Fluorescences and Light-up RNA aptamers

Light-up RNA aptamers are fluorescent RNA molecules that become fluorescent upon binding to specific fluorophores. This concept emerged from the need for non-invasive, real-time imaging tools to study RNA molecules and their interactions within living cells. Traditional fluorescent tags like GFP (green fluorescent protein) are not suitable for RNA because they are protein-based[1]. Thus, the development of RNA-based fluorescent tags (light-up RNA aptamers) became crucial for expanding the toolkit available for RNA visualization and functional studies.

The discovery of Spinach, the first light-up RNA aptamer, by Jaffrey and colleagues in 2011 marked a significant milestone. Spinach binds to a fluorophore called DFHBI and emits green fluorescence, mimicking GFP[2]. Following Spinach, several other light-up aptamers were developed, including Mango, Broccoli, and Corn, each binding to different fluorophores and exhibiting different fluorescence properties[3,4,5]. Researchers optimize light-up RNA aptamers for improved brightness, stability, and minimal background fluorescence. This involves iterative cycles of selection and mutation to enhance the aptamer’s performance for better performance in cellular environments[6,7,8].

Light-up RNA aptamers represent a significant advancement in the study of RNA biology, offering a versatile and powerful tool for real-time imaging and functional analysis of RNA molecules in living cells[9].
drawing

Fluorogenic mechanisms

Light-up RNA aptamers contain specific binding sites that interact with small-molecule fluorophores. These fluorophores are non-fluorescent or weakly fluorescent in the absence of the aptamer. Upon binding to the aptamer, the fluorophore undergoes a conformational change that enhances its fluorescence properties. This binding typically occurs through a combination of hydrogen bonding, Van der Waals interactions, and stacking interactions within the RNA’s three-dimensional structure.

Understanding their mechanism of action involves exploring how these aptamers interact with their fluorophores to produce fluorescence, the structural basis of their function, and the principles guiding their design and optimization[10].

Twisted Intramolecular Charge Transfer (TICT)

TICT refers to a mechanism where the binding of a target induces a conformational change in the RNA aptamer, leading to the activation or enhancement of fluorescence through a twisted charge-transfer state. This state involves the separation of charge within the molecule, resulting in a geometry that promotes fluorescence.

In the unbound state, the RNA aptamer and its fluorophore may be in a non-planar or less twisted conformation with limited charge separation, leading to weak or no fluorescence. Upon binding to a specific target, the aptamer undergoes a conformational change that facilitates a twist between the donor and acceptor moieties within the fluorophore. This twist maximizes the charge separation, creating an intramolecular charge transfer state with a large dipole moment. The TICT state stabilizes the fluorophore in a way that enhances its fluorescence emission, often resulting in a red-shifted and intensified fluorescence signal compared to the non-twisted state.
drawing

Contact Quenching (CQ)

Contact quenching in light-up RNA aptamers involves the reduction or elimination of fluorescence due to the close physical proximity of a quencher molecule to the fluorophore. This process is highly dependent on the spatial arrangement and interaction between the RNA aptamer and its target.

In the absence of the target, the fluorophore may be in close proximity to a quencher moiety within the RNA aptamer, resulting in minimal fluorescence. Binding of the target induces a conformational change in the RNA aptamer that separates the fluorophore from the quencher. The increased distance between the fluorophore and quencher reduces the efficiency of non-radiative energy transfer, allowing the fluorophore to emit fluorescence. The efficiency of contact quenching and subsequent fluorescence recovery can be modulated by the nature of the target-aptamer interaction and the specific design of the aptamer.

Light-up RNA aptamers exploiting contact quenching are used in biosensors to detect various biomolecules by monitoring changes in fluorescence.These aptamers facilitate the detection and quantification of analytes in complex mixtures.
drawing

Spirolactonization (SP)

Spirolactonization in light-up RNA aptamers involves the formation of a spirolactone ring structure upon binding to a target. This structural change enhances the fluorescence of the aptamer, providing a clear signal upon target recognition.

In the absence of the target, the RNA aptamer remains in a conformation that does not support spirolactone formation, leading to weak or no fluorescence. Binding of the target induces a conformational rearrangement in the aptamer, promoting the formation of a spirolactone ring. The formation of the spirolactone ring stabilizes the fluorophore in a fluorescent state, significantly enhancing its emission properties. The structural rigidity and planarity introduced by the spirolactone ring reduce non-radiative decay pathways, leading to a bright fluorescence signal.

Spirolactonization-based light-up RNA aptamers are used to detect specific targets with high sensitivity and specificity. These aptamers can serve dual functions in theranostics, providing both diagnostic imaging and therapeutic action.
drawing


Properties of Fluorophore-Aptamer pairs

Each light-up RNA aptamer is usually specific to a particular fluorophore. The specificity arises from the precise shape and chemical environment of the binding pocket within the RNA. The sequence and structure of the aptamer dictate the compatibility with the fluorophore, ensuring selective binding and fluorescence activation. Researchers focused on optimizing these aptamers for improved brightness, stability, and minimal background fluorescence. This included engineering the aptamers and fluorophores for better performance in cellular environments.

The table below lists several fluorescent small molecular-RNA pairings for which interaction patterns have been known through crystallographic studies or NMR. (The table only shows the representative individuals of each type of fluorescent small molecule. For more details, click on small molecule to jump to the corresponding view).

