Next Article in Journal
Magnetically Separable Base Catalysts: Heterogeneous Catalysis vs. Quasi-Homogeneous Catalysis
Next Article in Special Issue
Silicon-[18F]Fluorine Radiochemistry: Basics, Applications and Challenges
Previous Article in Journal / Special Issue
Optimization of Fluorine Plasma Treatment for Interface Improvement on HfO2/In0.53Ga0.47As MOSFETs
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Nucleic Acid Based Fluorinated Derivatives: New Tools for Biomedical Applications

1
Université de Bordeaux, Bordeaux F-33076, France
2
INSERM U869, Bordeaux F-33076, France
*
Author to whom correspondence should be addressed.
Appl. Sci. 2012, 2(2), 245-259; https://doi.org/10.3390/app2020245
Submission received: 14 January 2012 / Revised: 13 March 2012 / Accepted: 14 March 2012 / Published: 23 March 2012
(This article belongs to the Special Issue Organo-Fluorine Chemical Science)

Abstract

:
Nucleic acid-based fluorinated derivatives, e.g., nucleosides or oligonucleotides connected to highly fluorinated chains or labeled with one or more fluorine atoms, have been investigated recently due to their high potential for biomedical applications. This review deals with recent works on nucleoside and oligonucleotide fluorocarbon amphiphiles as well as with properties and applications of fluorine-labeled oligonucleotide analogues.

Graphical Abstract

1. Introduction

The nucleic acid architectures stabilized by base-pairing and aryl π−π stacking interactions provides to scientists one of the most sophisticated supramolecular models. Since the determination of the structure of double-stranded DNA (dsDNA) by Watson, Crick, Wilkins and Franklin [1,2,3], artificial bio-inspired molecules featuring nucleic acid units have become a very powerful strategy to construct supramolecular systems [4,5,6]. Of particular interest is the design of hybrid amphiphilic nucleolipids featuring a bi-functionality based on the combination of nucleic acids and lipids [7,8,9,10,11,12,13,14,15]. Interestingly, different bottom-up approaches involving nucleolipids have been used for the construction of nanostructures for different applications, including materials [16,17,18] and biomedical devices [19,20,21]. In parallel to nucleolipid derivatives, the combination of nucleobase units with fluorocarbon hydrophobic and lipophobic moieties to create novel materials, for diagnostics or in medical applications, is still in its infancy. It has been extensively reported that highly fluorinated derivatives possess unique properties, including chemical and biological inertness, strong hydro- and lipophobia. Interestingly, fluorocarbon colloids used for biomedical applications feature high gas solubility, low lipid and water solubility, and outstanding surface characteristics [22].
Hence, material and biomedical sciences could take advantage of the new properties coming from the combination of nucleic acids and fluorocarbon synthetic units for new applications. The availability of chemically modified nucleotides and fluorocarbon synthons and/or reagents, means that Nucleic Acids Fluorocarbon Amphiphiles (NAFAs) could be used in numerous fields of research including engineering, biology and medicine (Figure 1). Recently, the use of nucleoside-based fluorocarbon derivatives has been mainly motivated by three fundamental objectives: (i) construct novel supramolecular materials and/or devices; (ii) develop new drug delivery systems; and (iii) find new therapeutic molecules.
Figure 1. Nucleic Acids Fluorocarbon Amphiphiles (NAFAs), including Fluorocarbon Nucleoside and OligoNucleotide amphiphiles (FNs and FONs, respectively): building blocks for soft materials and biomedical applications.
Figure 1. Nucleic Acids Fluorocarbon Amphiphiles (NAFAs), including Fluorocarbon Nucleoside and OligoNucleotide amphiphiles (FNs and FONs, respectively): building blocks for soft materials and biomedical applications.
Applsci 02 00245 g001
This short, non-exhaustive review intends to illustrate the role that highly fluorinated amphiphiles may play in chemistry, biology and material science. It consists primarily of an account of the authors’ own efforts and experience in the field, hence will be limited to some specific aspects of this chemistry. It also intends to outline the potential applications of fluorinated nucleolipids, ranging from chemistry to biomedical research.
First, we will focus on the recent progress achieved in the fields of fluorocarbon and nucleoside amphiphiles. This first section includes hybrid molecules bearing both a nucleic acid and fluorocarbon chains: namely the Fluorocarbon Nucleoside amphiphiles (FNs). Second, we will present several Fluorocarbon OligoNucleotide amphiphiles (FONs) and fluorinated oligonucleotide analogues.

2. Fluorocarbon Nucleoside Amphiphiles (FNs)

2.1. Fluorocarbon Amphiphiles

Due to their unique properties, fluorocarbon chains represent a useful building block for the construction of supramolecular systems. Perfluoroalkyl chains have a larger cross section and are more rigid than their hydrogenated counterparts. They are considerably more hydrophobic and are lipophobic as well. As a result, when dispersed in water, fluorinated amphiphiles exhibit an enhanced capacity to self-assemble into highly stable and well-defined assemblies such as films, membranes, micelles, vesicles and other stable supramolecular systems [23,24]. The self-assembling behavior of a variety of synthesized single and double chain neutral, zwitterionicor anionic fluorinated phospholipids [25,26,27,28], glycolipid [29,30], glycophospholipids, glycopeptides [31,32] was already explored (Figure 2) [33], to evaluate the impact of the fluorinated chains on the properties of the resulting aggregates. It enhances stability, low permeability to both hydrophilic and lipophilic material and reduces interaction with biological media. These fluorinated amphiphiles possess a considerable potential for the delivery of materials such as drugs, prodrugs, markers, contrast agents and nucleic acids. Recently, new perfluorinated dimerizable polycationic amphiphiles were designed by Vierling et al. to evaluate the ability of the monomer to condense DNA and of the dimer to form stable nanoparticles capable of efficient cell transfection [34]. Although formulation of small-sized cationic monomolecular DNA nanoparticles (<40 nm) with the dimerizable perfluorinated succeeded, spermine-based amphiphiles proved to be poor non-specific transfection agents in vitro. Improvements are still in progress [35,36].
Figure 2. Examples of fluorocarbon amphiphiles; (a) sugar-based fluorocarbon amphiphile [30]; (b) fluorinated amphiphilic building block used for the synthesis of glycopeptide conjugates [32]; and (c) perfluorinated dimerizable polycationic amphiphile [34].
Figure 2. Examples of fluorocarbon amphiphiles; (a) sugar-based fluorocarbon amphiphile [30]; (b) fluorinated amphiphilic building block used for the synthesis of glycopeptide conjugates [32]; and (c) perfluorinated dimerizable polycationic amphiphile [34].
Applsci 02 00245 g002

