• No results found

Cover Page The handle http://hdl.handle.net/1887/44712

N/A
N/A
Protected

Academic year: 2021

Share "Cover Page The handle http://hdl.handle.net/1887/44712"

Copied!
154
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

The handle http://hdl.handle.net/1887/44712 holds various files of this Leiden University dissertation

Author: Jian Yang

Title: In vitro and in vivo delivery of functionalized nanoparticles via coiled-coil interactions

Issue Date: 2016-12-01

(2)
(3)
(4)

In vitro and in vivo delivery of functionalized nanoparticles

via coiled-coil interactions

JIAN YANG

(5)

Thesis layout by Jian Yang

Printing by GVO drukkers &vormgevers B.V. Janneke & Ineke, www.gvo.nl ISBN/EAN:

Copyright @ 2016, Jian Yang, Leiden

No part of this book may be reproduced, digitalized, or transmitted in any form or by any means, electronic or mechanical, including photocopying, recording, or by any information storage of retrieval system, without prior permission from the author.

(6)

In vitro and in vivo delivery of functionalized nanoparticles

via coiled-coil interactions

Proefschrift

ter verkrijging van

de graad van Doctor aan de Universiteit Leiden, Op gezag van Rector Magnificus Prof. mr. C.J.J.M. Stolker,

volgens het besluit van het College voor Promoties te verdedigen op donderdag 1 december 2016

Klokke 15.00 uur door Jian Yang

Geboren op 20 September te Huangshi, P.R.China

(7)

Promotor: Prof.dr. A. Kros

Co-promotor: Dr. René. C.L. Olsthoorn

Thesis Commissie:

Prof.dr.J.Brouwer (VZ)

Prof. dr. M.H.M. Noteborn (seretaris) Prof. dr. Bart Jan Ravoo1

Prof. dr. Sijbren Otto2 Dr. Schneider Gregory

1 Organisch-Chemisches Institut, Westfälische Wilhelms-Universität Münster, Germany 2 Centre for Systems Chemistry, University of Groningen, the Netherlands

The studies described in this thesis were primarily performed at the Leiden University (Netherlands), and in part at the Zernike Institute for Advanced Materials, University of Groningen (Netherlands).

This work was financially supported as indicated in each chapter.

(8)

but- I hope- into a better shape”

Charles Dickens (February 7, 1812 – June 9, 1870)

(9)
(10)

Outline of the thesis

Introduction

Drug Delivery via Cell Membrane Fusion using Lipopeptide Modified Liposomes

Application of Coiled Coil Peptides in Liposomal Anti-Cancer Drug Delivery using Zebrafish Xenograft Model

Coiled-coil forming peptides enhance intracellular delivery of lipid bilayer coated mesoporous silica nanoparticles Lipid-DNA induced fusion of phospholipid vesicles Summary and perspective

Nederlandse samenvatting Chapter 1

Chapter 2

Chapter 3

Chapter 4

Chapter 5 Chapter 6

11 23

55

81

107 137142

145 147 150 151 Appendices

Author affiliations List of publications Curriculum Vitae Acknowledgments

(11)
(12)

11 Introduction: Drug Delivery via cell membrane

1

1 Chapter 1 Introduction Drug Delivery via Cell Membrane

Jian Yang, René C.L. Olsthoorn and Alexander Kros

(13)

The eukaryotic cell membrane protects the cell from its environment, but at the same time it facilitates the exchange of the nutritional ingredients and signals, keeping the cell’s inner environment in balance.1-2 One way for a cell to take up extracellular material, like proteins or small particles, is by endocytosis. During this process, part of the cell membrane including receptor-bound ligands or particles is internalized and forms membranous compartments called endosomes. These endosomes eventually fuse with lysosomes and their content is largely degraded, resulting in very low delivery efficiency of the content.