Fluorescent molecule IUPAC Molecular Formula Molar mass CAS Excitation (nm) Emission (nm) Aptamer Fluorogenic mechanisms
DFHBI (5Z)-5-[(3,5-difluoro-4-hydroxyphenyl)methylidene]-2,3-dimethylimidazol-4-one C12H10F2N2O2 252.22 g/mol 1241390-29-3 469 501 Spinach aptamer TICT
MG [4-[[4-(dimethylamino)phenyl]-phenylmethylidene]cyclohexa-2,5-dien-1-ylidene]-dimethylazanium;chloride C23H25ClN2 364.9 g/mol 569-64-2 630 655 MG aptamer TICT
HBC 530 4-[(E)-1-cyano-2-[4-[2-hydroxyethyl(methyl)amino]phenyl]ethenyl]benzonitrile C19H17N3O 303.4 g/mol 156840-13-0 485 530 Pepper aptamer TICT
TO1 4-methylbenzenesulfonate;(2Z)-3-methyl-2-[(1-methylquinolin-1-ium-4-yl)methylidene]-1,3-benzothiazole C26H24N2O3S2 476.6 g/mol 107091-89-4 510 535 Mango aptamer TICT
DFHO (5Z)-5-[(3,5-difluoro-4-hydroxyphenyl)methylidene]-3-methyl-2-(nitrosomethylidene)imidazolidin-4-one C12H9F2N3O3 281.21 g/mol 1420815-34-4 505 545 Corn aptamer TICT
DMHBI 5-[(4-hydroxy-3,5-dimethoxyphenyl)methylidene]-2,3-dimethylimidazol-4-one C14H16N2O4 276.29 g/mol 1629243-34-0 400 537 Chili aptamer TICT
YO3 (2Z)-3-methyl-2-[(E)-3-(1-methylquinolin-1-ium-4-yl)prop-2-enylidene]-1,3-benzoxazole C21H19N2O+ 315.4 g/mol NA 595 620 Mango-III aptamer TICT
ThT 4-(3,6-dimethyl-1,3-benzothiazol-3-ium-2-yl)-N,N-dimethylaniline C17H19ClN2S 318.9 g/mol 2390-54-7 455 485 Beetroot aptamer TICT
TMR 2-[3-(dimethylamino)-6-dimethylazaniumylidenexanthen-9-yl]benzoate C24H22N2O3 386.4 g/mol 120718-52-7 564 587 TMR3 aptamer CQ
DFAME methyl 3-[4-[(3,5-difluoro-4-hydroxyphenyl)methylidene]-1-methyl-5-oxoimidazol-2-yl]prop-2-enoate C15H12F2N2O4 322.26 g/mol 1420815-55-9 514 619 Beetroot aptamer TICT
OTB 3-methyl-2-[(3-methyl-1,3-benzothiazol-3-ium-2-yl)methylidene]-1,3-benzoxazole C17H15N2OS+ 295.4 g/mol NA 380 421 DIRs2-Apt aptamer TICT

References

[1] Engineering and characterization of a superfolder green fluorescent protein.
Pédelacq, J. D., Cabantous, S., Tran, T., Terwilliger, T. C., & Waldo, G. S.
Nature biotechnology, 24(1), 79–88. (2006)
[2] RNA mimics of green fluorescent protein.
Paige, J. S., Wu, K. Y., & Jaffrey, S. R.
Science (New York, N.Y.), 333(6042), 642–646. (2011)
[3] RNA mango aptamer-fluorophore: a bright, high-affinity complex for RNA labeling and tracking.
Dolgosheina, E. V., Jeng, S. C., Panchapakesan, S. S., Cojocaru, R., Chen, P. S., Wilson, P. D., Hawkins, N., Wiggins, P. A., & Unrau, P. J.
ACS chemical biology, 9(10), 2412–2420. (2014)
[4] Imaging RNA polymerase III transcription using a photostable RNA-fluorophore complex.
Song, W., Filonov, G. S., Kim, H., Hirsch, M., Li, X., Moon, J. D., & Jaffrey, S. R.
Nature chemical biology, 13(11), 1187–1194. (2017)
[5] Broccoli: rapid selection of an RNA mimic of green fluorescent protein by fluorescence-based selection and directed evolution.
Filonov, G. S., Moon, J. D., Svensen, N., & Jaffrey, S. R.
Journal of the American Chemical Society, 136(46), 16299–16308. (2014)
[6] Plug-and-play fluorophores extend the spectral properties of Spinach.
Song, W., Strack, R. L., Svensen, N., & Jaffrey, S. R.
Journal of the American Chemical Society, 136(4), 1198–1201. (2014)
[7] Fluorogenic RNA Mango aptamers for imaging small non-coding RNAs in mammalian cells.
Autour, A., C Y Jeng, S., D Cawte, A., Abdolahzadeh, A., Galli, A., Panchapakesan, S. S. S., Rueda, D., Ryckelynck, M., & Unrau, P. J.
Nature communications, 9(1), 656. (2018)
[8] Corn-Based Fluorescent Light-Up Biosensors with Improved Signal-to-Background Ratio for Label-Free Detection of Long Noncoding RNAs.
Zhang, Q., Su, C., Tian, X., & Zhang, C. Y.
Analytical chemistry, 95(20), 8097–8104. (2023)
[9] RNA Structure and Cellular Applications of Fluorescent Light-Up Aptamers.
Neubacher, S., & Hennig, S.
Angewandte Chemie (International ed. in English), 58(5), 1266–1279. (2019)
[10] Harmonizing the growing fluorogenic RNA aptamer toolbox for RNA detection and imaging.
Lu, X., Kong, K. Y. S., & Unrau, P. J.
Chemical Society reviews, 52(12), 4071–4098. (2023)