2.2. Nucleoside Amphiphiles

Nucleoside-based lipids commonly named nucleolipids [7] are hybrid molecules composed of a nucleobase, a nucleoside, a nucleotide or an oligonucleotide (either DNA or RNA), and a lipophilic moiety, which might be either simply a single- or double-chained alkyl moiety. Several improved nucleolipids have been reported hereafter (Figure 3) [37,38,39,40,41,42,43,44,45,46,47]. Most nucleolipids have a polar head group at the 5’ position and hydrophobic groups at both the 2’ and 3’ positions, patterned after natural glycerol phospholipids, which are key components of the cellular membrane. Nucleolipids were designed to interact with nucleic acids through hydrogen bonding, π–π stacking, and nucleobase recognition, as well as electrostatic interactions and the hydrophobic effect. Several cationic [12,48,49,50,51] and anionic [52,53] nucleolipids were first investigated as potential agents for the delivery of nucleic acids into cells. Indeed their self-assembled structures into lipoplexes mimic cellular membranes and their cationic properties attract both anionic oligonucleotides and negatively charged cellular membranes. New versions comprising neutral, zwitterionic, and anionic nucleolipids are currently evaluated.
Figure 3. Examples of nucleoside amphiphiles; (a) diC8PA [37]; (b) a gemini nucleotide [39]; and (c) a zwitterionic nucleolipid.
Figure 3. Examples of nucleoside amphiphiles; (a) diC8PA [37]; (b) a gemini nucleotide [39]; and (c) a zwitterionic nucleolipid.
Applsci 02 00245 g003

2.3. Hybrid Fluorocarbon Nucleoside Amphiphiles

The combination of the self-assembly potential of nucleoside amphiphiles with the hydrophobic character of highly fluorinated chains is of high relevance to prepare new self-assembled structures with tunable physico-chemical properties and functions. To assess whether fluorinated chains can enhance the scope of supramolecular assemblies formed by hybrid fluorocarbon nucleoside amphiphiles, different molecules were synthesized (Figure 4).
The fluorocarbon amphiphile 1 is based on a uridine backbone modified by a phosphocholine moiety at the 5’ position, and two 2H,2H,3H,3H-perfluoroundecanoyl side chains at the 2’ and 3’ positions. One of the goals of the study was to characterize the properties of such new compounds and to compare those results with that observed for the analogous hydrocarbon nucleoamphiphiles. In aqueous solution, fluorocarbon nucleoamphiphiles were surprisingly able to form large vesicular assemblies and unexpected lamellar phases below their Tm, contrary to hydrocarbon analogs that self-assemble into helical structures below their Tm and bilayers above it [54]. These highly hydrophobic fluorocarbon amphiphiles exhibited interesting hybrid microspherical supramolecular assemblies in the presence of thorium that were characterized by Scanning Electron Microscopy (SEM) [55].
Figure 4. Chemical structures of compounds 1-3, and corresponding electronic microscopy images of DiF17UPC/Th4+ microspheres (SEM image from [55]), and supramolecular assemblies formed by GNFs 2 and 3 in water (TEM images from [58]).
Figure 4. Chemical structures of compounds 1-3, and corresponding electronic microscopy images of DiF17UPC/Th4+ microspheres (SEM image from [55]), and supramolecular assemblies formed by GNFs 2 and 3 in water (TEM images from [58]).
Applsci 02 00245 g004
Recently, two glycosyl-nucleoside fluorinated amphiphiles (GNFs) were designed to construct new supramolecular hydrogels. Molecules 2 and 3 feature both sugar moieties attached to a thymidine base and a 2H,2H,3H,3H-perfluoroundecanoyl side chain at the 5’ position, linked via triazole bridges resulting from “click” chemistry. The self-assembly and gelation properties of such molecules were investigated. Similarly to their hydrocarbon analogs [56,57] GNFs stabilize hydrogels at low concentrations (0.1% w/w for compound 2). GNFs gelators displayed supramolecular networks of varying morphologies, such as nanofibers and helical springs, which are correlated to the presence of fluorocarbon chains. Moreover, fluorocarbon amphiphile GNF 2 proved to be not toxic for human cells (Huh7) whereas hydrocarbon analogue becomes toxic above 100 µM, demonstrating the greater potential of such fluorocarbon nucleoside amphiphiles for biomedical applications [58].

3. Fluorocarbon OligoNucleotide Amphiphiles (FONs) and Fluorinated Oligonucleotide Analogues

In the previous section, several examples of nucleoside-based fluorinated amphiphiles have been discussed for gel stabilization. In this section we will discuss on Fluorocarbon OligoNucleotide amphiphiles (FONs). The fluorine atom is likely a mimic of either a hydroxyl group (in terms of electronegativity) [59,60,61,62] or a hydrogen atom (in terms of steric factor). In this context, we will also provide an overview on properties and applications of oligonucleotides carrying fluorine modifications at different positions.

3.1. Fluorocarbon OligoNucleotide Amphiphiles (FONs)

Recently our group has reported on the synthesis and biological properties of FONs [63]. In previous studies, our group already reported on the synthesis and cellular uptake studies of lipid-oligonucleotide conjugates (LONs) [64,65]. The main purpose of attaching a fluorocarbon chain to a polyanionic oligonucleotide (ONs) was to study how cellular uptake or internalization of ONs are influenced by the nature of the hydrophobic moieties. To address this issue, Godeau et al. have synthesized two FONs featuring C8F17 and C6F13 chains using 1,3-dipolar Huisgen’s reaction [66] via a “post synthetic” modification approach. Three different human cell lines were incubated with either FONs or LONs or unconjugated ONs for comparative study of cellular internalization (Figure 5). Moreover, internalization kinetics of FONs, LONs and unconjugated ONs with Huh7 cells were also evaluated. Keeping in mind the hydrophobic and lipophobic properties of fluorocarbon chains, it can be hypothesized that FONs self-assemble outside the cell to form nano aggregates. It is likely that these nanoobjects would be uptaken by the cell via an endocytosis pathway. It is worth mentioning here that to the best of our knowledge, there is no earlier report on oligonucleotide amphiphiles featuring perfluorinated chains.
Figure 5. Conjugation of fluorocarbon chain to the oligonucleotide (FONs) allows the delivery of highly polyanionic oligonucleotides into human cells.
Figure 5. Conjugation of fluorocarbon chain to the oligonucleotide (FONs) allows the delivery of highly polyanionic oligonucleotides into human cells.
Applsci 02 00245 g005

