Super-resolution Imaging of Structure, Molecular Composition, and Stability of Single Oligonucleotide PolyplexesClick to copy article linkArticle link copied!
- Natalia Feiner-GraciaNatalia Feiner-GraciaNanoscopy for Nanomedicine Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Carrer Baldiri Reixac 15-21, 08024 Barcelona, SpainDepartment of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5612AZ Eindhoven, The NetherlandsMore by Natalia Feiner-Gracia
- R. Alis OleaR. Alis OleaNanoscopy for Nanomedicine Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Carrer Baldiri Reixac 15-21, 08024 Barcelona, SpainDepartment of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The NetherlandsMore by R. Alis Olea
- Robert FitznerRobert FitznerDepartment of Mathematics and Computer Science, Eindhoven University of Technology, Post Office Box 513, 5600 MD Eindhoven, The NetherlandsMore by Robert Fitzner
- Najoua El BoujnouniNajoua El BoujnouniDepartment of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The NetherlandsMore by Najoua El Boujnouni
- Alexander H. van AsbeckAlexander H. van AsbeckDepartment of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The NetherlandsMore by Alexander H. van Asbeck
- Roland Brock*Roland Brock*E-mail: [email protected]Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The NetherlandsMore by Roland Brock
- Lorenzo Albertazzi*Lorenzo Albertazzi*E-mail: [email protected]Nanoscopy for Nanomedicine Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Carrer Baldiri Reixac 15-21, 08024 Barcelona, SpainDepartment of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5612AZ Eindhoven, The NetherlandsMore by Lorenzo Albertazzi
Abstract
The successful application of gene therapy relies on the development of safe and efficient delivery vectors. Cationic polymers such as cell-penetrating peptides (CPPs) can condense genetic material into nanoscale particles, called polyplexes, and induce cellular uptake. With respect to this point, several aspects of the nanoscale structure of polyplexes have remained elusive because of the difficulty in visualizing the molecular arrangement of the two components with nanometer resolution. This limitation has hampered the rational design of polyplexes based on direct structural information. Here, we used super-resolution imaging to study the structure and molecular composition of individual CPP-mRNA polyplexes with nanometer accuracy. We use two-color direct stochastic optical reconstruction microscopy (dSTORM) to unveil the impact of peptide stoichiometry on polyplex structure and composition and to assess their destabilization in blood serum. Our method provides information about the size and composition of individual polyplexes, allowing the study of such properties on a single polyplex basis. Furthermore, the differences in stoichiometry readily explain the differences in cellular uptake behavior. Thus, quantitative dSTORM of polyplexes is complementary to the currently used characterization techniques for understanding the determinants of polyplex activity in vitro and inside cells.
Cell-penetrating peptides (CPPs), also known as protein transduction domains, are a very promising class of vectors for gene delivery (1) because of their ability to facilitate cell entry of conjugated molecules. (2,3) Moreover, because of their positive charge, CPPs are ideal candidates for the noncovalent complexation of negatively charged oligonucleotides into nanosized particles named polyplexes. (4) The potential of polyplexes to deliver different oligonucleotide cargo such as plasmid DNA or RNA into cells for gene therapy has been widely explored. (5−7) Recently, mRNA has emerged as another promising option. (8−10) Polyplexes of mRNA only need to penetrate cells and escape from the endosomes or lysosomes to the cytoplasm, where mRNA gets translated immediately into proteins. Furthermore, mRNA is only transiently active without the risk of insertion into the host genome, and it is fully degraded via physiological mechanisms. Therefore, mRNA-loaded polyplexes have recently been proposed as efficient carriers for oligonucleotide therapy. (8−10)
Despite the many gene carriers proposed and studied, the design of the polyplexes is still very challenging, and activity varies greatly. (11) This may also be attributed to the fact that the structure space of delivery vectors has been navigated with a limited understanding of the structural properties and variations of the fabric of a polyplex. In fact, only a handful of methods for polyplex characterization are currently available. The average hydrodynamic size and charge of the polyplexes can be measured using dynamic light scattering (DLS) and zeta potential or electrophoretic shift assays, respectively. (7) Also, images from electron microscopy (EM) and atomic force microscopy (AFM) can indicate the polyplex size and shape, (12) thus providing information on morphological changes. (13,14) The efficiency of complexation is evaluated using gel electrophoresis by detecting the presence of free oligonucleotides, while the affinity is currently measured using a dye exclusion assay. (7) Overall, each of the current methods supplies valuable information; however, they exhibit two main limitations: (i) they are mostly ensemble techniques providing average information for the polyplexes and (ii) they are not able to distinguish between the two components of the polyplex and therefore miss quantitative and qualitative aspects of the composition and inner organization of individual polyplexes. Therefore, only very limited data is available for composing molecular models of the structure and the composition of polyplexes. (15−17)
Here, we propose the use of two-color direct stochastic optical reconstruction microscopy (dSTORM) to unveil the structure and composition of polyplexes on a single-particle basis. Recently, it has been demonstrated that dSTORM can be used to image synthetic nanoparticles with different chemical natures and sizes. (18−20) The nanometric resolution, down to 20 nm, provided by this technique together with the possibility to obtain two-color images is very promising for the characterization of multicomponent nanometric assemblies such as polyplexes. With the appropriate knowledge of the photophysical properties of the dye such as the duty cycle and bleaching rate, STORM provides the possibility to obtain quantitative information about stoichiometries and absolute molecule numbers. In this framework, membrane receptors, (21−23) DNA origami DNA strands, (24) and accessible binding sites on endocytic/endosomal vesicles (25) were quantified. Therefore, dSTORM can also contribute to the quantitative understanding of polyplexes. Despite this potential, to the best of our knowledge, the use of super-resolution for the study of gene delivery carriers has not been explored.
One of the most important experimental degrees of freedom in polyplex generation is the ratio of the cationic moiety and nucleic acids, which determines the N/P ratio, the ratio of positive over negative charge carriers. The N/P ratio determines several key properties of the formed polyplexes such as the net charge, size, and stability. (11,26) We considered the N/P ratio to be an ideal test case to establish a direct quantitative analysis of polyplex composition using super-resolution dSTORM microscopy. Gene-delivery carriers consisting of mRNA complexed with the peptide nona-arginine (R9) were imaged with molecular resolution. Owing to the ability to simultaneously measure the size and composition of polyplexes, our results show both qualitative and quantitative differences in the content of mRNA and CPPs for different N/P ratios. Moreover, dSTORM imaging can be performed in a complex matrix, which allowed us to visualize changes in polyplex structure in blood serum. This is key information becauase one of the reasons for the failure of gene carriers in vivo is the lack of stability in blood. Finally, we demonstrate that the information on polyplex stoichiometry obtained by STORM readily explains quantitative differences in the cellular uptake of the polyplexes. Our results highlight the potential of super-resolution microscopy to play a pivotal role in the study of polyplexes, complementing the existing methodologies and contributing to the rational design of new structures to enhance cell transfection.
For our dSTORM analysis, we prepared polyplexes formed by the CPP l-nona-arginine (R9) with a 1929-nucleotide-long mRNA coding for firefly luciferase. R9 is a well-known cell-penetrating peptide, and its capacity to complex and induce the cellular uptake of oligonucleotides has been previously studied. (11,27,28) An overview of our procedure is reported in Figure 1a. To perform dSTORM imaging, both components were labeled using the dSTORM-compatible, spectrally well-separated dyes, AlexaFluor488 and Cyanine 5 for R9 and mRNA, respectively. The peptide was labeled at the N-terminus. The mRNA was stochastically labeled with Cy5-UTP with an average labeling ratio of 27 dyes per molecule, as estimated from UV/vis absorption measurements (as detailed in Supporting Information). The labeling ratio (i.e,. the proportion of labeled molecules) was optimized to obtain an optimal signal density for dSTORM, as described extensively in the Materials and Methods section. STORM images of polyplexes with nonlabeled molecules were obtained as a control, showing almost no localization (Supporting Information Figure S6). After complexation, the polyplexes were deposited on a glass coverslip for dSTORM imaging. Figure 1B,C shows representative images of R9-mRNA polyplexes at an N/P ratio of 5. We had seen before that well-defined polyplexes were formed for N/P ratios larger than 3.
Figure 1
Figure 1. Imaging of polyplexes using dSTORM microscopy. (A) Schematic representation of polyplex formation from mRNA-Cy5 and AlexaFluor488-R9, with imaging and analysis. (B) Conventional fluorescent image of polyplexes at N/P 5 (red represents mRNA molecules and green R9 molecules). (C) dSTORM imaging of polyplexes, with the same field of view as in B. Scale bar 2 μm. (D) Close-up images of three different areas showing low-resolution data on the left and high-resolution data on the right. Scale bar 400 nm.
The direct comparison between conventional (B) and dSTORM (C) microscopy images shows how the improvement of resolution allows individual polyplexes to be resolved, while in the conventional image it is impossible to distinguish a single polyplex from clusters. Images revealed complexes of 50–75 nm in radius, in agreement with DLS measurements (Supporting Information Figure S7). Interestingly, a large amount of green signal (which corresponds to the peptide) was not associated with the polyplexes, indicating that not all of the peptide used in the complexation procedure was incorporated. This observation is of special interest as most of the other characterization methods (e.g., DLS, TEM, AFM, and gel electrophoresis) cannot detect individual peptides and are strongly biased toward the macromolecular assemblies. Notably, free cationic moieties may play a role in the transfection and/or toxicity. As an example, previous studies of Vaidyanathan et al. proved the enhanced pDNA endosomal escape of polyplexes due to the addition of free PEI polymer. (29) Moreover, for acylated TP10 analogs, nonincorporated peptide at higher N/P ratios showed enhanced toxicity. (30) Therefore, being able to visualize the noncomplex materials is of crucial importance.
In addition, dSTORM imaging allowed the study of the distribution of the peptide and the mRNA within the polyplexes as magnified in Figure 1D. The observed overlap of the two colors indicates that mRNA and R9 molecules are homogeneously intertwined inside the polyplex, at least within the resolution of dSTORM. A dSTORM 3D image was acquired to prove these observations and rule out the effect of the projection, and a middle stack of a polyplex in Supporting Information Figure S8 shows that both molecules are indeed intertwined. This result proves that mRNA molecules are not exposed on the polyplex surface but are shielded within the structure. This information is of utmost importance when designing effective gene delivery systems because confined mRNA will be more stable than exposed mRNA. (31)
Having successfully obtained super-resolution images of polyplexes formed at N/P 5, we further aimed to compare the structure and distribution of mRNA and R9 molecules for different N/P ratios. For this purpose, we imaged polyplexes formed at N/P 1, 3, 5, and 7. The mRNA concentration was maintained at a constant value in order to follow the complexation of the same amount of oligonucleotides. In Figure 2A, a representative image of each condition is shown. At N/P 1, the mRNA-Cy5 signal was not clustered, resulting in more spread structures compared to the images obtained for N/P 5. Only minimal mRNA signal colocalized with the Alexa488-R9 signal and mostly free mRNA and free peptides were present, suggesting that the amount of R9 present at N/P 1 was not sufficient to condense mRNA into polyplexes. The distribution of mRNA changed dramatically at N/P 3, for which clear rounded clusters of mRNA-Cy5 were present. Therefore, we concluded that the R9 concentration present at N/P 3 was sufficient to compact the mRNA into polyplexes. The N/P 5 and N/P 7 polyplexes clearly contained more R9 than the N/P 3 ones. Both N/P 5 and N/P 7 were very similar to each other, which indicates that the charges in the mRNA were already saturated and the extra R9 remained in solution and was not incorporated into the polyplexes. Overall, this qualitative evaluation of dSTORM images of polyplexes formed at different N/P ratios provided important structural information regarding the mRNA and R9 interplay in the formation of polyplexes.
Figure 2
Figure 2. Structure and stoichiometry differences of polyplexes. (A) dSTORM images of polyplexes prepared at N/P ratios of 1, 3, 5, and 7 from left to right. (B) Frequency histograms of the size of the polyplexes quantified from the STROM images for each N/P ratio. (C) Box plot comparing the number of localizations of mRNA and peptide quantified for each N/P ratio together with the quantification of single molecules and the number of localizations obtained from the simulations. Statistical analyses were run (Mann–Whitney test), showing significant differences between the peptide content in NP3 and NP5 (p < 0.0001) and the mRNA content between NP1 and NP3 (p < 0.0001). Scale bar 200 nm.
