dynamics for transcription analysis using new pGL4 luciferase reporter vectors Cell Notes 17, 3– 5There is no corresponding record for this reference.
- 26Auld, D. S., Zhang, Y. Q., Southall, N. T., Rai, G., Landsman, M., MacLure, J., Langevin, D., Thomas, C. J., Austin, C. P., and Inglese, J. (2009) A basis for reduced chemical library inhibition of firefly luciferase obtained from directed evolution J. Med. Chem. 52, 1450– 145826https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhvVWnt7o%253D&md5=aac37056be38a2c0ed06098d2e2f142aA Basis for Reduced Chemical Library Inhibition of Firefly Luciferase Obtained from Directed EvolutionAuld, Douglas S.; Zhang, Ya-Qin; Southall, Noel T.; Rai, Ganesha; Landsman, Marc; MacLure, Jennifer; Langevin, Daniel; Thomas, Craig J.; Austin, Christopher P.; Inglese, JamesJournal of Medicinal Chemistry (2009), 52 (5), 1450-1458CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)We measured the "druggability" of the ATP-dependent luciferase derived from the firefly Photuris pennsylvanica that was optimized using directed evolution (Ultra-Glo, Promega). Quant. high-throughput screening (qHTS) was used to det. IC50s of 198899 samples against a formulation of Ultra-Glo luciferase (Kinase-Glo). We found that only 0.1% of the Kinase-Glo inhibitors showed an IC50 < 10 μM compared to 0.9% found from a previous qHTS against the firefly luciferase from Photinus pyralis (lucPpy). Further, the max. affinity identified in the lucPpy qHTS was 50 nM, while for Kinase-Glo this value increased to 600 nM. Compds. with interactions stretching outside the luciferin binding pocket were largely lost with Ultra-Glo luciferase. Therefore, Ultra-Glo luciferase will show less compd. interference when used as an ATP sensor compared to lucPpy. This study demonstrates the power of large-scale quant. anal. of structure-activity relationships (>100K compds.) in addressing important questions such as a target's druggability.
- 27Fahey, R. C., Hunt, J. S., and Windham, G. C. (1977) On the cysteine and cystine content of proteins. Differences between intracellular and extracellular proteins J. Mol. Evol. 10, 155– 160There is no corresponding record for this reference.
- 28Goerke, A. R., Loening, A. M., Gambhir, S. S., and Swartz, J. R. (2008) Cell-free metabolic engineering promotes high-level production of bioactive Gaussia princeps luciferase Metab. Eng. 10, 187– 20028https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXnt1ektbc%253D&md5=1f54e550163e57beeae2a131e48a513cCell-free metabolic engineering promotes high-level production of bioactive Gaussia princeps luciferaseGoerke, Aaron R.; Loening, Andreas M.; Gambhir, Sanjiv S.; Swartz, James R.Metabolic Engineering (2008), 10 (3-4), 187-200CODEN: MEENFM; ISSN:1096-7176. (Elsevier Inc.)Due to its small size and intense luminescent signal, Gaussia princeps luciferase (GLuc) is attractive as a potential imaging agent in both cell culture and small animal research models. However, recombinant GLuc prodn. using in vivo techniques has only produced small quantities of active luciferase, likely due to five disulfide bonds being required for full activity. Cell-free biol. provides the freedom to control both the catalyst and chem. compns. in biol. reactions, and we capitalized on this to produce large amts. of highly active GLuc in cell-free reactions. Active yields were improved by mutating the cell ext. source strain to reduce proteolysis, adjusting reaction conditions to enhance oxidative protein folding, further activating energy metab., and encouraging post-translational activation. This cell-free protein synthesis procedure produced 412 μg/mL of purified GLuc, relative to 5 μg/mL isolated for intracellular Escherichia coli expression. The cell-free product had a specific activity of 4.2×1024 photons/s/mol, the highest reported activity for any characterized luciferase.
- 29Inouye, S. and Sasaki, S. (2007) Overexpression, purification and characterization of the catalytic component of Oplophorus luciferase in the deep-sea shrimp, Oplophorus gracilirostris Protein Expression Purif. 56, 261– 26829https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXht1anurrL&md5=0e4fd0d955d54530c24027053fb1cc18Overexpression, purification and characterization of the catalytic component of Oplophorus luciferase in the deep-sea shrimp, Oplophorus gracilirostrisInouye, Satoshi; Sasaki, SatokoProtein Expression and Purification (2007), 56 (2), 261-268CODEN: PEXPEJ; ISSN:1046-5928. (Elsevier B.V.)The luciferase secreted by the deep-sea shrimp Oplophorus consists of 19 and 35 kDa proteins. The 19-kDa protein (19kOLase), the catalytic component of luminescence reaction, was expressed in Escherichia coli using the cold-shock inducted expression system. 19KOLase, expressed as inclusion bodies, was solubilized with 6 M urea and purified by urea-nickel chelate affinity chromatog. The yield of 19kOLase was 16 mg from 400 mL of cultured cells. 19kOLase in 6 M urea could be refolded rapidly by diln. with 50 mM Tris-HCl (pH 7.8)-10 mM EDTA, and the refolded protein showed luminescence activity. The luminescence properties of refolded 19kOLase were characterized, in comparison with native Oplophorus luciferase. Luminescence intensity with bisdeoxycoelenterazine as a substrate was stimulated in the presence of org. solvents. The 19kOLase is a thermolabile protein and is 98 % inhibited by 1 μM Cu2+. The cysteine residue of 19kOLase is not essential for catalysis of the luminescence reaction.