3.2. Application of Fluorinated Oligonucleotide Analogues

Various groups have been interested in the synthesis and application of fluorinated oligonucleotide analogues. There are different possible sites for the modification of an oligonucleotide with one or more fluorine atoms for various biological and therapeutical applications. Fluorine atoms have a strong influence on both the electronic and the conformational properties of the nucleic acids’ furanose ring. The 2’-position in the ribose moiety of RNA is prime target for chemical modifications. Extensive research work is going on towards the study of structural properties, biological applications and delivery strategies of oligonucleotides-incorporated with 2’-deoxy-2’fluoronucleotide in combination with other structural modifications [67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84]. Oligonucleotides containing 2’-deoxy-2’fluoronucleosides have been synthesized for regiospecific cleavage of RNA by RNase H [67]. Incorporation of 2’-deoxy-2’-fluoronucleotide into oligonucleotides induces substantial enhancement in their binding affinities to RNA targets [68]. Damha et al. have shown that fluorine-induced sugar pucker in 2’-deoxy-2’fluoro-β-D-ribonucleicacid (2’-F RNA; C3’-endo) and 2’-deoxy-2’fluoro-β-D-arabinonucleicacid (2’-F ANA; C2’/O4’-endo) can improve the activity of siRNA (Figure 6) and enhance its serum stability. They have also provided the evidences for an important role of a pseudohydrogen bond between the F2’ and H8 of the nucleobase for 2’-F ANA:RNA duplex stability [72,73,74,75,76,77,78,79,80,81,82,83,84].
Figure 6. Structure and sugar puckering equilibrium for (a) 2’-deoxy-2’-fluororibonucleic acid (2’-F RNA); C3’-endo is the most stable conformation for 2’-F RNA. (b) 2’-deoxy-2’-fluoroarabonucleic acid (2’-F ANA); C2’-endo is the most stable conformation for 2’-F ANA.
Figure 6. Structure and sugar puckering equilibrium for (a) 2’-deoxy-2’-fluororibonucleic acid (2’-F RNA); C3’-endo is the most stable conformation for 2’-F RNA. (b) 2’-deoxy-2’-fluoroarabonucleic acid (2’-F ANA); C2’-endo is the most stable conformation for 2’-F ANA.
Applsci 02 00245 g006
On the other side, very interesting and novel studies have been carried out by Erande et al. using 3’-deoxy-3’ribofluoro/xylofluoro uridine incorporated oligonucleotides (2’-5’-strand) [85]. They have shown the effect of incorporation of one or more unit of 3’-deoxy-3’-fluoronucleosides (having either locked or frozen S-type/Ntype sugar conformations) on the stability of isoDNA:RNA duplexes. For the first time, evidences were given that the DNA strand prefers S-type geometry in stable isoDNA:RNA duplexes (Figure 7) [85].
Figure 7. Structure and sugar puckering equilibrium for (a) 3’-deoxy-3’-ribofluorouridine. (b) 3’-deoxy-3’-xylofluorouridine.S-type (C2’-endo) conformation (preferred by 3’-deoxy-3’-ribofluorouridine) stabilized isoDNA:RNA duplexed while N-type (C3’-endo) conformation (preferred by 3’-deoxy-3’-xylofluorouridine) destabilized isoDNA:RNA duplexes.
Figure 7. Structure and sugar puckering equilibrium for (a) 3’-deoxy-3’-ribofluorouridine. (b) 3’-deoxy-3’-xylofluorouridine.S-type (C2’-endo) conformation (preferred by 3’-deoxy-3’-ribofluorouridine) stabilized isoDNA:RNA duplexed while N-type (C3’-endo) conformation (preferred by 3’-deoxy-3’-xylofluorouridine) destabilized isoDNA:RNA duplexes.
Applsci 02 00245 g007
To investigate the forces (hydrogen bonding, base stacking and solvation) which are responsible for stability of the secondary structure of nucleic acids, Engels et al. have synthesized novel nucleic acid analogues in which nucleobases are replaced by their steric mimics, containing one or more fluorine atom. They have utilized fluorobenzene and fluorobenzimidazole derivatives (Figure 8) [86,87]. They have discussed how C–F…H–C hydrogen bonds affect the stability of the secondary structure of nucleic acids in details.
Figure 8. Monomer building blocks for the incorporation of fluorine derivative into RNA oligonucleotides by solid-phase synthesis.
Figure 8. Monomer building blocks for the incorporation of fluorine derivative into RNA oligonucleotides by solid-phase synthesis.
Applsci 02 00245 g008
Owing to the favorable NMR properties of fluorine atom (19F), fluorinated oligonucleotide derivatives provide a sensitive probe for structural study of nucleic acids in the solution state and their interactions with other biological moieties [71,88,89,90,91,92,93,94,95]. Barhate et al. have synthesized nucleosides with perfluorinated tert-butyl group and successfully incorporated it into the oligonucleotide at different positions and demonstrated as a sensitive 19F NMR probe of nucleic acid conformation [95]. Oligonucleotides, labeled with radioactive isotopes, are new imaging tools to study gene expression at the nucleic acid and protein levels. Due to its positron-emission properties, fluorine-18 (radioactive isotope of fluorine) is widely used for oligonucleotide labeling for in vivo PET (Positron Emission Tomography) imaging. Dollé et al. [96,97,98,99,100]. have developed an original method for labeling oligonucleotides with 18F in different positions. Several other research groups have also synthesized 18F labeled oligonucleotide analogues for imaging studies with PET [101,102,103].

4. Conclusions

The nucleic acids fluorocarbon amphiphiles belong to a new class of hybrid bioinspired molecules, which is clearly connected to soft materials, nanosciences and biomedical technology. It is a promising and exciting time for NAFAs and the emerging devices developed so far clearly open up interesting routes for the design and development of nanodevices, therapeutic strategies and biotechnological tools. The potential innovations using these fluorocarbon molecules are numerous in terms of chemical architectures, physicochemical and biological properties. This short survey highlights the interest to combine nucleic acids with fluorocarbons for developing soft and complex matters. The history of this field is still at the beginning and we can imagine that remarkable achievements will be realized in the future.

Acknowledgments

P.B. acknowledges financial support from the Army Research Office. This work has been supported by the French National Agency (ANR) in the frame of its programme PNANO (project NANAN, n° ANR-08-NANO-028).