In dSTORM, the image reconstruction is based on single-molecule localizations detected during the imaging. (32) Therefore, dSTORM provides a highly powerful approach to analyzing the polyplexes not only qualitatively but also quantitatively. (33) Using an adapted version of a previously developed MatLab script, (34) the localizations in the mRNA-Cy5 channel were clustered in order to remove the free R9 localizations on the glass coverslip and to provide an estimation of the localizations of mRNA and CPP molecules at different complexation ratios. (The procedures are described in the SI.) In Figure 2B, the size of the polyplexes quantified for each N/P ratio is plotted on a frequency histogram. The radius of the polyplexes was between 60 and 80 nm under all conditions; however, the polydispersity of N/P 1 was significantly higher compared to that of the other ratios. N/P 1 polyplexes had a significant population of polyplexes exceeding 100 nm in size, indicating that under this condition most of the mRNA molecules were not packaged. For N/P 3 to 7, the formation of polyplexes reduced the size of the structures, also obtaining more monodisperse populations with no significant differences between N/P 3 and 7. Notably, for all conditions, the sizes quantified by dSTORM matched the DLS ensemble measurements (Supporting Information Figure S7).
In Figure 2C, the median number of localizations per polyplex detected for each N/P ratio, which is proportional to the number of molecules of the specific component, is plotted to unveil the composition of the polyplexes for the different conditions. More than 15 000 polyplexes were measured per N/P ratio. The frequency histogram of the number of localizations is represented in Supporting Information Figures S9–S12.
To better understand the data in Figure 2C, the median number of localizations per polyplex was plotted together with two more conditions: (i) polyplexes with only one labeled mRNA molecule or only one labeled R9 molecule and (ii) simulated data obtained from a stochastic blinking model. (See the Supporting Information for further details.) The results for polyplexes with only one labeled molecule are crucial to interpreting the data from the measured polyplexes because they provide a reference for the estimation of the number of molecules per polyplex (e.g., they distinguish single uncomplexed RNA strands from multiple RNAs in a particle). On the other hand, the simulations can unmask the stochastics of the dSTORM blinking, one of the main issues of dSTORM quantification, and provide the possibility to compare the median number of localizations that was detected experimentally with a theoretical framework. As previously shown, (35) stochastic simulations can predict the expected number and distribution of localizations of a set number of molecules. Therefore, comparing the number of localizations per polyplex that were experimentally detected to simulated data can provide a more accurate estimation of the composition of individual polyplexes. To obtain the simulated data presented in Figure 2C, the following steps were performed: (i) we acquired dSTORM images of polyplexes with a single labeled R9 molecule from which we estimated the photophysical parameters of the AlexaFluor488 dye; (ii) we introduced these parameters together with the known experimental data such as the number of frames and the number of polyplexes to be simulated; (iii) we simulated the blinking behavior of a single R9 molecule and verified that the output number of localizations per cluster corresponded to the experimental one (detailed information on the simulation can be found in the Supporting Information); and (iv) data reported in Figure 2C was obtained by simulating the blinking behavior of a Poissonian population of polyplexes with a specific number of peptides. The distributions of simulated and experimentally detected localizations were compared to determine which simulated peptide number matched the peak of the experimental data (e.g., higher peptide numbers for high N/P ratios). Notably, for mRNA no simulation was conducted because of the heterogeneous labeling of the mRNA, which together with the stochastic nature of the detection could not be approximated.
From the experimental and simulated data presented in Figure 2C, comprehensive information on the polyplex structure and composition can be obtained. For N/P 1, the number of mRNA localizations was comparable to that for a single molecule supporting the hypothesis that single mRNA molecules were present that were not complexed into polyplexes. For N/P 3, the experimentally determined median number of mRNA localizations significantly increased to about 2 to 3 times that of the single molecule and remained stable at N/P ratios of 5 and 7. These results indicate that inside each polyplex there were only a few mRNA molecules. Interestingly, from N/P 3 to N/P 5 the box width was enlarged, which indicated a high polydispersity with respect to the number of encapsulated mRNAs. In contrast, the density of the peptides followed different behavior compared to that of the mRNA molecules. At N/P 1, the number of R9 localizations per polyplex was very low and similar to that of a single peptide. Considering the labeling density (about 1.25% of labeled peptides were mixed with 98.75% unlabeled ones), this is compatible with individual mRNA molecules with about 100 peptides attached, an insufficient number to compact the oligonucleotides into a polyplex. At N/P 3, the number of R9 localizations slightly increased, but it was not until N/P 5 that the number abruptly grew and remained stable for higher ratios. At N/P 5 and N/P 7, the box widths were similar and again larger than that at N/P 3. The median number of simulated localizations matched the experimental data, demonstrating the validity of our model. The number of peptides for which the simulated number of localizations best matched the experimental data was about 25–30 labeled peptides per polyplexes; considering that 1.25% of the peptides were labeled, we can estimate approximately 2000–2400 peptides per polyplex. Still, for N/P 5 and 7 the box plot was broader. The simulated data always corresponded to the lower range of the plots. The frequency histograms of the number of localizations in Supporting Information Figures S9–S11 show the presence of a major population in the experimental sample, which fits the simulated data, together with a tail of polyplexes with a larger number of molecules that cannot be fitted to the simulated data. The fact that the experimental data was always broader demonstrates heterogeneity in the samples, which the simulation did not recapitulate. Thus, the measured heterogeneity cannot be simply explained by the stochastic behavior of the dye; therefore, it can be concluded that the polyplex formulation does not consist of a single monodisperse population but rather of a mixture where polyplexes of different sizes and RNA quantities coexist.
To summarize these data in a molecular picture, R9 binds mRNA at N/P 1, but only from N/P 3 is this binding sufficient to condense the mRNA into polyplexes. Polyplexes from N/P 3 to N/P 7 are similar in size and amount of mRNA but change significantly with respect to the amount of peptide present in the particles. Moreover, a significant heterogeneity of the molecular composition is present, especially at N/P 5 and 7. Notably, the differences between the last three conditions and the heterogeneity in the samples would have been difficult or impossible to measure with other methods, showing the potential of dSTORM to complement existing technique for polyplex characterization.
Above, the median number of localizations per component and condition was analyzed to determine the approximate number of molecules per polyplex and the polydispersity of the samples. However, dual-color dSTORM simultaneously provides information regarding the number of localizations for each component and the size for each individual polyplex. Figure 3A,B shows a correlative analysis of these three variables. The scatter plots in Figure 3A represents the data from over 15 000 polyplexes: every dot in the chart represents one polyplex that is positioned in the graph depending on its amount of R9 or mRNA localizations. Moreover, every point is plotted in a different color depending on the size of the polyplex. With this representation, we can draw conclusions about the relationship between the size and amount of the two components on a single-particle basis and thus with respect to the heterogeneity of the sample. At N/P 1, the mRNA localization is rather monodisperse compared to that of other samples, corresponding to the presence of single mRNA molecules. At N/P 3, the number of mRNA localizations significantly increased as did the polydispersity of the mRNA content, indicating the existence of polyplexes with multiple RNA molecules. Moreover, a correlation between the amounts of R9 and mRNA is present. This is even more evident for N/P ratios 5 and 7, where the number of R9 localizations increased overall. In particular, the polyplexes with higher mRNA localizations are the ones that show a pronounced increase in the number of R9 localizations. Following this pattern, at N/P 5 the polyplexes with high mRNA localizations display an increase in the R9 localizations, and at N/P 7, a similar correlation was observed, suggesting that the charges were saturated at N/P 5. These results prove a clear correlation in the number of molecules of each component inside individual polyplexes.
Figure 3
Figure 3. Correlation between the number of mRNA localizations and R9 localizations. (A) Scatter plots of the number of mRNA localizations versus the number of R9 localizations, together with a color code representing the size for each polyplex. (B) Number of localizations plotted as a function of polyplex size.
Moreover, the color code provides information on the interplay between composition and size. In all of he N/P studied, higher amounts of mRNA and R9 were found in larger polyplexes (green-blue color). The plots in Figure 3B, where the RNA and R9 content are plotted against size, also confirm this. However, the data also indicate that the density of the packing inside the polyplexes must be heterogeneous. Within the size category of a 65–85 nm radius, polyplexes differ in volume by a factor of maximally 2.2 while the number of localization varies by up to a factor of 8. Similarly, with increasing N/P more localizations for both mRNA and peptide are found for polyplexes of the same size.
Moreover, we calculated the ratio of R9 localizations per mRNA localizations and found that it was constant for N/P 1 and 3 but slightly increased with higher radii at N/P 5 and 7 (Supporting Information Figure S13). These results indicate that the density of polyplexes prepared at the same N/P is overall similar, although there is a trend of higher ratios at higher radii.
Importantly, the estimated number of about 2000 peptides per polyplex is fully consistent with the increase in mRNA localizations by about a factor of 3 when going from a single mRNA molecule to a polyplex. Two thousand peptides correspond to 18 000 positive charges, which, at an N/P ratio of 3, can accommodate 6000 negative charges. Because the mRNA is 1929 nucleotides in length, about 3 mRNA molecules carry this charge.
Altogether our measurements depict some important structural aspects of polyplexes: (i) a significant heterogeneity is present, with polyplexes varying significantly in size and composition; (ii) a correlation between the two components is present, with more R9 necessary to pack a higher copy number of mRNA inside the same polyplexe; and (iii) higher mRNA content results in larger sizes.
Next, we aimed to assess whether the STORM analyses could lead to a better understanding of cell biological experiments. For this purpose, polyplexes were formed with Cy5-mRNA and fluorescein-labeled R9. We had shown before that fluorescein and Alexa488-labeled R9 yield equivalent results. (36) In this case, fluorescein-labeled R9 was mixed with unlabeled peptide in a ratio of 1:10. As confirmed by DLS, polyplexes with diameters ranging from 70 to 130 nm were formed for all N/P ratios. HeLa cells were incubated with polyplexes at constant concentrations of mRNA, resulting in increasing concentrations of peptide. These concentrations were proved to be noncytotoxic for cells at any of the N/P ratios (Supporting Information Figure S14). Then, intracellular fluorescence was recorded by confocal microscopy after 1 h of incubation (Figure 4A). For N/P 1, corresponding to a concentration of fluorescein-labeled peptide of 0.05 μM, hardly any cell-associated fluorescence could be detected. From N/P 3, vesicular fluorescence was presented that further increased to an N/P ratio of 5 with no further increase to N/P 7 (Figure 4A). Quantitative image analysis demonstrated that both the mean intensity of vesicular fluorescence (Figure 4B) and the total number of vesicular pixels per cell as a measure of the number of endosomes increased from N/P 1 to N/P 5 (Figure 4C). At N/P 1, almost no polyplex was internalized, and at N/P 3, the amount increased, as it also did at N/P 5 and 7. These data demonstrate that at N/P 5 the polyplexes had the right composition to be internalized.
Figure 4
Figure 4. Dependence of cellular nanoparticle uptake on the N/P ratio. HeLa cells were incubated for 1 h with polyplexes consisting of Cy5-mRNA and fluorescein-labeled R9 at the indicated N/P ratios, followed by washing and confocal microscopy of living cells. Labeled peptide was mixed with unlabeled peptide in a ratio of 1:10. The mRNA concentration was constant at 4.5 pM for all N/P ratios, corresponding to peptide concentrations of 0.5, 1.5, 2.5, and 3.5 μM for N/P 1, 3, 5 and 7, respectively. (A) Cellular uptake and intracellular distribution. To facilitate the discrimination of intensities, a false color look-up table has been employed. The scale bar corresponds to 20 μm. (B) Average pixel intensities for vesicular fluorescence. (C) Pixels per cell as a measure of the number and size of endocytic vesicles. (D) Average pixel intensity for fluorescence outside cells, recorded at a higher detector gain. (E) Ratio of peptide over mRNA fluorescence intensity. Ratios were calculated for the vesicular intensities of each analysis image and averaged. Error bars correspond to the standard deviation for normalized data of two independent experiments.
Even though the cells were washed in order to reduce extracellular out-of-focus fluorescence, there was still some fluorescence left. For the peptide, this extracellular fluorescence strongly increased with increasing N/P ratio, consistent with the STORM results that had demonstrated the incomplete incorporation of peptide from N/P ratios larger than 3 (Figure 4D). Moreover, we divided both mean fluorescence intensities (peptide vs mRNA) to assess the ratio of peptide per mRNA in the internalized poyplexes (Figure 4E). Interestingly, from N/P 3 to 7 this value remained constant. This observation proves that a specific composition of the polyplexes is needed to be internalized by the cells. From our STORM data, we demonstrated that at N/P 3 most of the polyplexes did not have enough peptide incorporated to pack the mRNA; therefore, we hypothesis that only a small population of polyplexes at N/P 3 have the right composition, which corresponds to the lower internalization observed. Finally, the higher ratio at N/P 1 is consistent with the absence of polyplex formation so that only free peptide was internalized.