- 30Wong, G. G., Witek-Giannotti, J., Hewick, R. M., Clark, S. C., and Ogawa, M. (1988) Interleukin 6: identification as a hematopoietic colony-stimulating factor Behring Inst. Mitt. 83, 40– 4730https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL1MXitFSmur8%253D&md5=b253f52b9e95b28e7ca725a9ef526756Interleukin 6: identification as a hematopoietic colony-stimulating factorWong, G. G.; Witek-Giannotti, J.; Hewick, R. M.; Clark, S. C.; Ogawa, M.Behring Institute Mitteilungen (1988), 83 (), 40-7CODEN: BHIMA2; ISSN:0301-0457.A cDNA encoding a novel myeloid colony-stimulating activity from a human T cell line was identified and mol. cloned. This cytokine proved to be identical to the factor currently known as interleukin (IL)-6, thereby demonstrating effects of IL-6 with hematopoietic target cells. In addn. to its ability to support murine granulocyte-macrophage colony formation, IL-6 acted synergistically with IL-3 in both the murine and human systems in support of colony formation by the primitive blast cell colony forming cell. This multitude of biol. activities suggests that IL-6 plays a prominent role within a network of cytokines in integrating the different arms of the host response to infection.
- 31Lippincott-Schwartz, J., Roberts, T. H., and Hirschberg, K. (2000) Secretory protein trafficking and organelle dynamics in living cells Annu. Rev. Cell Dev. Biol. 16, 557– 58931https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXpvFyq&md5=b5cf93a3d1fd4b750c843c43780bd567Secretory protein trafficking and organelle dynamics in living cellsLippincott-Schwartz, Jennifer; Roberts, Theresa H.; Hirschberg, KoretAnnual Review of Cell and Developmental Biology (2000), 16 (), 557-589CODEN: ARDBF8; ISSN:1081-0706. (Annual Reviews Inc.)A review with many refs. Green fluorescent protein chimerae acting as reporters for protein localization and trafficking within the secretory membrane system of living cells have been used in a wide variety of applications, including time-lapse imaging, double-labeling, energy transfer, quantitation, and photobleaching expts. Results from this work are clarifying the steps involved in the formation, translocation, and fusion of transport intermediates; the organization and biogenesis of organelles; and the mechanisms of protein retention, sorting, and recycling in the secretory pathway. In so doing, they are broadening our thinking about the temporal and spatial relationships among secretory organelles and the membrane trafficking pathways that operate between them.
- 32Simmons, S. O. (2011) Fireflies in the coalmine: luciferase technologies in next-generation toxicity testing Comb. Chem. High Throughput Screening 14, 688– 70232https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtVKqtrnI&md5=1224daf1cf9b892c342b3c87d13c478bFireflies in the coalmine: luciferase technologies in next-generation toxicity testingSimmons, Steven O.Combinatorial Chemistry & High Throughput Screening (2011), 14 (8), 688-702CODEN: CCHSFU; ISSN:1386-2073. (Bentham Science Publishers Ltd.)Whole-animal studies have been the mainstay of toxicity testing for decades. These approaches are too expensive and laborious to effectively characterize all of the chems. currently in com. use. In addn., there are social and ethical pressures to reduce, refine and replace animal testing in toxicol. The National Research Council (NRC) has outlined a new strategy to transition from animal-based tests to high throughput, cell-based assays and computational modeling approaches to characterize chem. toxicants. Crit. to this vision, assays that measure toxicity pathways assocd. with adverse health effects must be developed. Bioluminescent assays are particularly well suited to the demands of next-generation toxicity testing because they measure a wide range of biol. activities in a quant. and high throughput manner. This review describes the limitations of traditional, animal-based toxicity testing and discusses the current and developing uses of bioluminescent technologies in next-generation testing based on three general assays formats: luciferase-limited assays, ATP-limited assays and luciferin-limited assays.