Conflict of Interest

The authors declare no conflict of interest.

References

  1. Watson, J.D. A structure for deoxyribose nucleic acid. Nature 1953, 171, 737–738. [Google Scholar] [CrossRef]
  2. Franklin, R.E. Molecular configuration in sodium thymonucleate. Nature 1953, 171, 740–741. [Google Scholar]
  3. Wilkins, M.H.F. Molecular structure of deoxypentose nucleic acids. Nature 1953, 171, 738–740. [Google Scholar] [CrossRef]
  4. Sessler, J.L.; Lawrence, C.M.; Jayawickramarajah, J.G.M. Molecular recognition via base-pairing. Chem. Soc. Rev. 2007, 36, 314–325. [Google Scholar]
  5. Whitesides, G.M.; Mathias, J.P.; Seto, C.T. Molecular self-assembly and nanochemistry: A chemical strategy for the synthesis of nanostructures. Science 1991, 254, 1312–1319. [Google Scholar]
  6. Bowden, N.; Terfort, A.; Carbeck, J.; Whitesides, G.M. Self-assembly of mesoscale objects into ordered two-dimensional arrays. Science 1997, 276, 233–235. [Google Scholar]
  7. Rosemeyer, H. Nucleolipids: Natural occurrence, synthesis, molecular recognition, and supramolecular assemblies as potential precursors of life and bioorganic materials. Chem. Biodivers. 2005, 2, 977–1062. [Google Scholar] [CrossRef]
  8. Gissot, A.; Camplo, M.; Grinstaff, M.W.; Barthélémy, P. Nucleoside, nucleotide and oligonucleotide based amphiphiles: A successful marriage of nucleic acids with lipids. Org. Biomol. Chem. 2008, 6, 1324–1333. [Google Scholar] [CrossRef]
  9. Ahlers, M.; Ringsdorf, H.; Rosemeyer, H.; Seela, F. Orientation, recognition, and photoreaction of nucleolipids in model membranes. Colloid Polym. Sci. 1990, 268, 132–142. [Google Scholar] [CrossRef]
  10. Huang, J.; Li, C.; Liang, Y. FT-SERS studies on molecular recognition capabilities of monolayers of novel nucleolipid amphiphiles. Langmuir 2000, 16, 3937–3940. [Google Scholar] [CrossRef]
  11. Lebeau, L.; Olland, S.; Oudet, P.; Mioskowski, C. Rational design and synthesis of phospholipids for the two-dimensional crystallization of DNA gyrase, a key element in chromosome organization. Chem. Phys. Lipid 1992, 62, 93–103. [Google Scholar] [CrossRef]
  12. Barthélémy, P.; Lee, S.J.; Grinstaff, M. Supramolecular assemblies with DNA. Pure Appl. Chem. 2005, 77, 2133–2148. [Google Scholar]
  13. Berti, D.; Franchi, L.; Baglioni, P.; Luisi, P.L. Molecular recognition in monolayers. Complementary base pairing in dioleoylphosphatidyl derivatives of adenosine, uridine, and cytidine. Langmuir 1997, 13, 3438–3444. [Google Scholar] [CrossRef]
  14. Baglioni, P.; Berti, D. Self assembly in micelles combining stacking and H-bonding. Curr. Opin. Colloid Interface Sci. 2003, 8, 55–61. [Google Scholar] [CrossRef]
  15. Fortini, M.; Berti, D.; Baglioni, P.; Ninham, B.W. Specific anion effects on the aggregation properties of anionic nucleolipids. Curr. Opin. Colloid Interface Sci. 2004, 9, 168–172. [Google Scholar] [CrossRef]
  16. Mulet, X.; Kaasgaard, T.; Conn, C.E.; Waddington, L.J.; Kennedy, D.F.; Weerawardena, A.; Drummond, C.J. Nanostructured nonionic thymidine nucleolipid self-assembly materials. Langmuir 2010, 26, 18415–18423. [Google Scholar]
  17. Murgia, S.; Lampis, S.; Angius, R.; Berti, D.; Monduzzi, M. Orientation and specific interactions of nucleotides and nucleolipids inside monoolein-based liquid crystals. J. Phys. Chem. B 2009, 113, 9205–9215. [Google Scholar] [CrossRef]
  18. Godeau, G.; Barthélémy, P. Glycosyl-nucleoside lipids as low-molecular-weight gelators. Langmuir 2009, 25, 8447–8450. [Google Scholar] [CrossRef]
  19. Khiati, S.; Luvino, D.; Oumzil, K.; Chauffert, B.; Camplo, M.; Barthélémy, P. Nucleoside-lipid-based nanoparticles for cisplatin delivery. ACS Nano 2011, 5, 8649–8655. [Google Scholar] [CrossRef]
  20. Hong, C.I.; Nechaev, A.; Kirisits, A.J.; Vig, R.; West, C.R.; Manouilov, K.K.; Chu, C.K. Synthesis and biological activity of anti-HIV nucleoside conjugates of ether and thioether phospholipids. J. Med. Chem. 1996, 39, 1771–1777. [Google Scholar] [CrossRef]
  21. Ambike, A.; Rosilio, V.; Stella, B.; Lepétre-Mouelhi, S.; Couvreur, P. Interaction of self-assembled squalenoyl gemcitabine nanoparticles with phospholipid−cholesterol monolayers mimicking a biomembrane. Langmuir 2011, 27, 4891–4899. [Google Scholar]
  22. Riess, J.G. Highly fluorinated amphiphilic molecules and self-assemblies with biomedical potential. Curr. Opin. Colloid Interface Sci. 2009, 14, 294–304. [Google Scholar] [CrossRef]
  23. Krafft, M.P. Controlling phospholipid self-assembly and film properties using highly fluorinated components—Fluorinated monolayers, vesicles, emulsions and microbubbles. Biochimie 2012, 94, 11–25. [Google Scholar]
  24. Krafft, M.P.; Riess, J.G. Highly fluorinated amphiphiles and colloidal systems, and their applications in the biomedical field. A contribution. Biochimie 1998, 80, 489–514. [Google Scholar] [CrossRef]