Having established the methodology to measure the polyplex size and composition, we exploited the ability of dSTORM to perform analyses in complex media such as blood serum. Polyplexes are designed to be intravenously injected into the bloodstream, being in contact with proteins and other biomolecules that can destabilize the complex. Therefore, it is of major importance to understand the stability of the polyplexes in serum. Classically, gel retardation assays are used to assess the release and decomposition of polyplexes. (7) Using dSTORM imaging, we expected to obtain information on the mechanism of decomplexation because we were able to track the composition and size of polyplexes. To this end, we incubated polyplexes at 37 °C with fetal bovine serum (FBS) at concentrations of 1, 20, and 100% for 1 min and 1 h followed by deposition on coverslips. Then, dSTORM images of the complexes were acquired (Figure 5 and Supporting Information Figure S15). The behavior of the polyplexes was similar when incubated with 1 or 20% FBS. Under these conditions, after 1 min the mRNA content in the polyplexes remained stable compared to the initial composition, indicating that the polyplexes were not releasing their payload. However, we observed a decrease in the number of peptide molecules. After 1 min of serum incubation, only half of the initial peptide localizations were detected. In contrast, after 1 h of FBS incubation both the numbers of mRNA and R9 molecules abruptly dropped (Figure 5A–C). Altogether, these results indicate that the serum proteins interact with the polyplexes in two stages as schematically represented in Figure 5D: (i) serum rapidly removes peptides from the polyplexes without compromising the mRNA compacted inside the particle (minute timescale) and (ii) proteins destabilize the complexes causing the release of mRNA molecules (hour timescale). We envision that the ability of dSTORM to provide additional information about the mechanism of polyplex disassembly will support the study and design of gene carriers with improved serum stability.
Figure 5
Figure 5. Stability of polyplexes in serum. (A) dSTORM image of the N/P 5 polyplexes initially and after being incubated with 20% FBS for 1 min and 1 h. Scale bar 200 nm. (B) Number of mRNA localizations of N/P 5 polyplexes after being incubated with 20% FBS for 1 min and 1 h. (C) Number of R9 localizations of N/P 5 polyplexes after being incubated with 20% FBS for 1 min and 1 h. (D) Schematic representation of the destabilization of the polyplexes in time in the presence of serum.
We presented an in-depth study of the structure and composition of polyplexes containing mRNA and cell-penetrating peptide R9 using dSTORM imaging. Our results show both qualitative and quantitative differences in the mRNA and peptide content of individual polyplexes formed at different N/P ratios. Importantly, a minimum excess of peptide was required to condense the mRNA into polyplexes because at N/P 1 no distinct clusters could be observed. At N/P 3, the number of peptides was sufficient to pack the mRNA into compact polyplexes. With an increase in N/P, the R9 content and peptide to mRNA ratio in the polyplexes further increased, reaching a plateau at around N/P 5. The possibility to image individual polyplexes allowed us to dissect the relationship between the relative content of the two components and the size of the polyplexes, demonstrating a next-to-positive correlation of both components with respect to size, some heterogeneity in the packing density, and an increase in the packing density with increasing N/P ratio. Our analyses clearly demonstrate that the insights gained through the STORM analysis of the polyplexes directly translate into a better understanding of cellular uptake experiments. From N/P 5 to 7, the characteristics of the polyplexes are the same, which explains why they have the same cellular uptake efficiency. At N/P 3, sufficient mRNA encapsulation had been achieved for only a fraction of the polyplexes as evident from the large heterogeneity of polyplexes at the molecular level, demonstrated by STORM. Finally, the imaging of the destabilization of polyplexes in the presence of serum provided mechanistic insights into the disassembly mechanism, a crucial aspect for the in vivo performance of gene carriers. Our study highlights the potential of multicolor super-resolution microscopy to shed new light on the nanoscale structure and stoichiometry of polyplexes, a key aspect for the understanding of gene delivery carriers. Future perspectives include a study of structure–activity relationships for a variety of polymeric, lipidic, and peptidic carriers and a study of the structure of polyplexes in cells after internalization. In this framework it has to be considered that STORM imaging is possible only in fixed cells and therefore optimization of the fixation procedure to retain the polyplex structure is necessary. In conclusion, we believe that super-resolution microscopy can nicely complement the current techniques for polyplex characterization contributing to the guidance of the design of more effective oligonucleotide therapeutics.
Supporting Information
The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acs.nanolett.8b04407.
Materials and methods, a detailed explanation of the simulations used, and additional experimental results (PDF)
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.
Acknowledgments
L.A. thanks the Spanish Ministry of Economy, Industry and Competitiveness, through Project SAF2016-75241-R, as part of the Generalitat de Catalunya through the CERCA program. R.A.O. received scholarships from the Radboudumc scholarship fund and the Erasmus+ programme. The authors also acknowledge the EuroNanoMed II platform through project NANOVAX and the foundation Obra Social La Caixa. The authors acknowledge Remco van der Hofstad for useful discussions. L.A. thanks the the European Research Council (ERC-StG- 757397).
dSTORM | direct stochastic optical reconstruction microscopy |
CPP | cell-penetrating peptide |
DLS | dynamic light scattering |
R9 | l-nona-arginine |
FBS | fetal bovine serum |
References
This article references 36 other publications.
- 1Koren, E.; Torchilin, V. P. Trends Mol. Med. 2012, 18 (7), 385– 393, DOI: 10.1016/j.molmed.2012.04.012Google ScholarThere is no corresponding record for this reference.
- 2Collado Camps, E.; Brock, R. Bioorg. Med. Chem. 2018, 26 (10), 2780– 2787, DOI: 10.1016/j.bmc.2017.11.004Google ScholarThere is no corresponding record for this reference.
- 3Heitz, F.; Morris May, C.; Divita, G. Br. J. Pharmacol. 2009, 157 (2), 195– 206, DOI: 10.1111/j.1476-5381.2009.00057.xGoogle Scholar3Twenty years of cell-penetrating peptides: from molecular mechanisms to therapeuticsHeitz, Frederic; Morris, May Catherine; Divita, GillesBritish Journal of Pharmacology (2009), 157 (2), 195-206CODEN: BJPCBM; ISSN:1476-5381. (Wiley-Blackwell)A review. The recent discovery of new potent therapeutic mols. that do not reach the clinic due to poor delivery and low bioavailability have made of delivery a key stone in therapeutic development. Several technologies have been designed to improve cellular uptake of therapeutic mols., including cell-penetrating peptides (CPPs). CPPs were first discovered based on the potency of several proteins to enter cells. Numerous CPPs have been described so far, which can be grouped into two major classes, the first requiring chem. linkage with the drug for cellular internalization and the second involving formation of stable, non-covalent complexes with drugs. Nowadays, CPPs constitute very promising tools for non-invasive cellular import of cargo and have been successfully applied for in vitro and in vivo delivery of therapeutic mols. varying from small chem. mol., nucleic acids, proteins, peptides, liposomes and particles. This review will focus on the structure/function and cellular uptake mechanism of CPPs in the general context of drug delivery. We will also highlight the application of peptide carriers for the delivery of therapeutic mols. and provide an update of their clin. evaluation.
- 4Lee, S. H.; Castagner, B.; Leroux, J.-C. Eur. J. Pharm. Biopharm. 2013, 85 (1), 5– 11, DOI: 10.1016/j.ejpb.2013.03.021Google Scholar4Is there a future for cell-penetrating peptides in oligonucleotide delivery?Lee, Soo Hyeon; Castagner, Bastien; Leroux, Jean-ChristopheEuropean Journal of Pharmaceutics and Biopharmaceutics (2013), 85 (1), 5-11CODEN: EJPBEL; ISSN:0939-6411. (Elsevier B.V.)A review. Cell-penetrating peptides have been widely investigated as delivery vehicles for oligonucleotides (e.g., siRNA and antisense oligonucleotides). Different delivery strategies can be used, such as co-incubation, direct conjugation, non-covalent complex, and modification on the surface of liposome or polymer complexes. However, several challenges remain for their preclin. and clin. development. Endosomal escape, lack of cell/tissue specificity, and toxicity are major concerns in the design of cell-penetrating peptide-mediated delivery systems. In this commentary, we highlight recent reports of cell-penetrating peptide incorporation into oligonucleotide delivery systems and underline the remaining challenges, particularly for preclin. and clin. applications.
- 5Zhang, P.; Wagner, E. Top. Curr. Chem. 2017, 375 (2), 26, DOI: 10.1007/s41061-017-0112-0Google ScholarThere is no corresponding record for this reference.
- 6Lächelt, U.; Wagner, E. Chem. Rev. 2015, 115 (19), 11043– 11078, DOI: 10.1021/cr5006793Google ScholarThere is no corresponding record for this reference.
- 7Scomparin, A.; Polyak, D.; Krivitsky, A.; Satchi-Fainaro, R. Biotechnol. Adv. 2015, 33 (6, Part 3), 1294– 1309, DOI: 10.1016/j.biotechadv.2015.04.008Google ScholarThere is no corresponding record for this reference.
- 8Debus, H.; Baumhof, P.; Probst, J.; Kissel, T. J. Controlled Release 2010, 148 (3), 334– 343, DOI: 10.1016/j.jconrel.2010.09.007Google ScholarThere is no corresponding record for this reference.
- 9Meng, Z.; O’Keeffe-Ahern, J.; Lyu, J.; Pierucci, L.; Zhou, D.; Wang, W. Biomater. Sci. 2017, 5 (12), 2381– 2392, DOI: 10.1039/C7BM00712DGoogle Scholar9A new developing class of gene delivery: messenger RNA-based therapeuticsMeng, Zhao; O'Keeffe-Ahern, Jonathan; Jing, Lyu; Pierucci, Luca; Zhou, Dezhong; Wang, WenxinBiomaterials Science (2017), 5 (12), 2381-2392CODEN: BSICCH; ISSN:2047-4849. (Royal Society of Chemistry)A review. Gene therapy has long been held as having the potential to become a front line treatment for various genetic disorders. However, the direct delivery of nucleic acids to correct a genetic disorder has numerous limitations owing to the inability of naked nucleic acids (DNA and RNA) to traverse the cell membrane. Recently, mRNA (mRNA) based delivery has become a more attractive alternative to DNA due to the relatively easier transfection process, higher efficiency and safety profile. As with all gene therapies, the central challenge that remains is the efficient delivery of nucleic acids intracellularly. This review presents the recent progress in mRNA delivery, focusing on comparing the advantages and limitations of non-viral based delivery vectors.
- 10Sahin, U.; Karikó, K.; Türeci, Ö. Nat. Rev. Drug Discovery 2014, 13 (10), 759– 780, DOI: 10.1038/nrd4278Google ScholarThere is no corresponding record for this reference.
- 11van Asbeck, A. H.; Beyerle, A.; McNeill, H.; Bovee-Geurts, P. H. M.; Lindberg, S.; Verdurmen, W. P. R.; Hällbrink, M.; Langel, Ü.; Heidenreich, O.; Brock, R. ACS Nano 2013, 7 (5), 3797– 3807, DOI: 10.1021/nn305754cGoogle ScholarThere is no corresponding record for this reference.
- 12Lee, J. B.; Hong, J.; Bonner, D. K.; Poon, Z.; Hammond, P. T. Nat. Mater. 2012, 11 (4), 316– 322, DOI: 10.1038/nmat3253Google ScholarThere is no corresponding record for this reference.
- 13Patil, M. L.; Zhang, M.; Taratula, O.; Garbuzenko, O. B.; He, H.; Minko, T. Biomacromolecules 2009, 10 (2), 258– 266, DOI: 10.1021/bm8009973Google Scholar13Internally Cationic Polyamidoamine PAMAM-OH Dendrimers for siRNA Delivery: Effect of the Degree of Quaternization and Cancer TargetingPatil, Mahesh L.; Zhang, Min; Taratula, Oleh; Garbuzenko, Olga B.; He, Huixin; Minko, TamaraBiomacromolecules (2009), 10 (2), 258-266CODEN: BOMAF6; ISSN:1525-7797. (American Chemical Society)A novel cancer targeted, internally cationic and surface neutral polyamidoamine (PAMAM) dendrimer, was designed, synthesized, and evaluated as a nanocarrier for the targeted intracellular delivery of siRNA. The dendrimer contained a synthetic analog of LH-releasing hormone as cancer targeting moiety. The proposed delivery system possesses the following advantages: (1) internal cationic charges for complexation with siRNA and enhanced siRNA protection; (2) low cytotoxicity; (3) lesser degree of quaternization offering free tertiary amines for potential proton sponge effect; and (4) targeting specifically to cancer cells for enhancing siRNA uptake and efficiency and potential limitation of adverse side effects of chemotherapy on healthy organs. Both nontargeted and targeted dendrimer-siRNA complexes formed compact nanometer size spherical particles, exhibited very low cytotoxicity even at the higher concn., and efficiently penetrated cancer cells in vitro. However, only the targeted dendrimer-siRNA complex was able to substantially decrease the expression of a targeted BCL2 gene.