- 33Rehemtulla, A., Taneja, N., and Ross, B. D. (2004) Bioluminescence detection of cells having stabilized p53 in response to a genotoxic event Mol. Imaging 3, 63– 6833https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXkslGjsLc%253D&md5=e3a74f1ffe9966213c680a7668f356e3Bioluminescence detection of cells having stabilized p53 in response to a genotoxic eventRehemtulla, Alnawaz; Taneja, Neelam; Ross, Brian D.Molecular Imaging (2004), 3 (1), 63-68CODEN: MIOMBP; ISSN:1535-3508. (MIT Press)Inactivation of p53 is one of the most frequent mol. events in neoplastic transformation. Approx. 60% of all human tumors have mutations in both p53 alleles. Wild-type p53 activity is regulated in large part by the proteosome-dependent degrdn. of p53, resulting in a short p53 half-life in unstressed and untransformed cells. Activation of p53 by a variety of stimuli, including DNA damage induced by genotoxic drugs or radiation, is accomplished by stabilization of wild-type p53. The stabilized and active p53 can result in either cell-cycle arrest or apoptosis. Surprisingly, the majority of tumor-assocd., inactivating p53 mutations also result in p53 accumulation. Thus, constitutive elevation of p53 levels in cells is a reliable measure of p53 inactivation, whereas transiently increased p53 levels reflect a recent genotoxic stress. In order to facilitate noninvasive imaging of p53 accumulation, the authors here describe the construction of a p53-luciferase fusion protein. Induction of DNA damage in cells expressing, the fusion protein resulted in a time-dependent accumulation of the fusion that was noninvasively detected using bioluminescence imaging and validated by Western blot anal. The p53-Luc protein retains p53 function because its expression in HCT116 cells lacking functional p53 resulted in activation of p21 expression as well as induction of apoptosis in response to a DNA damaging event. Employed in a transgenic animal model, the proposed p53-reporter fusion protein will be useful for studying p53 activation in response to exposure to DNA-damaging carcinogenic agents. It could also be used to study p53 stabilization as a result of inactivating p53 mutations. Such studies will further the understanding of p53's role as the "guardian of the genome" and its function in tumorigenesis.
- 34Horn, H. F. and Vousden, K. H. (2007) Coping with stress: multiple ways to activate p53 Oncogene 26, 1306– 131634https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXitVKhsLY%253D&md5=2d7b83d1eee3c060185f2441868ed16dCoping with stress: multiple ways to activate p53Horn, H. F.; Vousden, K. H.Oncogene (2007), 26 (9), 1306-1316CODEN: ONCNES; ISSN:0950-9232. (Nature Publishing Group)A review. Over the years, p53 has been shown to sit at the center of an increasingly complex web of incoming stress signals and outgoing effector pathways. The no. and diversity of stress signals that lead to p53 activation illustrates the breadth of p53's remit - responding to a wide variety of potentially oncogenic insults to prevent tumor development. Interestingly, different stress signals can use different and independent pathways to activate p53, and there is some evidence that different stress signals can mediate different responses. How each of the responses to p53 contributes to inhibition of malignant progression is beginning to be clarified, with the hope that identification of responses that are key to tumor suppression will allow a more focused and effective search for new therapeutic targets. In this review, we will highlight some recently identified roles for p53 in tumor suppression, and discuss some of the numerous mechanisms through which p53 can be regulated and activated.
- 35Nishizuka, Y. (1984) The role of protein kinase C in cell surface signal transduction and tumour promotion Nature 308, 693– 69835https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL2cXitVCmtrk%253D&md5=33d139697753d3f7e10417573e131ff9The role of protein kinase C in cell surface signal transduction and tumor promotionNishizuka, YasutomiNature (London, United Kingdom) (1984), 308 (5961), 693-8CODEN: NATUAS; ISSN:0028-0836.A review with 80 refs. on the title role of protein kinase C [9026-43-1].
- 36Htun, H., Barsony, J., Renyi, I., Gould, D. L., and Hager, G. L. (1996) Visualization of glucocorticoid receptor translocation and intranuclear organization in living cells with a green fluorescent protein chimera Proc. Natl. Acad. Sci. U.S.A. 93, 4845– 4850There is no corresponding record for this reference.
- 37Nakajima, Y., Yamazaki, T., Nishii, S., Noguchi, T., Hoshino, H., Niwa, K., Viviani, V. R., and Ohmiya, Y. (2010) Enhanced beetle luciferase for high-resolution bioluminescence imaging PLoS One 5, e10011There is no corresponding record for this reference.
- 38Suzuki, T., Kondo, C., Kanamori, T., and Inouye, S. (2011) Video rate bioluminescence imaging of secretory proteins in living cells: localization, secretory frequency, and quantification Anal. Biochem. 415, 182– 189There is no corresponding record for this reference.
Supporting Information
Supporting Information
This material is available free of charge via the Internet at http://pubs.acs.org
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.