  25. Riess, J.G.; Krafft, M.P. Fluorinated phosphocholine-based amphiphiles as components of fluorocarbon emulsions and fluorinated vesicles. Chem. Phys. Lipids 1995, 75, 1–14. [Google Scholar] [CrossRef]
  26. Held, P.; Lach, F.; Lebeau, L.; Mioskowski, C. Synthesis and preliminary evaluation of a new class of fluorinated amphiphiles designed for in-plane immobilisation of biological macromolecules. Tetrahedron Lett. 1997, 38, 1937–1940. [Google Scholar] [CrossRef]
  27. Lebeau, L.; Lach, F.; Vénien-Bryan, C.; Renault, A.; Dietrich, J.; Jahn, T.; Palmgren, M.G.; Kühlbrandt, W.; Mioskowski, C. Two-dimensional crystallization of a membrane protein on a detergent-resistant lipid monolayer. J. Mol. Biol. 2001, 308, 639–647. [Google Scholar] [CrossRef]
  28. Courty, S.; Lebeau, L.; Martel, L.; Lenné, P.-F.; Balavoine, F.; Dischert, W.; Konovalov, O.; Mioskowski, C.; Legrand, J.-F.; Vénien-Bryan, C. Two-dimensional crystallization of a histidine-tagged protein on monolayers of fluidity-enhanced Ni2+-chelating lipids. Langmuir 2002, 18, 9502–9512. [Google Scholar]
  29. Clary, L.; Gadras, C.; Greiner, J.; Rolland, J.-P.; Santaella, C.; Vierling, P.; Gulik, A. Phase behavior of fluorocarbon and hydrocarbon double-chain hydroxylated and galactosylated amphiphiles and bolaamphiphiles. Long-term shelf-stability of their liposomes. Chem. Phys. Lipids 1999, 99, 125–137. [Google Scholar] [CrossRef]
  30. Pasc-Banu, A.; Blanzat, M.; Belloni, M.; Perez, E.; Mingotaud, C.; Rico-Lattes, I.; Labrot, T.; Oda, R. Spontaneous vesicles of single-chain sugar-based fluorocarbon surfactants. J. Fluor. Chem. 2005, 126, 33–38. [Google Scholar] [CrossRef]
  31. Roytman, R.; Adler-Abramovich, L.; Ajish, K.S.; Kuan, T.-C.; Lin, C.-C.; Gazit, E.; Brik, A. Exploring the self-assembly of glycopeptides using a diphenylalanine scaffold. Org. Biomol. Chem. 2011, 9, 5755–5761. [Google Scholar]
  32. Platen, T.; Schüler, T.; Tremel, W.; Hoffmann-Röder, A. Synthesis and antibody bonding of higly fluorinated amphiphilic MUC1 glycopeptide antigens. Eur. J. Org. Chem. 2011, 3878–3887. [Google Scholar]
  33. Riess, J.G. Fluorinated vesicles. J. Drug Target. 1994, 2, 455–468. [Google Scholar] [CrossRef]
  34. Fabio, K.; Di Giorgio, C.; Vierling, P. New perfluorinated polycationic dimerizable detergents for the formulation of monomolecular DNA nanoparticles and their in vitro transfection efficiency. Biochim. Biophys. Acta 1724, 203–214. [Google Scholar]
  35. Klein, E.; Leborgne, C.; Ciobanu, M.; Klein, J.; Frisch, B.; Pons, F.; Zuber, G.; Scherman, D.; Kichler, A.; Lebeau, L. Nucleic acid transfer with hemifluorinated polycationic lipids. Biomaterials 2010, 31, 4781–4788. [Google Scholar]
  36. Klein, E.; Ciobanu, M.; Klein, J.; MacHi, V.; Leborgne, C.; Vandamme, T.; Frisch, B.; Pons, F.; Kichler, A.; Zuber, G.; Lebeau, L. "HFP" fluorinated cationic lipids for enhanced lipoplex stability and gene delivery. Bioconjug. Chem. 2010, 21, 360–371. [Google Scholar] [CrossRef]
  37. Berti, D.; Montis, C.; Baglioni, P. Self-assembly of designer biosurfactants. Soft Matter 2011, 7, 7150–7158. [Google Scholar]
  38. Wang, Y.; Moreau, L.; Barthélémy, P.; El Maataoui, M.; Grinstaff, M.W. Supramolecular assemblies of nucleoside phosphocholine amphiphiles. J. Am. Chem. Soc. 2004, 126, 7533–7539. [Google Scholar]
  39. Desbat, B.; Manet, S.; Aimé, C.; Labrot, T.; Oda, R. Aggregation behaviors of gemini nucleotide at the air-water interface and in solutions induced by adenine-uracil interaction. J. Colloid Interface Sci. 2005, 283, 555–564. [Google Scholar] [CrossRef]
  40. Pincet, F.; Perez, E.; Bryant, G.; Lebeau, L.; Mioskowski, C. Long-range attraction between nucleosides with short-range specificity: Direct measurements. Phys. Rev. Lett. 1994, 73, 2780–2783. [Google Scholar] [CrossRef]
  41. Pincet, F.; Perez, E.; Lebeau, L.; Mioskowski, C. Long range H-bond specific interactions between nucleosides. J. Chem. Soc. Faraday Trans. 1995, 91, 4329–4330. [Google Scholar] [CrossRef]
  42. Pincet, F.; Perez, E.; Bryant, G.; Lebeau, L.; Mioskowski, C. Specific forces between DNA bases. Mod. Phys. Lett. B 1996, 10, 81–90. [Google Scholar] [CrossRef]
  43. Pincet, F.; Rawicz, W.; Perez, E.; Lebeau, L.; Mioskowski, C.; Evans, E. Electrostatic nanotitration of weak biochemical bonds. Phys. Rev. Lett. 1997, 79, 1949–1952. [Google Scholar] [CrossRef]
  44. Pincet, F.; Lebeau, L.; Cribier, S. Short-range specific forces are able to induce hemifusion. Eur. Biophys. J. 2001, 30, 91–97. [Google Scholar] [CrossRef]
  45. Perez, E.; Pincet, F.; Goldmann, M.; Mioskowski, C.; Lebeau, L. Translational order in liquid-expanded lipid monolayers functionalized with nucleosides. Eur. Phys. J. B 1998, 6, 1–4. [Google Scholar] [CrossRef]