- 14Ofek, P.; Fischer, W.; Calderón, M.; Haag, R.; Satchi-Fainaro, R. FASEB J. 2010, 24 (9), 3122– 3134, DOI: 10.1096/fj.09-149641Google ScholarThere is no corresponding record for this reference.
- 15García-Sosa, A. T.; Tulp, I.; Langel, K.; Langel, Ü. BioMed Res. Int. 2014, 2014, 257040, DOI: 10.1155/2014/257040Google ScholarThere is no corresponding record for this reference.
- 16Meneksedag-Erol, D.; Tang, T.; Uludağ, H. Biomaterials 2014, 35 (25), 7068– 7076, DOI: 10.1016/j.biomaterials.2014.04.103Google Scholar16Molecular modeling of polynucleotide complexesMeneksedag-Erol, Deniz; Tang, Tian; Uludag, HasanBiomaterials (2014), 35 (25), 7068-7076CODEN: BIMADU; ISSN:0142-9612. (Elsevier Ltd.)A review. Delivery of polynucleotides into patient cells is a promising strategy for treatment of genetic disorders. Gene therapy aims to either synthesize desired proteins (DNA delivery) or suppress expression of endogenous genes (siRNA delivery). Carriers constitute an important part of gene therapeutics due to limitations arising from the pharmacokinetics of polynucleotides. Non-viral carriers such as polymers and lipids protect polynucleotides from intra and extracellular threats and facilitate formation of cell-permeable nanoparticles through shielding and/or bridging multiple polynucleotide mols. Formation of nanoparticulate systems with optimal features, their cellular uptake and intracellular trafficking are crucial steps for an effective gene therapy. Despite the great amt. of exptl. work pursued, crit. features of the nanoparticles as well as their processing mechanisms are still under debate due to the lack of instrumentation at at. resoln. Mol. modeling based computational approaches can shed light onto the at. level details of gene delivery systems, thus provide valuable input that cannot be readily obtained with exptl. techniques. Here, we review the mol. modeling research pursued on crit. gene therapy steps, highlight the knowledge gaps in the field and providing future perspectives. Existing modeling studies revealed several important aspects of gene delivery, such as nanoparticle formation dynamics with various carriers, effect of carrier properties on complexation, carrier conformations in endosomal stages, and release of polynucleotides from carriers. Rate-limiting steps related to cellular events (i.e. internalization, endosomal escape, and nuclear uptake) are now beginning to be addressed by computational approaches. Limitations arising from current computational power and accuracy of modeling have been hindering the development of more realistic models. With the help of rapidly-growing computational power, the crit. aspects of gene therapy are expected to be better investigated and direct comparison between more realistic mol. modeling and expts. may open the path for design of next generation gene therapeutics.
- 17Majzoub, R. N.; Ewert, K. K.; Safinya, C. R. Philos. Trans. R. Soc., A 2016, 374 (2072), 20150129, DOI: 10.1098/rsta.2015.0129Google Scholar17Cationic liposome-nucleic acid nanoparticle assemblies with applications in gene delivery and gene silencingMajzoub, Ramsey N.; Ewert, Kai K.; Safinya, Cyrus R.Philosophical Transactions of the Royal Society, A: Mathematical, Physical & Engineering Sciences (2016), 374 (2072), 20150129/1-20150129/14CODEN: PTRMAD; ISSN:1364-503X. (Royal Society)Cationic liposomes (CLs) are synthetic carriers of nucleic acids in gene delivery and gene silencing therapeutics. The introduction will describe the structures of distinct liq. cryst. phases of CL-nucleic acid complexes, which were revealed in earlier synchrotron small-angle X-ray scattering expts. When mixed with plasmid DNA, CLs contg. lipids with distinct shapes spontaneously undergo topol. transitions into self-assembled lamellar, inverse hexagonal, and hexagonal CL-DNA phases. CLs contg. cubic phase lipids are obsd. to readily mix with short interfering RNA (siRNA) mols. creating double gyroid CL-siRNA phases for gene silencing. Custom synthesis of multivalent lipids and a range of novel polyethylene glycol (PEG)-lipids with attached targeting ligands and hydrolysable moieties have led to functionalized equil. nanoparticles (NPs) optimized for cell targeting, uptake or endosomal escape. Very recent expts. are described with surface-functionalized PEGylated CL-DNA NPs, including fluorescence microscopy colocalization with members of the Rab family of GTPases, which directly reveal interactions with cell membranes and NP pathways. In vitro optimization of CL-DNA and CL-siRNA NPs with relevant primary cancer cells is expected to impact nucleic acid therapeutics in vivo.
- 18van der Zwaag, D.; Vanparijs, N.; Wijnands, S.; De Rycke, R.; De Geest, B. G.; Albertazzi, L. ACS Appl. Mater. Interfaces 2016, 8 (10), 6391– 6399, DOI: 10.1021/acsami.6b00811Google ScholarThere is no corresponding record for this reference.
- 19Duro-Castano, A.; Nebot Vicent, J.; Niño-Pariente, A.; Armiñán, A.; Arroyo-Crespo, J.; Paul, A.; Feiner-Gracia, N.; Albertazzi, L.; Vicent, M. Adv. Mater. 2017, 29 (39), 1702888, DOI: 10.1002/adma.201702888Google ScholarThere is no corresponding record for this reference.
- 20Ardizzone, A.; Kurhuzenkau, S.; Illa-Tuset, S.; Faraudo, J.; Bondar, M.; Hagan, D.; Van Stryland, E. W.; Painelli, A.; Sissa, C.; Feiner, N. Small 2018, 14, 1703851, DOI: 10.1002/smll.201703851Google ScholarThere is no corresponding record for this reference.
- 21Burgert, A.; Schlegel, J.; Bécam, J.; Doose, S.; Bieberich, E.; Schubert-Unkmeir, A.; Sauer, M. Angew. Chem. 2017, 129 (22), 6227– 6231, DOI: 10.1002/ange.201700570Google ScholarThere is no corresponding record for this reference.
- 22Letschert, S.; Göhler, A.; Franke, C.; Bertleff-Zieschang, N.; Memmel, E.; Doose, S.; Seibel, J.; Sauer, M. Angew. Chem., Int. Ed. 2014, 53 (41), 10921– 10924, DOI: 10.1002/anie.201406045Google ScholarThere is no corresponding record for this reference.
- 23Fricke, F.; Beaudouin, J.; Eils, R.; Heilemann, M. Sci. Rep. 2015, 5, 14072, DOI: 10.1038/srep14072Google Scholar23One, two or three? Probing the stoichiometry of membrane proteins by single-molecule localization microscopyFricke Franziska; Heilemann Mike; Beaudouin Joel; Eils RolandScientific reports (2015), 5 (), 14072 ISSN:.Probing the oligomeric state of abundant molecules, such as membrane proteins in intact cells, is essential, but has not been straightforward. We address this challenge with a simple counting strategy that is capable of reporting the oligomeric state of dense, membrane-bound protein complexes. It is based on single-molecule localization microscopy to super-resolve protein structures in intact cells and basic quantitative evaluation. We validate our method with membrane-bound monomeric CD86 and dimeric cytotoxic T-lymphocyte-associated protein as model proteins and confirm their oligomeric states. We further detect oligomerization of CD80 and vesicular stomatitis virus glycoprotein and propose coexistence of monomers and dimers for CD80 and trimeric assembly of the viral protein at the cell membrane. This approach should prove valuable for researchers striving for reliable molecular counting in cells.
- 24Zanacchi, F. C.; Manzo, C.; Alvarez, A. S.; Derr, N. D.; Garcia-Parajo, M. F.; Lakadamyali, M. Nat. Methods 2017, 14 (8), 789– 792, DOI: 10.1038/nmeth.4342Google Scholar24A DNA origami platform for quantifying protein copy number in super-resolutionZanacchi, Francesca Cella; Manzo, Carlo; Alvarez, Angel S.; Derr, Nathan D.; Garcia-Parajo, Maria F.; Lakadamyali, MelikeNature Methods (2017), 14 (8), 789-792CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)Single-mol.-based super-resoln. microscopy offers researchers a unique opportunity to quantify protein copy no. with nanoscale resoln. However, while fluorescent proteins have been characterized for quant. imaging using calibration stds., similar calibration tools for immunofluorescence with small org. fluorophores are lacking. Here we show that DNA origami, in combination with GFP antibodies, is a versatile platform for calibrating fluorophore and antibody labeling efficiency to quantify protein copy no. in cellular contexts using super-resoln. microscopy.
- 25Puchner, E. M.; Walter, J. M.; Kasper, R.; Huang, B.; Lim, W. A. Proc. Natl. Acad. Sci. U. S. A. 2013, 110 (40), 16015– 16020, DOI: 10.1073/pnas.1309676110Google ScholarThere is no corresponding record for this reference.
- 26Ezzat, K.; EL Andaloussi, S.; Zaghloul, E. M.; Lehto, T.; Lindberg, S.; Moreno, P. M. D.; Viola, J. R.; Magdy, T.; Abdo, R.; Guterstam, P. Nucleic Acids Res. 2011, 39 (12), 5284– 5298, DOI: 10.1093/nar/gkr072Google ScholarThere is no corresponding record for this reference.
- 27Wu, R. P.; Youngblood, D. S.; Hassinger, J. N.; Lovejoy, C. E.; Nelson, M. H.; Iversen, P. L.; Moulton, H. M. Nucleic Acids Res. 2007, 35 (15), 5182– 5191, DOI: 10.1093/nar/gkm478Google ScholarThere is no corresponding record for this reference.
- 28Futaki, S.; Ohashi, W.; Suzuki, T.; Niwa, M.; Tanaka, S.; Ueda, K.; Harashima, H.; Sugiura, Y. Bioconjugate Chem. 2001, 12 (6), 1005– 1011, DOI: 10.1021/bc015508lGoogle Scholar28Stearylated Arginine-Rich Peptides: A New Class of Transfection SystemsFutaki, Shiroh; Ohashi, Wakana; Suzuki, Tomoki; Niwa, Miki; Tanaka, Seigo; Ueda, Kunihiro; Harashima, Hideyoshi; Sugiura, YukioBioconjugate Chemistry (2001), 12 (6), 1005-1011CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)Membrane-permeable arginine-rich peptides, such as HIV-1 Tat-(48-60), HIV-1 Rev-(34-50), and flock house virus (FHV) coat-(35-49), have been shown to possess the ability to transfect COS-7 cells with luciferase-coding plasmid as efficiently as polyarginine (MW 5000-15 000) and polylysine (MW 9800). Not only these virus-derived cationic peptides but also oligoarginines of 4-16 residues were found to be able to transfect cells. In the case of the Tat, FHV, and octaarginine peptides, N-terminal stearylation of the peptides increases the transfection efficiency by approx. 100 times to reach the same order of magnitude as that of LipofectAMINE, one of the most efficient com. available transfection agents. Also, a certain correlation was obsd. between the transfection efficiency of stearyl-(Arg)n peptides (stearyl-Rn: n = 4, 8, 12, 16) and the membrane permeability of the corresponding (Arg)n peptides (Rn).
- 29Vaidyanathan, S.; Chen, J.; Orr, B. G.; Banaszak Holl, M. M. Mol. Pharmaceutics 2016, 13 (6), 1967– 1978, DOI: 10.1021/acs.molpharmaceut.6b00139Google Scholar29Cationic Polymer Intercalation into the Lipid Membrane Enables Intact Polyplex DNA Escape from Endosomes for Gene DeliveryVaidyanathan, Sriram; Chen, Junjie; Orr, Bradford G.; Banaszak Holl, Mark M.Molecular Pharmaceutics (2016), 13 (6), 1967-1978CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)Developing improved cationic polymer-DNA polyplexes for gene delivery requires improved understanding of DNA transport from endosomes into the nucleus. Using a FRET-capable oligonucleotide mol. beacon (OMB), we monitored the transport of intact DNA to cell organelles. We obsd. that for effective (jetPEI) and ineffective (G5 PAMAM) vectors, the fraction of cells displaying intact OMB in the cytosol (jetPEI » G5 PAMAM) quant. predicted the fraction expressing transgene (jetPEI » G5 PAMAM). Intact OMB delivered with PAMAM and confined to endosomes could be released to the cytosol by the subsequent addn. of L-PEI, with a corresponding 10-fold increase in transgene expression. These results suggest that future vector development should optimize vectors for intercalation into, and destabilization of, the endosomal membrane. Finally, the study highlights a two-step strategy in which the pDNA is loaded in cells using one vector and endosomal release is mediated by a second agent.
- 30Lehto, T.; Vasconcelos, L.; Margus, H.; Figueroa, R.; Pooga, M.; Hällbrink, M.; Langel, Ü. Bioconjugate Chem. 2017, 28 (3), 782– 792, DOI: 10.1021/acs.bioconjchem.6b00680Google Scholar30Saturated Fatty Acid Analogues of Cell-Penetrating Peptide PepFect14: Role of Fatty Acid Modification in Complexation and Delivery of Splice-Correcting OligonucleotidesLehto, Tonis; Vasconcelos, Luis; Margus, Helerin; Figueroa, Ricardo; Pooga, Margus; Haellbrink, Mattias; Langel, UloBioconjugate Chemistry (2017), 28 (3), 782-792CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)Modifying cell-penetrating peptides (CPPs) with fatty acids has long been used to improve peptide-mediated nucleic acid delivery. In this study we have revisited this phenomenon with a systematic approach where we developed a structure-activity relationship to describe the role of the acyl chain length in the transfection process. For that we took a well-studied CPP, PepFect14, as the basis and varied its N-terminal acyl chain length from 2 to 22 carbons. To evaluate the delivery efficiency, the peptides were noncovalently complexed with a splice-correcting oligonucleotide (SCO) and tested in HeLa pLuc705 reporter cell line. Our results demonstrate that biol. splice-correction activity emerges from acyl chain of 12 carbons and increases linearly with each addnl. carbon. To assess the underlying factors regarding how the transfection efficacy of these complexes is dependent on hydrophobicity, we used an array of different methods. For the functionally active peptides (C12-22) there was no apparent difference in their physicochem. properties, including complex formation efficiency, hydrodynamic size, and zeta potential. Moreover, membrane activity studies with peptides and their complexes with SCOs confirmed that the toxicity of the complexes at higher molar ratios is mainly caused by the free fraction of the peptide which is not incorporated into the peptide/oligonucleotide complexes. Finally, we show that the increase in splice-correcting activity correlates with the ability of the complexes to assoc. with the cells. Collectively these studies lay the ground work for how to design highly efficient CPPs and how to optimize their oligonucleotide complexes for lowest toxicity without losing efficiency.
- 31Jekhmane, S.; de Haas, R.; Paulino da Silva Filho, O.; van Asbeck, A. H.; Favretto, M. E.; Hernandez Garcia, A.; Brock, R.; de Vries, R. Nucleic Acid Ther. 2017, 27 (3), 159– 167, DOI: 10.1089/nat.2016.0660Google Scholar31Virus-Like Particles of mRNA with Artificial Minimal Coat Proteins: Particle Formation, Stability, and Transfection EfficiencyJekhmane, Shehrazade; de Haas, Rob; Paulino da Silva Filho, Omar; van Asbeck, Alexander H.; Favretto, Marco Emanuele; Hernandez Garcia, Armando; Brock, Roland; de Vries, RenkoNucleic Acid Therapeutics (2017), 27 (3), 159-167CODEN: NATUCU; ISSN:2159-3337. (Mary Ann Liebert, Inc.)RNA has enormous potential as a therapeutic, yet, the successful application depends on efficient delivery strategies. In this study, we demonstrate that a designed artificial viral coat protein, which self-assembles with DNA to form rod-shaped virus-like particles (VLPs), also encapsulates and protects mRNA encoding enhanced green fluorescent protein (EGFP) and luciferase, and yields cellular expression of these proteins. The artificial viral coat protein consists of an oligolysine (K12) for binding to the oligonucleotide, a silk protein-like midblock S10 = (GAGAGAGQ)10 that self-assembles into stiff rods, and a long hydrophilic random coil block C that shields the nucleic acid cargo from its environment. With mRNA, the C-S10-K12 protein coassembles to form rod-shaped VLPs each encapsulating about one to five mRNA mols. Inside the rod-shaped VLPs, the mRNAs are protected against degrdn. by RNAses, and VLPs also maintain their shape following incubation with serum. Despite the lack of cationic surface charge, the mRNA VLPs transfect cells with both EGFP and luciferase, although with a much lower efficiency than obtained by a lipoplex transfection reagent. The VLPs have a negligible toxicity and minimal hemolytic activity. Our results demonstrate that VLPs yield efficient packaging and shielding of mRNA and create the basis for implementation of addnl. virus-like functionalities to improve transfection and cell specificity, such as targeting functionalities.
- 32Zhuang, X. Nat. Photonics 2009, 3 (7), 365– 367, DOI: 10.1038/nphoton.2009.101Google Scholar32Nano-imaging with STORMZhuang, XiaoweiNature Photonics (2009), 3 (7), 365-367CODEN: NPAHBY; ISSN:1749-4885. (Nature Publishing Group)Multicolor, three-dimensional stochastic optical reconstruction microscopy now makes it possible to image cellular structures with near mol.-scale resoln.
- 33Coltharp, C.; Yang, X.; Xiao, J. Curr. Opin. Struct. Biol. 2014, 28, 112– 121, DOI: 10.1016/j.sbi.2014.08.008Google Scholar33Quantitative analysis of single-molecule superresolution imagesColtharp, Carla; Yang, Xinxing; Xiao, JieCurrent Opinion in Structural Biology (2014), 28 (), 112-121CODEN: COSBEF; ISSN:0959-440X. (Elsevier Ltd.)This review highlights the quant. capabilities of single-mol. localization-based superresoln. imaging methods. In addn. to revealing fine structural details, the mol. coordinate lists generated by these methods provide the crit. ability to quantify the no., clustering, and colocalization of mols. with 10-50 nm resoln. Here we describe typical workflows and precautions for quant. anal. of single-mol. superresoln. images. These guidelines include potential pitfalls and essential control expts., allowing crit. assessment and interpretation of superresoln. images.
- 34Feiner-Gracia, N.; Beck, M.; Pujals, S.; Tosi, S.; Mandal, T.; Buske, C.; Linden, M.; Albertazzi, L. Small 2017, 13 (41), 1701631, DOI: 10.1002/smll.201701631Google ScholarThere is no corresponding record for this reference.
- 35Albertazzi, L.; van der Zwaag, D.; Leenders, C. M. A.; Fitzner, R.; van der Hofstad, R. W.; Meijer, E. W. Science 2014, 344 (6183), 491– 495, DOI: 10.1126/science.1250945Google Scholar35Probing Exchange Pathways in One-Dimensional Aggregates with Super-Resolution MicroscopyAlbertazzi, Lorenzo; van der Zwaag, Daan; Leenders, Christianus M. A.; Fitzner, Robert; van der Hofstad, Remco W.; Meijer, E. W.Science (Washington, DC, United States) (2014), 344 (6183), 491-495CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Supramol. fibers are prominent structures in biol. and chem. A quant. understanding of mol. exchange pathways in these one-dimensional aggregates was obtained by a combination of super-resoln. stochastic optical reconstruction microscopy and stochastic simulation. The potential of this methodol. is demonstrated with a set of well-defined synthetic building blocks that self-assemble into supramol. fibrils. Previous ensemble measurements hid all mol. phenomena underpinning monomer exchange, but the mol. pathway detd. from single-aggregate studies revealed unexpected homogeneous exchange along the polymer backbone. These results pave the way for exptl. investigation of the structure and exchange pathways of synthetic and natural supramol. fibers.
- 36Wallbrecher, R.; Ackels, T.; Olea, R. A.; Klein, M. J.; Caillon, L.; Schiller, J.; Bovée-Geurts, P. H.; van Kuppevelt, T. H.; Ulrich, A. S.; Spehr, M. J. Controlled Release 2017, 256, 68– 78, DOI: 10.1016/j.jconrel.2017.04.013Google ScholarThere is no corresponding record for this reference.
Cited By
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by ACS Publications if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
This article is cited by 33 publications.
- Rik Oude Egberink, Alexander H. van Asbeck, Milou Boswinkel, Grigor Muradjan, Jürgen Dieker, Roland Brock. Deciphering Structural Determinants Distinguishing Active from Inactive Cell-Penetrating Peptides for Cytosolic mRNA Delivery. Bioconjugate Chemistry 2023, 34
(10)
, 1822-1834. https://doi.org/10.1021/acs.bioconjchem.3c00346
- Yijiao Qu, Haiyan Zhu, Zhixing Lin, Domitilla Vanni, Sukhvir Kaur Bhangu, Brendan Dyett, Marc-Antoine Sani, Christina Cortez-Jugo, Frank Caruso, Francesca Cavalieri. Supramolecular Assembly of Polyphenols and Nucleic Acids by Thermal Cycling for Immune Cell Activation. Chemistry of Materials 2023, 35
(17)
, 7103-7112. https://doi.org/10.1021/acs.chemmater.3c01378
- Christian C. M. Sproncken, J. Rodrigo Magana, Ilja K. Voets. 100th Anniversary of Macromolecular Science Viewpoint: Attractive Soft Matter: Association Kinetics, Dynamics, and Pathway Complexity in Electrostatically Coassembled Micelles. ACS Macro Letters 2021, 10
(2)
, 167-179. https://doi.org/10.1021/acsmacrolett.0c00787
- Gregory V. Hartland (Senior Editor). Virtual Issue on Super-Resolution Far-Field Optical Microscopy. The Journal of Physical Chemistry A 2020, 124
(9)
, 1669-1672. https://doi.org/10.1021/acs.jpca.0c01500
- Gregory V. Hartland (Senior Editor). Virtual Issue on Super-Resolution Far-Field Optical Microscopy. The Journal of Physical Chemistry B 2020, 124
(9)
, 1581-1584. https://doi.org/10.1021/acs.jpcb.0c01501
- Gregory V. Hartland (Senior Editor). Virtual Issue on Super-Resolution Far-Field Optical Microscopy. The Journal of Physical Chemistry C 2020, 124
(9)
, 4927-4930. https://doi.org/10.1021/acs.jpcc.0c01502
- Silvia Pujals, Lorenzo Albertazzi. Super-resolution Microscopy for Nanomedicine Research. ACS Nano 2019, 13
(9)
, 9707-9712. https://doi.org/10.1021/acsnano.9b05289
- Xiaoming Zhang, Zhanshu Yang, Jiaxuan Lin, Wei Zhou, Nan Sun, Yi Jia. Probing Peptide Assembly and Interaction via High-Resolution Imaging Techniques: A Mini Review. International Journal of Molecular Sciences 2025, 26
(9)
, 3998. https://doi.org/10.3390/ijms26093998
- Elisa De Luca, Chiara Coricciati, Marcella Trombetta, Giuseppe Gigli, Pamela Mozetic, Alberto Rainer. Optical Nanoscopy for Elucidating Nano–Bio Interactions: A One Health Perspective. Advanced Optical Materials 2025, 20 https://doi.org/10.1002/adom.202401397
- Lauren Toms, Lorna FitzPatrick, Philip Auckland. Super-resolution microscopy as a drug discovery tool. SLAS Discovery 2025, 31 , 100209. https://doi.org/10.1016/j.slasd.2025.100209
- Xhorxhina Shaulli, Aura Maria Moreno‐Echeverri, Mariza Andoni, Eileen Waeber, Shivaprakash N. Ramakrishna, Cornelia Fritsch, Dimitri Vanhecke, Barbara Rothen‐Rutishauser, Frank Scheffold. Polymer Nano‐Carrier‐Mediated Gene Delivery: Visualizing and Quantifying DNA Encapsulation Using dSTORM. Small 2025, 21
(1)
https://doi.org/10.1002/smll.202405929