  46. Tareste, D.; Pincet, F.; Perez, E.; Rickling, S.; Mioskowski, C.; Lebeau, L. Energy of hydrogen bonds probed by the adhesion of functionalized lipid layers. Biophys. J. 2002, 83, 3675–3681. [Google Scholar] [CrossRef]
  47. Tareste, D.; Pincet, F.; Lebeau, L.; Perez, É. Hydrophobic forces and hydrogen bonds in the adhesion between retinoid-coated surfaces. Langmuir 2007, 23, 3225–3229. [Google Scholar] [CrossRef]
  48. Yang, H.W.; Yi, J.W.; Bang, E.-K.; Jeon, E.M.; Kim, B.H. Cationic nucleolipids as efficient siRNA carriers. Org. Biomol. Chem. 2011, 9, 291–296. [Google Scholar]
  49. Ceballos, C.; Khiati, S.; Prata, C.A.; Zhang, X.X.; Giorgio, S.; Marsal, P.; Grinstaff, M.W.; Barthélémy, P.; Camplo, M. Cationic nucleoside lipids derived from universal bases: A rational approach for siRNA transfection. Bioconjug. Chem. 2010, 21, 1062–1069. [Google Scholar] [CrossRef]
  50. Moreau, L.; Barthélémy, P.; Li, Y.; Luo, D.; Prata, C.A.H.; Grinstaff, M.W. Nucleoside Phosphocholine Amphiphile for in vitro DNA transfection. Mol. BioSyst. 2005, 1, 260–264. [Google Scholar] [CrossRef]
  51. Chabaud, P.; Camplo, M.; Payet, D.; Serin, G.; Moreau, L.; Barthélémy, P.; Grinstaff, M.W. Cationic nucleoside lipids for gene delivery. Bioconjug. Chem. 2006, 17, 466–472. [Google Scholar] [CrossRef]
  52. Céballos, C.; Khiati, S.; Barthélémy, P.; Camplo, M. Acyclic anionic nucleolipids for DNA delivery. J. Nanosci. Lett. 2012, 2, 20. [Google Scholar]
  53. Khiati, S.; Pierre, N.; Andriamanarivo, S.; Grinstaff, M.W.; Arazam, N.; Nallet, F.; Navailles, L.; Barthélémy, P. Anionic nucleotide-lipids for in vitro DNA transfection. Bioconjug. Chem. 2009, 20, 1765–1772. [Google Scholar] [CrossRef]
  54. Moreau, L.; Barthélémy, P.; El Maataoui, M.; Grinstaff, M.W. Supramolecular assemblies of nucleoside phosphocholine amphiphiles. J. Am. Chem. Soc. 2004, 126, 7533–7539. [Google Scholar]
  55. Moreau, L.; Campins, N.; Grinstaff, M.W.; Barthélémy, P. A fluorocarbon nucleoamphiphile for the construction of actinide loaded microspheres. Tetrahedron Lett. 2006, 47, 7117–7120. [Google Scholar]
  56. Godeau, G.; Barthélémy, P. Glycosyl-nucleoside lipids as low-molecular-weight gelators. Langmuir 2009, 25, 8447–8450. [Google Scholar] [CrossRef]
  57. Godeau, G.; Bernard, L.; Staedel, C.; Barthélémy, P. Glycosyl-nucleoside-lipid based supramolecular assembly as a nanostructured material with nucleic acid delivery capabilities. Chem. Commun. 2009, 34, 5127–5129. [Google Scholar]
  58. Godeau, G.; Brun, C.; Arnion, H.; Staedel, C.; Barthélémy, P. Glycosyl-nucleoside fluorinated amphiphiles as compoents of nanostructured hydrogels. Tetrahedron Lett. 2010, 51, 1012–1015. [Google Scholar]
  59. Studer, A.; Hadida, S.; Ferritto, R.; Kim, S.-Y.; Jeger, P.; Wipf, P.; Curran, D.P. Fluorous synthesis: A fluorous-phase strategy for improving separation efficiency in organic synthesis. Science 1997, 275, 823–826. [Google Scholar] [CrossRef]
  60. Pongdee, R.; Liu, H.-W. Elucidation of enzyme mechanisms using fluorinated substrate analogues. Bioorg. Chem. 2004, 32, 393–437. [Google Scholar] [CrossRef]
  61. Müller, K.; Faeh, C.; Diederich, F. Fluorine in pharmaceuticals: Looking beyond intuition. Science 2007, 317, 1881–1886. [Google Scholar]
  62. Curran, D.P. Chemistry: Fluorous tags unstick messy chemical biology problems. Science 2008, 321, 1645–1646. [Google Scholar] [CrossRef]
  63. Godeau, G.; Arnion, H.; Brun, C.; Staedel, C.; Barthélémy, P. Fluorocarbon oligonucleotide conjugates for nucleic acids delivery. Med. Chem. Commun. 2010, 1, 76–78. [Google Scholar] [CrossRef]
  64. Patwa, A.; Gissot, A.; Bestel, I.; Barthélémy, P. Hybrid lipid oligonucleotide conjugates: Synthesis, self-assemblies and biomedical applications. Chem. Soc. Rev. 2011, 40, 5844–5854. [Google Scholar] [CrossRef]
  65. Godeau, G.; Staedel, C.; Barthélémy, P. Lipid-conjugated oligonucleotides via “click chemistry” efficiently inhibit hepatitis C virus translation. J. Med. Chem. 2008, 51, 4374–4376. [Google Scholar] [CrossRef]
  66. Huisgen, R. 1,3-Dipolar Cycloaddition Chemistry; Wiley: New York, NY ,USA, 1984; Volume 1, pp. 1–176. [Google Scholar]
  67. Schmidt, S.; Niemann, A.; Krynetskaya, N.F.; Oretskaya, T.S.; Metelev, V.G.; Suchomlinov, V.V.; Shabarova, Z.A.; Cech, D. The use of oligonucleotide probes containing 2'-deoxy-2'-fluoronucleosides for regiospecific cleavage of RNA by RNase H from Escherichia coli. Biochim. Biophys. Acta 1130, 41–46. [Google Scholar]
  68. Kawasaki, A.M.; Casper, M.D.; Freier, S.M.; Lesnik, E.A.; Zounes, M.C.; Cummins, L.L.; Gonzalez, C.; Dan Cook, P. Uniformly modified 2′-deoxy-2′-fluoro phosphorothioate oligonucleotides as nuclease-resistant antisense compounds with high affinity and specificity for RNA targets. J. Med. Chem. 1993, 36, 831–841. [Google Scholar] [CrossRef]