- Rasmus Münter, Jannik B. Larsen, Thomas L. Andresen. The vast majority of nucleic acid-loaded lipid nanoparticles contain cargo. Journal of Colloid and Interface Science 2024, 674 , 139-144. https://doi.org/10.1016/j.jcis.2024.06.158
- Rik Oude Egberink, Deni M. van Schie, Ben Joosten, Lisa.T.A. de Muynck, Ward Jacobs, Jenny van Oostrum, Roland Brock. Unraveling mRNA delivery bottlenecks of ineffective delivery vectors by co-transfection with effective carriers. European Journal of Pharmaceutics and Biopharmaceutics 2024, 202 , 114414. https://doi.org/10.1016/j.ejpb.2024.114414
- Sukhvir Kaur Bhangu, Liviana Mummolo, Soraia Fernandes, Alessia Amodio, Agata Radziwon, Brendan Dyett, Marco Savioli, Nitin Mantri, Christina Cortez‐Jugo, Frank Caruso, Francesca Cavalieri. Tracking the Endosomal Escape of Nanoparticles in Live Cells Using a Triplex‐Forming Oligonucleotide. Advanced Functional Materials 2024, 34
(17)
https://doi.org/10.1002/adfm.202311240
- Øystein Øvrebø, Miina Ojansivu, Kimmo Kartasalo, Hanna M. G. Barriga, Petter Ranefall, Margaret N. Holme, Molly M. Stevens. RegiSTORM: channel registration for multi-color stochastic optical reconstruction microscopy. BMC Bioinformatics 2023, 24
(1)
https://doi.org/10.1186/s12859-023-05320-1
- Lenny Van Daele, Íngrid Amer Cid, Sven Vereecken, Kristiaan Neyts, Filip Strubbe, Peter Dubruel. Single-particle electrophoresis to study the adsorption of polyplexes onto anionic particles: A model system for investigating polyplex-cell interactions. European Polymer Journal 2023, 200 , 112537. https://doi.org/10.1016/j.eurpolymj.2023.112537
- Satoshi Uchida, Chun Yin Jerry Lau, Makoto Oba, Kanjiro Miyata. Polyplex designs for improving the stability and safety of RNA therapeutics. Advanced Drug Delivery Reviews 2023, 199 , 114972. https://doi.org/10.1016/j.addr.2023.114972
- Jiangkang Xu, Fenghua Wang, Lei Ye, Rui Wang, Lixia Zhao, Xiaoye Yang, Jianbo Ji, Anchang Liu, Guangxi Zhai. Penetrating peptides: Applications in drug delivery. Journal of Drug Delivery Science and Technology 2023, 84 , 104475. https://doi.org/10.1016/j.jddst.2023.104475
- Ülo Langel. Methods for Molecular Imaging, Detection and Visualization of CPPs. 2023, 263-311. https://doi.org/10.1007/978-3-031-38731-9_10
- Benjamin Winkeljann, David C. Keul, Olivia M. Merkel. Engineering poly- and micelleplexes for nucleic acid delivery – A reflection on their endosomal escape. Journal of Controlled Release 2023, 353 , 518-534. https://doi.org/10.1016/j.jconrel.2022.12.008
- Shuaibin Chang, Divya Varadarajan, Jiarui Yang, Ichun Anderson Chen, Sreekanth Kura, Caroline Magnain, Jean C. Augustinack, Bruce Fischl, Douglas N. Greve, David A. Boas, Hui Wang. Scalable mapping of myelin and neuron density in the human brain with micrometer resolution. Scientific Reports 2022, 12
(1)
https://doi.org/10.1038/s41598-021-04093-y
- Jinsong Zhang, Tianyuan Zhang, Jianqing Gao. Biocompatible Iron Oxide Nanoparticles for Targeted Cancer Gene Therapy: A Review. Nanomaterials 2022, 12
(19)
, 3323. https://doi.org/10.3390/nano12193323
- Huapan Fang, Qian Chen. Applications and challenges of biomaterial mediated mRNA delivery. Exploration of Targeted Anti-tumor Therapy 2022, , 428-444. https://doi.org/10.37349/etat.2022.00093
- Shikha Dhiman, Teodora Andrian, Beatriz Santiago Gonzalez, Marrit M. E. Tholen, Yuyang Wang, Lorenzo Albertazzi. Can super-resolution microscopy become a standard characterization technique for materials chemistry?. Chemical Science 2022, 13
(8)
, 2152-2166. https://doi.org/10.1039/D1SC05506B
- Alexander H. van Asbeck, Jürgen Dieker, Rik Oude Egberink, Lennard van den Berg, Johan van der Vlag, Roland Brock. Protein Expression Correlates Linearly with mRNA Dose over Up to Five Orders of Magnitude In Vitro and In Vivo. Biomedicines 2021, 9
(5)
, 511. https://doi.org/10.3390/biomedicines9050511
- Sreejesh Sreedharan, Rajeshwari Tiwari, Deepak Tyde, Stephen O. Aderinto, Sumit Kumar Pramanik, Amitava Das, James A. Thomas. Nanocarriers used as probes for super-resolution microscopy. Materials Chemistry Frontiers 2021, 5
(3)
, 1268-1282. https://doi.org/10.1039/D0QM00684J
- Laura Woythe, Nicholas B. Tito, Lorenzo Albertazzi. A quantitative view on multivalent nanomedicine targeting. Advanced Drug Delivery Reviews 2021, 169 , 1-21. https://doi.org/10.1016/j.addr.2020.11.010
- Teodora Andrian, Roger Riera, Silvia Pujals, Lorenzo Albertazzi. Nanoscopy for endosomal escape quantification. Nanoscale Advances 2021, 3
(1)
, 10-23. https://doi.org/10.1039/D0NA00454E
- Teodora Andrian, Thomas Bakkum, Daphne M. van Elsland, Erik Bos, Abraham J. Koster, Lorenzo Albertazzi, Sander I. van Kasteren, Sílvia Pujals. Super-resolution correlative light-electron microscopy using a click-chemistry approach for studying intracellular trafficking. 2021, 303-331. https://doi.org/10.1016/bs.mcb.2020.09.001
- Jose Rodrigo Magana, Christian C. M. Sproncken, Ilja K. Voets. On Complex Coacervate Core Micelles: Structure-Function Perspectives. Polymers 2020, 12
(9)
, 1953. https://doi.org/10.3390/polym12091953
- Weiqi Zhang. Protein Corona of Nanoparticles and Its Application in Drug Delivery. 2020, 389-419. https://doi.org/10.1007/978-981-15-5386-8_9
- Kaido Kurrikoff, Ülo Langel. Recent CPP-based applications in medicine. Expert Opinion on Drug Delivery 2019, 16
(11)
, 1183-1191. https://doi.org/10.1080/17425247.2019.1665021
- Roger Riera, Natalia Feiner-Gracia, Cristina Fornaguera, Anna Cascante, Salvador Borrós, Lorenzo Albertazzi. Tracking the DNA complexation state of pBAE polyplexes in cells with super resolution microscopy. Nanoscale 2019, 11
(38)
, 17869-17877. https://doi.org/10.1039/C9NR02858G
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
Abstract
Figure 1
Figure 1. Imaging of polyplexes using dSTORM microscopy. (A) Schematic representation of polyplex formation from mRNA-Cy5 and AlexaFluor488-R9, with imaging and analysis. (B) Conventional fluorescent image of polyplexes at N/P 5 (red represents mRNA molecules and green R9 molecules). (C) dSTORM imaging of polyplexes, with the same field of view as in B. Scale bar 2 μm. (D) Close-up images of three different areas showing low-resolution data on the left and high-resolution data on the right. Scale bar 400 nm.
Figure 2
Figure 2. Structure and stoichiometry differences of polyplexes. (A) dSTORM images of polyplexes prepared at N/P ratios of 1, 3, 5, and 7 from left to right. (B) Frequency histograms of the size of the polyplexes quantified from the STROM images for each N/P ratio. (C) Box plot comparing the number of localizations of mRNA and peptide quantified for each N/P ratio together with the quantification of single molecules and the number of localizations obtained from the simulations. Statistical analyses were run (Mann–Whitney test), showing significant differences between the peptide content in NP3 and NP5 (p < 0.0001) and the mRNA content between NP1 and NP3 (p < 0.0001). Scale bar 200 nm.
Figure 3
Figure 3. Correlation between the number of mRNA localizations and R9 localizations. (A) Scatter plots of the number of mRNA localizations versus the number of R9 localizations, together with a color code representing the size for each polyplex. (B) Number of localizations plotted as a function of polyplex size.
Figure 4
Figure 4. Dependence of cellular nanoparticle uptake on the N/P ratio. HeLa cells were incubated for 1 h with polyplexes consisting of Cy5-mRNA and fluorescein-labeled R9 at the indicated N/P ratios, followed by washing and confocal microscopy of living cells. Labeled peptide was mixed with unlabeled peptide in a ratio of 1:10. The mRNA concentration was constant at 4.5 pM for all N/P ratios, corresponding to peptide concentrations of 0.5, 1.5, 2.5, and 3.5 μM for N/P 1, 3, 5 and 7, respectively. (A) Cellular uptake and intracellular distribution. To facilitate the discrimination of intensities, a false color look-up table has been employed. The scale bar corresponds to 20 μm. (B) Average pixel intensities for vesicular fluorescence. (C) Pixels per cell as a measure of the number and size of endocytic vesicles. (D) Average pixel intensity for fluorescence outside cells, recorded at a higher detector gain. (E) Ratio of peptide over mRNA fluorescence intensity. Ratios were calculated for the vesicular intensities of each analysis image and averaged. Error bars correspond to the standard deviation for normalized data of two independent experiments.
Figure 5
Figure 5. Stability of polyplexes in serum. (A) dSTORM image of the N/P 5 polyplexes initially and after being incubated with 20% FBS for 1 min and 1 h. Scale bar 200 nm. (B) Number of mRNA localizations of N/P 5 polyplexes after being incubated with 20% FBS for 1 min and 1 h. (C) Number of R9 localizations of N/P 5 polyplexes after being incubated with 20% FBS for 1 min and 1 h. (D) Schematic representation of the destabilization of the polyplexes in time in the presence of serum.
References
This article references 36 other publications.
- 1Koren, E.; Torchilin, V. P. Trends Mol. Med. 2012, 18 (7), 385– 393, DOI: 10.1016/j.molmed.2012.04.012There is no corresponding record for this reference.
- 2Collado Camps, E.; Brock, R. Bioorg. Med. Chem. 2018, 26 (10), 2780– 2787, DOI: 10.1016/j.bmc.2017.11.004There is no corresponding record for this reference.
- 3Heitz, F.; Morris May, C.; Divita, G. Br. J. Pharmacol. 2009, 157 (2), 195– 206, DOI: 10.1111/j.1476-5381.2009.00057.x3Twenty years of cell-penetrating peptides: from molecular mechanisms to therapeuticsHeitz, Frederic; Morris, May Catherine; Divita, GillesBritish Journal of Pharmacology (2009), 157 (2), 195-206CODEN: BJPCBM; ISSN:1476-5381. (Wiley-Blackwell)A review. The recent discovery of new potent therapeutic mols. that do not reach the clinic due to poor delivery and low bioavailability have made of delivery a key stone in therapeutic development. Several technologies have been designed to improve cellular uptake of therapeutic mols., including cell-penetrating peptides (CPPs). CPPs were first discovered based on the potency of several proteins to enter cells. Numerous CPPs have been described so far, which can be grouped into two major classes, the first requiring chem. linkage with the drug for cellular internalization and the second involving formation of stable, non-covalent complexes with drugs. Nowadays, CPPs constitute very promising tools for non-invasive cellular import of cargo and have been successfully applied for in vitro and in vivo delivery of therapeutic mols. varying from small chem. mol., nucleic acids, proteins, peptides, liposomes and particles. This review will focus on the structure/function and cellular uptake mechanism of CPPs in the general context of drug delivery. We will also highlight the application of peptide carriers for the delivery of therapeutic mols. and provide an update of their clin. evaluation.
- 4Lee, S. H.; Castagner, B.; Leroux, J.-C. Eur. J. Pharm. Biopharm. 2013, 85 (1), 5– 11, DOI: 10.1016/j.ejpb.2013.03.0214Is there a future for cell-penetrating peptides in oligonucleotide delivery?Lee, Soo Hyeon; Castagner, Bastien; Leroux, Jean-ChristopheEuropean Journal of Pharmaceutics and Biopharmaceutics (2013), 85 (1), 5-11CODEN: EJPBEL; ISSN:0939-6411. (Elsevier B.V.)A review. Cell-penetrating peptides have been widely investigated as delivery vehicles for oligonucleotides (e.g., siRNA and antisense oligonucleotides). Different delivery strategies can be used, such as co-incubation, direct conjugation, non-covalent complex, and modification on the surface of liposome or polymer complexes. However, several challenges remain for their preclin. and clin. development. Endosomal escape, lack of cell/tissue specificity, and toxicity are major concerns in the design of cell-penetrating peptide-mediated delivery systems. In this commentary, we highlight recent reports of cell-penetrating peptide incorporation into oligonucleotide delivery systems and underline the remaining challenges, particularly for preclin. and clin. applications.
- 5Zhang, P.; Wagner, E. Top. Curr. Chem. 2017, 375 (2), 26, DOI: 10.1007/s41061-017-0112-0There is no corresponding record for this reference.
- 6Lächelt, U.; Wagner, E. Chem. Rev. 2015, 115 (19), 11043– 11078, DOI: 10.1021/cr5006793There is no corresponding record for this reference.
- 7Scomparin, A.; Polyak, D.; Krivitsky, A.; Satchi-Fainaro, R. Biotechnol. Adv. 2015, 33 (6, Part 3), 1294– 1309, DOI: 10.1016/j.biotechadv.2015.04.008There is no corresponding record for this reference.
- 8Debus, H.; Baumhof, P.; Probst, J.; Kissel, T. J. Controlled Release 2010, 148 (3), 334– 343, DOI: 10.1016/j.jconrel.2010.09.007There is no corresponding record for this reference.
- 9Meng, Z.; O’Keeffe-Ahern, J.; Lyu, J.; Pierucci, L.; Zhou, D.; Wang, W. Biomater. Sci. 2017, 5 (12), 2381– 2392, DOI: 10.1039/C7BM00712D9A new developing class of gene delivery: messenger RNA-based therapeuticsMeng, Zhao; O'Keeffe-Ahern, Jonathan; Jing, Lyu; Pierucci, Luca; Zhou, Dezhong; Wang, WenxinBiomaterials Science (2017), 5 (12), 2381-2392CODEN: BSICCH; ISSN:2047-4849. (Royal Society of Chemistry)A review. Gene therapy has long been held as having the potential to become a front line treatment for various genetic disorders. However, the direct delivery of nucleic acids to correct a genetic disorder has numerous limitations owing to the inability of naked nucleic acids (DNA and RNA) to traverse the cell membrane. Recently, mRNA (mRNA) based delivery has become a more attractive alternative to DNA due to the relatively easier transfection process, higher efficiency and safety profile. As with all gene therapies, the central challenge that remains is the efficient delivery of nucleic acids intracellularly. This review presents the recent progress in mRNA delivery, focusing on comparing the advantages and limitations of non-viral based delivery vectors.
- 10Sahin, U.; Karikó, K.; Türeci, Ö. Nat. Rev. Drug Discovery 2014, 13 (10), 759– 780, DOI: 10.1038/nrd4278There is no corresponding record for this reference.
- 11van Asbeck, A. H.; Beyerle, A.; McNeill, H.; Bovee-Geurts, P. H. M.; Lindberg, S.; Verdurmen, W. P. R.; Hällbrink, M.; Langel, Ü.; Heidenreich, O.; Brock, R. ACS Nano 2013, 7 (5), 3797– 3807, DOI: 10.1021/nn305754cThere is no corresponding record for this reference.
- 12Lee, J. B.; Hong, J.; Bonner, D. K.; Poon, Z.; Hammond, P. T. Nat. Mater. 2012, 11 (4), 316– 322, DOI: 10.1038/nmat3253There is no corresponding record for this reference.
- 13Patil, M. L.; Zhang, M.; Taratula, O.; Garbuzenko, O. B.; He, H.; Minko, T. Biomacromolecules 2009, 10 (2), 258– 266, DOI: 10.1021/bm800997313Internally Cationic Polyamidoamine PAMAM-OH Dendrimers for siRNA Delivery: Effect of the Degree of Quaternization and Cancer TargetingPatil, Mahesh L.; Zhang, Min; Taratula, Oleh; Garbuzenko, Olga B.; He, Huixin; Minko, TamaraBiomacromolecules (2009), 10 (2), 258-266CODEN: BOMAF6; ISSN:1525-7797. (American Chemical Society)A novel cancer targeted, internally cationic and surface neutral polyamidoamine (PAMAM) dendrimer, was designed, synthesized, and evaluated as a nanocarrier for the targeted intracellular delivery of siRNA. The dendrimer contained a synthetic analog of LH-releasing hormone as cancer targeting moiety. The proposed delivery system possesses the following advantages: (1) internal cationic charges for complexation with siRNA and enhanced siRNA protection; (2) low cytotoxicity; (3) lesser degree of quaternization offering free tertiary amines for potential proton sponge effect; and (4) targeting specifically to cancer cells for enhancing siRNA uptake and efficiency and potential limitation of adverse side effects of chemotherapy on healthy organs. Both nontargeted and targeted dendrimer-siRNA complexes formed compact nanometer size spherical particles, exhibited very low cytotoxicity even at the higher concn., and efficiently penetrated cancer cells in vitro. However, only the targeted dendrimer-siRNA complex was able to substantially decrease the expression of a targeted BCL2 gene.
- 14Ofek, P.; Fischer, W.; Calderón, M.; Haag, R.; Satchi-Fainaro, R. FASEB J. 2010, 24 (9), 3122– 3134, DOI: 10.1096/fj.09-149641There is no corresponding record for this reference.
- 15García-Sosa, A. T.; Tulp, I.; Langel, K.; Langel, Ü. BioMed Res. Int. 2014, 2014, 257040, DOI: 10.1155/2014/257040There is no corresponding record for this reference.
- 16Meneksedag-Erol, D.; Tang, T.; Uludağ, H. Biomaterials 2014, 35 (25), 7068– 7076, DOI: 10.1016/j.biomaterials.2014.04.10316Molecular modeling of polynucleotide complexesMeneksedag-Erol, Deniz; Tang, Tian; Uludag, HasanBiomaterials (2014), 35 (25), 7068-7076CODEN: BIMADU; ISSN:0142-9612. (Elsevier Ltd.)A review. Delivery of polynucleotides into patient cells is a promising strategy for treatment of genetic disorders. Gene therapy aims to either synthesize desired proteins (DNA delivery) or suppress expression of endogenous genes (siRNA delivery). Carriers constitute an important part of gene therapeutics due to limitations arising from the pharmacokinetics of polynucleotides. Non-viral carriers such as polymers and lipids protect polynucleotides from intra and extracellular threats and facilitate formation of cell-permeable nanoparticles through shielding and/or bridging multiple polynucleotide mols. Formation of nanoparticulate systems with optimal features, their cellular uptake and intracellular trafficking are crucial steps for an effective gene therapy. Despite the great amt. of exptl. work pursued, crit. features of the nanoparticles as well as their processing mechanisms are still under debate due to the lack of instrumentation at at. resoln. Mol. modeling based computational approaches can shed light onto the at. level details of gene delivery systems, thus provide valuable input that cannot be readily obtained with exptl. techniques. Here, we review the mol. modeling research pursued on crit. gene therapy steps, highlight the knowledge gaps in the field and providing future perspectives. Existing modeling studies revealed several important aspects of gene delivery, such as nanoparticle formation dynamics with various carriers, effect of carrier properties on complexation, carrier conformations in endosomal stages, and release of polynucleotides from carriers. Rate-limiting steps related to cellular events (i.e. internalization, endosomal escape, and nuclear uptake) are now beginning to be addressed by computational approaches. Limitations arising from current computational power and accuracy of modeling have been hindering the development of more realistic models. With the help of rapidly-growing computational power, the crit. aspects of gene therapy are expected to be better investigated and direct comparison between more realistic mol. modeling and expts. may open the path for design of next generation gene therapeutics.
- 17Majzoub, R. N.; Ewert, K. K.; Safinya, C. R. Philos. Trans. R. Soc., A 2016, 374 (2072), 20150129, DOI: 10.1098/rsta.2015.012917Cationic liposome-nucleic acid nanoparticle assemblies with applications in gene delivery and gene silencingMajzoub, Ramsey N.; Ewert, Kai K.; Safinya, Cyrus R.Philosophical Transactions of the Royal Society, A: Mathematical, Physical & Engineering Sciences (2016), 374 (2072), 20150129/1-20150129/14CODEN: PTRMAD; ISSN:1364-503X. (Royal Society)Cationic liposomes (CLs) are synthetic carriers of nucleic acids in gene delivery and gene silencing therapeutics. The introduction will describe the structures of distinct liq. cryst. phases of CL-nucleic acid complexes, which were revealed in earlier synchrotron small-angle X-ray scattering expts. When mixed with plasmid DNA, CLs contg. lipids with distinct shapes spontaneously undergo topol. transitions into self-assembled lamellar, inverse hexagonal, and hexagonal CL-DNA phases. CLs contg. cubic phase lipids are obsd. to readily mix with short interfering RNA (siRNA) mols. creating double gyroid CL-siRNA phases for gene silencing. Custom synthesis of multivalent lipids and a range of novel polyethylene glycol (PEG)-lipids with attached targeting ligands and hydrolysable moieties have led to functionalized equil. nanoparticles (NPs) optimized for cell targeting, uptake or endosomal escape. Very recent expts. are described with surface-functionalized PEGylated CL-DNA NPs, including fluorescence microscopy colocalization with members of the Rab family of GTPases, which directly reveal interactions with cell membranes and NP pathways. In vitro optimization of CL-DNA and CL-siRNA NPs with relevant primary cancer cells is expected to impact nucleic acid therapeutics in vivo.
- 18van der Zwaag, D.; Vanparijs, N.; Wijnands, S.; De Rycke, R.; De Geest, B. G.; Albertazzi, L. ACS Appl. Mater. Interfaces 2016, 8 (10), 6391– 6399, DOI: 10.1021/acsami.6b00811There is no corresponding record for this reference.
- 19Duro-Castano, A.; Nebot Vicent, J.; Niño-Pariente, A.; Armiñán, A.; Arroyo-Crespo, J.; Paul, A.; Feiner-Gracia, N.; Albertazzi, L.; Vicent, M. Adv. Mater. 2017, 29 (39), 1702888, DOI: 10.1002/adma.201702888There is no corresponding record for this reference.
- 20Ardizzone, A.; Kurhuzenkau, S.; Illa-Tuset, S.; Faraudo, J.; Bondar, M.; Hagan, D.; Van Stryland, E. W.; Painelli, A.; Sissa, C.; Feiner, N. Small 2018, 14, 1703851, DOI: 10.1002/smll.201703851There is no corresponding record for this reference.
- 21Burgert, A.; Schlegel, J.; Bécam, J.; Doose, S.; Bieberich, E.; Schubert-Unkmeir, A.; Sauer, M. Angew. Chem. 2017, 129 (22), 6227– 6231, DOI: 10.1002/ange.201700570There is no corresponding record for this reference.
- 22Letschert, S.; Göhler, A.; Franke, C.; Bertleff-Zieschang, N.; Memmel, E.; Doose, S.; Seibel, J.; Sauer, M. Angew. Chem., Int. Ed. 2014, 53 (41), 10921– 10924, DOI: 10.1002/anie.201406045There is no corresponding record for this reference.
- 23Fricke, F.; Beaudouin, J.; Eils, R.; Heilemann, M. Sci. Rep. 2015, 5, 14072, DOI: 10.1038/srep1407223One, two or three? Probing the stoichiometry of membrane proteins by single-molecule localization microscopyFricke Franziska; Heilemann Mike; Beaudouin Joel; Eils RolandScientific reports (2015), 5 (), 14072 ISSN:.Probing the oligomeric state of abundant molecules, such as membrane proteins in intact cells, is essential, but has not been straightforward. We address this challenge with a simple counting strategy that is capable of reporting the oligomeric state of dense, membrane-bound protein complexes. It is based on single-molecule localization microscopy to super-resolve protein structures in intact cells and basic quantitative evaluation. We validate our method with membrane-bound monomeric CD86 and dimeric cytotoxic T-lymphocyte-associated protein as model proteins and confirm their oligomeric states. We further detect oligomerization of CD80 and vesicular stomatitis virus glycoprotein and propose coexistence of monomers and dimers for CD80 and trimeric assembly of the viral protein at the cell membrane. This approach should prove valuable for researchers striving for reliable molecular counting in cells.