  69. Kois, P.; Tocik, Z.; Spassova, M.; Ren, W.-Y.; Rosenberg, I.; Soler, J.F.; Watanabe, K.A. Synthesis and some properties of modified oligonucleotides. 2. oligonucleotides containing 2'-deoxy-2'-fluoro-β-D-arabinofuranosyl pyrimidine nucleosides. Nucleosides Nucleotides 1993, 12, 1093–1109. [Google Scholar] [CrossRef]
  70. Rosenberg, I.; Soler, J.F.; Tocik, Z.; Ren, W.-Y.; Ciszewski, L.A.; Kois, P.; Pankiewicz, K.W.; Spassova, M.; Watanabe, K.A. Synthesis of oligodeoxynucleotides containing the C-nucleoside and 2'- deoxy-2'-fluoro-ara-nucleoside moieties by the H-phosphonate method. Nucleosides Nucleotides 1993, 12, 381–401. [Google Scholar] [CrossRef]
  71. Reif, B.; Wittmann, V.; Schwalbe, H.; Griesinger, C.; Wörner, K.; Jahn-Hofmann, K.; Engels, J.W.; Bermel, W. 132. Structural comparison of oligoribonucleotides and their 2′-deoxy-2′-fluoro analogs by heteronuclear NMR spectroscopy. Helv. Chim. Acta 1997, 80, 1952–1971. [Google Scholar] [CrossRef]
  72. Damha, M.J.; Wilds, C.J.; Noronha, A.; Brukner, I.; Borkow, G.; Arion, D.; Parniak, M.A. Hybrids of RNA and arabinonucleic acids (ANA and 2'F-ANA) are substrates of ribonuclease H. J. Am. Chem. Soc. 1998, 120, 12976–12977. [Google Scholar]
  73. Wilds, C.J.; Damha, M.J. Duplex recognition by oligonucleotides containing 2'-deoxy-2'-fluoro-D-arabinose and 2'-deoxy-2'-fluoro-D-ribose. Intermolecular 2'-OH-phosphate contacts versus sugar puckering in the stabilization of triple-helical complexes. Bioconjugate Chem. 1999, 10, 299–305. [Google Scholar] [CrossRef]
  74. Wilds, C.J.; Damha, M.J. 2'-Deoxy-2'-fluoro-β-D-arabinonucleosides and oligonucleotides (2'F-ANA): Synthesis and physicochemical studies. Nucleic Acids Res. 2000, 28, 3625–3635. [Google Scholar] [CrossRef]
  75. Damha, M.J.; Noronha, A.M.; Wilds, C.J.; Trempe, J.-F.; Denisov, A.; Pon, R.T.; Gehring, K. Properties of arabinonucleic acids (ANA & 20′F-ANA): Implications for the design of antisense therapeutics that invoke RNase H cleavage of RNA. Nucleosides Nucleotides Nucleic Acids 2001, 20, 429–440. [Google Scholar] [CrossRef]
  76. Denisov, A.Yu.; Noronha, A.M.; Wilds, C.J.; Trempe, J.-F.; Pon, R.T.; Gehring, K.; Damha, M.J. Solution structure of an arabinonucleic acid (ANA)/RNA duplex in a chimeric hairpin: Comparison with 2′-fluoro-ANA/RNA and DNA/RNA hybrids. Nucleic Acids Res. 2001, 29, 4284–4293. [Google Scholar] [CrossRef]
  77. Lok, C.-N.; Viazovkina, E.; Min, K.-L.; Nagy, E.; Wilds, C.J.; Damha, M.J.; Parniak, M.A. Potent gene-specific inhibitory properties of mixed-backbone antisense oligonucleotides comprised of 2′-deoxy-2′-fluoro-D-arabinose and 2′-deoxyribose nucleotides. Biochemistry 2002, 41, 3457–3467. [Google Scholar]
  78. Doi, Y.; Katafuchi, A.; Fujiwara, Y.; Hitomi, K.; Tainer, J.A.; Ide, H.; Iwai, S. Synthesis and characterization of oligonucleotides containing 2′-fluorinated thymidine glycol as inhibitors of the endonuclease III reaction. Nucleic Acids Res. 2006, 34, 1540–1551. [Google Scholar] [CrossRef]
  79. Dowler, T.; Bergeron, D.; Tedeschi, A.-L.; Paquet, L.; Ferrari, N.; Damha, M.J. Improvements in siRNA properties mediated by 2′-deoxy-2′-fluoro-β-D-arabinonucleic acid (FANA). Nucleic Acids Res. 2006, 34, 1669–1675. [Google Scholar] [CrossRef]
  80. Watts, J.K.; Choubdar, N.; Sadalapure, K.; Robert, F.; Wahba, A.S.; Pelletier, J.; Pinto, B.M.; Damha, M.J. 2′-Fluoro-4′-thioarabino-modified oligonucleotides: Conformational switches linked to siRNA activity. Nucleic Acids Res. 2007, 35, 1441–1451. [Google Scholar] [CrossRef]
  81. Peng, C.G.; Damha, M.J. Polymerase-directed synthesis of 2′-deoxy-2′-fluoro-β-D- arabinonucleic acids. J. Am. Chem. Soc. 2007, 129, 5310–5311. [Google Scholar] [CrossRef]
  82. Peng, C.G.; Damha, M.J. G-quadruplex induced stabilization by 2′-deoxy-2′-fluoro-d-arabinonucleic acids (2′F-ANA). Nucleic Acids Res. 2007, 35, 4977–4988. [Google Scholar] [CrossRef]
  83. Watts, J.K.; Martín-Pintado, N.; Gómez-Pinto, I.; Schwartzentruber, J.; Portella, G.; Orozco, M.; González, C.; Damha, M.J. Differential stability of 2′F-ANA•RNA and ANA•RNA hybrid duplexes: Roles of structure, pseudohydrogen bonding, hydration, ion uptake and flexibility. Nucleic Acids Res. 2010, 38, 2498–2511. [Google Scholar]
  84. Deleavey, G.F.; Watts, J.K.; Alain, T.; Robert, F.; Kalota, A.; Aishwarya, V.; Pelletier, J.; Gewirtz, A.M.; Sonenberg, N.; Damha, M.J. Synergistic effects between analogs of DNA and RNA improve the potency of siRNA-mediated gene silencing. Nucleic Acids Res. 2010, 38, 4547–4557. [Google Scholar]
  85. Erande, N.; Gunjal, A.D.; Fernandes, M.; Kumar, V.A. Probing the furanose conformation in the 2’-5’ strand of isoDNA:RNA duplexes by freezing the nucleoside conformations. Chem. Commun. 2011, 47, 4007–4009. [Google Scholar]
  86. Parsch, J.; Engels, J.W. Synthesis of fluorobenzene and benzimidazole nucleic-acid analogues and their influence on stability of RNA duplexes. Helv. Chim. Acta 2000, 83, 1791–1808. [Google Scholar] [CrossRef]
  87. Parsch, J.; Engels, J.W. C–F … H–C hydrogen bonds in ribonucleic acids. J. Am. Chem. Soc. 2002, 124, 5664–5672. [Google Scholar] [CrossRef]
  88. Rastinejad, F.; Evilia, C.; Lu, P. Studies of nucleic acids and their protein interactions by 19F NMR. Methods Enzymol. 1995, 261, 560–575. [Google Scholar] [CrossRef]
  89. Rastinejad, F.; Lu, P. Bacteriophage T7 RNA polymerase. 19F-nuclear magnetic resonance observations at 5-fluorouracil-substituted promoter DNA and RNA transcript. J. Mol. Biol. 1993, 232, 105–122. [Google Scholar] [CrossRef]
  90. Chu, W.-C.; Feiz, V.; Derrick, W.B.; Horowitz, J. Fluorine-19 nuclear magnetic resonance as a probe of the solution structure of mutants of 5-Fluorouracil-substituted Escherichia coli valine tRNA. J. Mol. Biol. 1992, 227, 1164–1172. [Google Scholar] [CrossRef]
  91. Chu, W.-C.; Horowitz, J. Fluorine-19 NMR studies of the thermal unfolding of 5-fluorouracil-substituted Escherichia coli valine transfer RNA. FEBS Lett. 1991, 295, 159–162. [Google Scholar] [CrossRef]
  92. Parisot, D.; Malet-Martino, M.C.; Martino, R.; Crasnier, P. 19F nuclear magnetic resonance analysis of 5-fluorouracil metabolism in four differently pigmented strains of Nectria haematococca. Appl. Environ. Microbil. 1991, 57, 3605–3612. [Google Scholar]
  93. Tanabe, K.; Sugiura, M.; Nishimoto, S.-I. Monitoring of duplex and triplex formation by 19F NMR using oligodeoxynucleotides possessing 5-fluorodeoxyuridine unit as 19F signal transmitter. Bioorg. Med. Chem. 2010, 18, 6690–6694. [Google Scholar] [CrossRef] [Green Version]
  94. Danielson, M.A.; Falke, J.J. Use of 19F NMR to probe protein structure and conformational changes. Annu. Rev. Biophys. Biomol. Struct. 1996, 25, 163–195. [Google Scholar] [CrossRef]
  95. Barhate, N.B.; Barhate, R.N.; Cekan, P.; Drobny, G.; Sigurdsson, S.Th. A nonafluoro nucleoside as a sensitive 19F NMR probe of nucleic acid conformation. Org. Lett. 2008, 10, 2745–2747. [Google Scholar] [CrossRef]
  96. Dollé, F.; Hinnen, F.; Vaufrey, F.; Tavitian, B.; Crouzel, C. A general method for labeling oligodeoxynucleotides with 18F for in vivo PET imaging. J. Label. Compd. Radiopharm. 1997, 39, 319–330. [Google Scholar] [CrossRef]
  97. Tavitian, B.; Terrazzino, S.; Kühnast, B.; Marzabal, S.; Stettler, O.; Dollé, F.; Deverre, J.-R.; Jobert, A.; Hinnen, F.; Bendriem, B.; et al. In vivo imaging of oligonucleotides with positron emission tomography. Nat. Med. 1998, 4, 467–470. [Google Scholar] [CrossRef]
  98. Kuhnast, B.; Dollé, F.; Vaufrey, F.; Hinnen, F.; Crouzel, C.; Tavitian, B. Fluorine-18 labeling of oligonucleotides bearing chemically-modified ribose-phosphate backbones. J. Label. Compd. Radiopharm. 2000, 43, 837–848. [Google Scholar] [CrossRef]
  99. Kuhnast, B.; Hinnen, F.; Boisgard, R.; Tavitian, B.; Dollé, F. Fluorine-18 labelling of oligonucleotides: Prosthetic labelling at the 5′-end using the N-(4-[18F]fluorobenzyl)-2-bromoacetamide reagent. J. Label. Compd. Radiopharm. 2003, 46, 1093–1103. [Google Scholar] [CrossRef]
  100. Viel, T.; Kuhnast, B.; Hinnen, F.; Boisgard, R.; Tavitian, B.; Dollé, F. Fluorine-18 labelling of small interfering RNAs (siRNAs) for PET imaging. J. Label. Compd. Radiopharm. 2007, 50, 1159–1168. [Google Scholar] [CrossRef]
  101. Hedberg, E.; Långström, B. 18F-labelling of oligonucleotides using succinimido 4-[18F]fluorobenzoate. Acta Chem. Scand. 1998, 52, 1034–1039. [Google Scholar] [CrossRef]
  102. Pan, D.; Gambhir, S.S.; Toyokuni, T.; Iyer, M.R.; Acharya, N.; Phelps, M.E.; Barrio, J.R. Rapid synthesis of a 5'-fluorinated oligodeoxy-nucleotide: A model antisense probe for use in imaging with positron emission tomography (PET). Bioorg. Med. Chem. Lett. 1998, 8, 1317–1320. [Google Scholar] [CrossRef]
  103. Mercier, F.; Paris, J.; Kaisin, G.; Thonon, D.; Flagothier, J.; Teller, N.; Lemaire, C.; Luxen, A. General method for labeling siRNA by click chemistry with fluorine-18 for the purpose of PET imaging. Bioconjug. Chem. 2011, 22, 108–114. [Google Scholar] [CrossRef]

Share and Cite

MDPI and ACS Style

Dolain, C.; Patwa, A.; Godeau, G.; Barthélémy, P. Nucleic Acid Based Fluorinated Derivatives: New Tools for Biomedical Applications. Appl. Sci. 2012, 2, 245-259. https://doi.org/10.3390/app2020245

AMA Style

Dolain C, Patwa A, Godeau G, Barthélémy P. Nucleic Acid Based Fluorinated Derivatives: New Tools for Biomedical Applications. Applied Sciences. 2012; 2(2):245-259. https://doi.org/10.3390/app2020245

Chicago/Turabian Style

Dolain, Christel, Amit Patwa, Guilhem Godeau, and Philippe Barthélémy. 2012. "Nucleic Acid Based Fluorinated Derivatives: New Tools for Biomedical Applications" Applied Sciences 2, no. 2: 245-259. https://doi.org/10.3390/app2020245

Article Metrics

Back to TopTop