- 24Zanacchi, F. C.; Manzo, C.; Alvarez, A. S.; Derr, N. D.; Garcia-Parajo, M. F.; Lakadamyali, M. Nat. Methods 2017, 14 (8), 789– 792, DOI: 10.1038/nmeth.434224A DNA origami platform for quantifying protein copy number in super-resolutionZanacchi, Francesca Cella; Manzo, Carlo; Alvarez, Angel S.; Derr, Nathan D.; Garcia-Parajo, Maria F.; Lakadamyali, MelikeNature Methods (2017), 14 (8), 789-792CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)Single-mol.-based super-resoln. microscopy offers researchers a unique opportunity to quantify protein copy no. with nanoscale resoln. However, while fluorescent proteins have been characterized for quant. imaging using calibration stds., similar calibration tools for immunofluorescence with small org. fluorophores are lacking. Here we show that DNA origami, in combination with GFP antibodies, is a versatile platform for calibrating fluorophore and antibody labeling efficiency to quantify protein copy no. in cellular contexts using super-resoln. microscopy.
- 25Puchner, E. M.; Walter, J. M.; Kasper, R.; Huang, B.; Lim, W. A. Proc. Natl. Acad. Sci. U. S. A. 2013, 110 (40), 16015– 16020, DOI: 10.1073/pnas.1309676110There is no corresponding record for this reference.
- 26Ezzat, K.; EL Andaloussi, S.; Zaghloul, E. M.; Lehto, T.; Lindberg, S.; Moreno, P. M. D.; Viola, J. R.; Magdy, T.; Abdo, R.; Guterstam, P. Nucleic Acids Res. 2011, 39 (12), 5284– 5298, DOI: 10.1093/nar/gkr072There is no corresponding record for this reference.
- 27Wu, R. P.; Youngblood, D. S.; Hassinger, J. N.; Lovejoy, C. E.; Nelson, M. H.; Iversen, P. L.; Moulton, H. M. Nucleic Acids Res. 2007, 35 (15), 5182– 5191, DOI: 10.1093/nar/gkm478There is no corresponding record for this reference.
- 28Futaki, S.; Ohashi, W.; Suzuki, T.; Niwa, M.; Tanaka, S.; Ueda, K.; Harashima, H.; Sugiura, Y. Bioconjugate Chem. 2001, 12 (6), 1005– 1011, DOI: 10.1021/bc015508l28Stearylated Arginine-Rich Peptides: A New Class of Transfection SystemsFutaki, Shiroh; Ohashi, Wakana; Suzuki, Tomoki; Niwa, Miki; Tanaka, Seigo; Ueda, Kunihiro; Harashima, Hideyoshi; Sugiura, YukioBioconjugate Chemistry (2001), 12 (6), 1005-1011CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)Membrane-permeable arginine-rich peptides, such as HIV-1 Tat-(48-60), HIV-1 Rev-(34-50), and flock house virus (FHV) coat-(35-49), have been shown to possess the ability to transfect COS-7 cells with luciferase-coding plasmid as efficiently as polyarginine (MW 5000-15 000) and polylysine (MW 9800). Not only these virus-derived cationic peptides but also oligoarginines of 4-16 residues were found to be able to transfect cells. In the case of the Tat, FHV, and octaarginine peptides, N-terminal stearylation of the peptides increases the transfection efficiency by approx. 100 times to reach the same order of magnitude as that of LipofectAMINE, one of the most efficient com. available transfection agents. Also, a certain correlation was obsd. between the transfection efficiency of stearyl-(Arg)n peptides (stearyl-Rn: n = 4, 8, 12, 16) and the membrane permeability of the corresponding (Arg)n peptides (Rn).
- 29Vaidyanathan, S.; Chen, J.; Orr, B. G.; Banaszak Holl, M. M. Mol. Pharmaceutics 2016, 13 (6), 1967– 1978, DOI: 10.1021/acs.molpharmaceut.6b0013929Cationic Polymer Intercalation into the Lipid Membrane Enables Intact Polyplex DNA Escape from Endosomes for Gene DeliveryVaidyanathan, Sriram; Chen, Junjie; Orr, Bradford G.; Banaszak Holl, Mark M.Molecular Pharmaceutics (2016), 13 (6), 1967-1978CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)Developing improved cationic polymer-DNA polyplexes for gene delivery requires improved understanding of DNA transport from endosomes into the nucleus. Using a FRET-capable oligonucleotide mol. beacon (OMB), we monitored the transport of intact DNA to cell organelles. We obsd. that for effective (jetPEI) and ineffective (G5 PAMAM) vectors, the fraction of cells displaying intact OMB in the cytosol (jetPEI » G5 PAMAM) quant. predicted the fraction expressing transgene (jetPEI » G5 PAMAM). Intact OMB delivered with PAMAM and confined to endosomes could be released to the cytosol by the subsequent addn. of L-PEI, with a corresponding 10-fold increase in transgene expression. These results suggest that future vector development should optimize vectors for intercalation into, and destabilization of, the endosomal membrane. Finally, the study highlights a two-step strategy in which the pDNA is loaded in cells using one vector and endosomal release is mediated by a second agent.
- 30Lehto, T.; Vasconcelos, L.; Margus, H.; Figueroa, R.; Pooga, M.; Hällbrink, M.; Langel, Ü. Bioconjugate Chem. 2017, 28 (3), 782– 792, DOI: 10.1021/acs.bioconjchem.6b0068030Saturated Fatty Acid Analogues of Cell-Penetrating Peptide PepFect14: Role of Fatty Acid Modification in Complexation and Delivery of Splice-Correcting OligonucleotidesLehto, Tonis; Vasconcelos, Luis; Margus, Helerin; Figueroa, Ricardo; Pooga, Margus; Haellbrink, Mattias; Langel, UloBioconjugate Chemistry (2017), 28 (3), 782-792CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)Modifying cell-penetrating peptides (CPPs) with fatty acids has long been used to improve peptide-mediated nucleic acid delivery. In this study we have revisited this phenomenon with a systematic approach where we developed a structure-activity relationship to describe the role of the acyl chain length in the transfection process. For that we took a well-studied CPP, PepFect14, as the basis and varied its N-terminal acyl chain length from 2 to 22 carbons. To evaluate the delivery efficiency, the peptides were noncovalently complexed with a splice-correcting oligonucleotide (SCO) and tested in HeLa pLuc705 reporter cell line. Our results demonstrate that biol. splice-correction activity emerges from acyl chain of 12 carbons and increases linearly with each addnl. carbon. To assess the underlying factors regarding how the transfection efficacy of these complexes is dependent on hydrophobicity, we used an array of different methods. For the functionally active peptides (C12-22) there was no apparent difference in their physicochem. properties, including complex formation efficiency, hydrodynamic size, and zeta potential. Moreover, membrane activity studies with peptides and their complexes with SCOs confirmed that the toxicity of the complexes at higher molar ratios is mainly caused by the free fraction of the peptide which is not incorporated into the peptide/oligonucleotide complexes. Finally, we show that the increase in splice-correcting activity correlates with the ability of the complexes to assoc. with the cells. Collectively these studies lay the ground work for how to design highly efficient CPPs and how to optimize their oligonucleotide complexes for lowest toxicity without losing efficiency.
- 31Jekhmane, S.; de Haas, R.; Paulino da Silva Filho, O.; van Asbeck, A. H.; Favretto, M. E.; Hernandez Garcia, A.; Brock, R.; de Vries, R. Nucleic Acid Ther. 2017, 27 (3), 159– 167, DOI: 10.1089/nat.2016.066031Virus-Like Particles of mRNA with Artificial Minimal Coat Proteins: Particle Formation, Stability, and Transfection EfficiencyJekhmane, Shehrazade; de Haas, Rob; Paulino da Silva Filho, Omar; van Asbeck, Alexander H.; Favretto, Marco Emanuele; Hernandez Garcia, Armando; Brock, Roland; de Vries, RenkoNucleic Acid Therapeutics (2017), 27 (3), 159-167CODEN: NATUCU; ISSN:2159-3337. (Mary Ann Liebert, Inc.)RNA has enormous potential as a therapeutic, yet, the successful application depends on efficient delivery strategies. In this study, we demonstrate that a designed artificial viral coat protein, which self-assembles with DNA to form rod-shaped virus-like particles (VLPs), also encapsulates and protects mRNA encoding enhanced green fluorescent protein (EGFP) and luciferase, and yields cellular expression of these proteins. The artificial viral coat protein consists of an oligolysine (K12) for binding to the oligonucleotide, a silk protein-like midblock S10 = (GAGAGAGQ)10 that self-assembles into stiff rods, and a long hydrophilic random coil block C that shields the nucleic acid cargo from its environment. With mRNA, the C-S10-K12 protein coassembles to form rod-shaped VLPs each encapsulating about one to five mRNA mols. Inside the rod-shaped VLPs, the mRNAs are protected against degrdn. by RNAses, and VLPs also maintain their shape following incubation with serum. Despite the lack of cationic surface charge, the mRNA VLPs transfect cells with both EGFP and luciferase, although with a much lower efficiency than obtained by a lipoplex transfection reagent. The VLPs have a negligible toxicity and minimal hemolytic activity. Our results demonstrate that VLPs yield efficient packaging and shielding of mRNA and create the basis for implementation of addnl. virus-like functionalities to improve transfection and cell specificity, such as targeting functionalities.
- 32Zhuang, X. Nat. Photonics 2009, 3 (7), 365– 367, DOI: 10.1038/nphoton.2009.10132Nano-imaging with STORMZhuang, XiaoweiNature Photonics (2009), 3 (7), 365-367CODEN: NPAHBY; ISSN:1749-4885. (Nature Publishing Group)Multicolor, three-dimensional stochastic optical reconstruction microscopy now makes it possible to image cellular structures with near mol.-scale resoln.
- 33Coltharp, C.; Yang, X.; Xiao, J. Curr. Opin. Struct. Biol. 2014, 28, 112– 121, DOI: 10.1016/j.sbi.2014.08.00833Quantitative analysis of single-molecule superresolution imagesColtharp, Carla; Yang, Xinxing; Xiao, JieCurrent Opinion in Structural Biology (2014), 28 (), 112-121CODEN: COSBEF; ISSN:0959-440X. (Elsevier Ltd.)This review highlights the quant. capabilities of single-mol. localization-based superresoln. imaging methods. In addn. to revealing fine structural details, the mol. coordinate lists generated by these methods provide the crit. ability to quantify the no., clustering, and colocalization of mols. with 10-50 nm resoln. Here we describe typical workflows and precautions for quant. anal. of single-mol. superresoln. images. These guidelines include potential pitfalls and essential control expts., allowing crit. assessment and interpretation of superresoln. images.
- 34Feiner-Gracia, N.; Beck, M.; Pujals, S.; Tosi, S.; Mandal, T.; Buske, C.; Linden, M.; Albertazzi, L. Small 2017, 13 (41), 1701631, DOI: 10.1002/smll.201701631There is no corresponding record for this reference.
- 35Albertazzi, L.; van der Zwaag, D.; Leenders, C. M. A.; Fitzner, R.; van der Hofstad, R. W.; Meijer, E. W. Science 2014, 344 (6183), 491– 495, DOI: 10.1126/science.125094535Probing Exchange Pathways in One-Dimensional Aggregates with Super-Resolution MicroscopyAlbertazzi, Lorenzo; van der Zwaag, Daan; Leenders, Christianus M. A.; Fitzner, Robert; van der Hofstad, Remco W.; Meijer, E. W.Science (Washington, DC, United States) (2014), 344 (6183), 491-495CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Supramol. fibers are prominent structures in biol. and chem. A quant. understanding of mol. exchange pathways in these one-dimensional aggregates was obtained by a combination of super-resoln. stochastic optical reconstruction microscopy and stochastic simulation. The potential of this methodol. is demonstrated with a set of well-defined synthetic building blocks that self-assemble into supramol. fibrils. Previous ensemble measurements hid all mol. phenomena underpinning monomer exchange, but the mol. pathway detd. from single-aggregate studies revealed unexpected homogeneous exchange along the polymer backbone. These results pave the way for exptl. investigation of the structure and exchange pathways of synthetic and natural supramol. fibers.
- 36Wallbrecher, R.; Ackels, T.; Olea, R. A.; Klein, M. J.; Caillon, L.; Schiller, J.; Bovée-Geurts, P. H.; van Kuppevelt, T. H.; Ulrich, A. S.; Spehr, M. J. Controlled Release 2017, 256, 68– 78, DOI: 10.1016/j.jconrel.2017.04.013There is no corresponding record for this reference.
Supporting Information
Supporting Information
The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acs.nanolett.8b04407.
Materials and methods, a detailed explanation of the simulations used, and additional experimental results (PDF)
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.