Nanocarriers (defined sizes of diameter as <1000 nm) are currently being widely studied as drug delivery tools in biomedical and clinical therapeutic fields.3,4 Because of their sizes, they can deliver drugs to otherwise inaccessible sites around the body. The types of nanocarriers that exist are diverse and consisting of polymers,5-8 liposomes,9-10 silica,11-12 metal nanoparticles,13-15 microfluidic devices16-17 and dendrimers18-19. Due to their chemical properties and physical structures, like size, shape, molecular weight, surface charge etc., there are five different common endocytic uptake mechanisms, that is, phagocytosis, macropinocytosis, clathrin-, caveolin-mediated and clathrin/caveolin-independent endocytosis. For example, Figure 1 shows that the size of engulfed particles affects their uptake routes.20 Giant particles (~1 µm) are typically taken up by cells through phagocytosis or macropinocytosis, following phagocytic vesicle directly fuse with lysosome instead of early endosome; When the size of particles is less than 200 nm, the dominant mechanisms are clathrin-mediated endocytosis or caveolin mediated endocytosis, which are based on receptor mediated endocytosis. Nanoparticles of ~90 nm are normally taken up by clathrin- and caveolin independent pathways.20 When size of nanoparticles changes, the major cellular uptake mechanism of nanoparticles also can be shifted.21

The surface charge also affects the route of cellular uptake of nanoparticles.22-24 Positively charged nanoparticles seem to undergo a higher extent of internalization, apparently as a result of the ionic interactions occurring between positively charged particles and negatively charged cell membranes.22, 25 Moreover, positively charged nanoparticles seem to be able to escape from lysosomes after being internalized and exhibit perinuclear localization, whereas negatively and neutrally charged nanoparticles remain localized within lysosomes.26 In general, the present findings establish that particle diameters of 0.5 μm and below were optimal for cell uptake;

Moreover, uptake of larger particles could be greatly enhanced by rendering the particle surface positive.22

(14)

1

Figure 1. Modes of cellular internalization of nanoparticles and respective size limitations.

Internalization of large particles is facilitated by phagocytosis (a). Nonspecific internal- ization of smaller particles (>1 μm) can occur through macropinocytosis (b). Smaller nanoparticles can be internalized through several pathways, including caveolar-mediated endocytosis (c), clathrin-mediated endocytosis (d) and clathrin-independent and caveolin independent endocytosis (e), with each being subject to slightly different size constraints.

Nanoparticles are represented by blue circles (> 1 μm), blue stars (about 120 nm), red stars (about 90 nm) and yellow rods (about 60 nm).20

In order to control the fate of a nanocarrier upon contact with a cell, a wide variety of chemical methods has been developed to produce functionalized nanoparticles while maintaining their biocompatibility and degradability. For example, uptake of positively charged poly (L-lysine) (PLL) coated nanoparticles normally does not depend on receptor mediated endocytosis, but when PLL is covalently grafted with PEG resulting in PLL-g-PEG, the uptake pathway has be switched to clathrin- mediated receptor endocytosis due to the changed surface. Moreover, it lowers the

(15)

cytotoxicity of PLL thereby enhancing its biocompatibility,.26

However, during the endocytic processes, endosomal excretion and lysosomal degradation of nanoparticles and their cargo can result in a very low delivery efficiency. For example, during the intravenous delivery of siRNA mediated by lipid nanoparticles, siRNA enters target cells by endocytosis, resulting in the release of only 1-2% of functional siRNA from endosomes.28 Therefore, there is a high need to develop new alternative delivery approaches that enhance the delivery efficiency.

So far, researchers have attempted to enhance endosomal escape and minimize the risk of degradation by employing fusogenic ligands. For example, fusogenic lipids, such as dioleoylphosphatidyl-ethanolamine (DOPE), when inserted into a lipoplex have been shown to promote endosomal release by enhancing the interaction between liposomal and endosomal membranes.29 Nanoparticles containing dimethylaminoethyl methacrylate (DMAEMA), propylacrylic acid (PAA) and hydrophobic butyl methacrylate (BMA) enable endosomal escape by a hydrophilic-to-hydrophobic transition at endosomal pH thereby mediating endosomal membrane disruption. This results in, for example, enhanced intracellular siRNA delivery and knockdown of GAPDH (glyceraldehyde-3-phosphate dehydrogenase) in HeLa cells.30 In addition, the positively charged Tat peptide, a so-called cell penetrating peptide (CPP) can lead to energy-independent cytoplasmic delivery of cargo;31 however Tat has high toxicity. Moreover, the current view for the cellular uptake pathway of CPP-coupled cargo is that they are predominantly internalized via endocytosis.32-34

Compared to these endocytic pathways, delivery systems based on a direct cytosolic delivery mechanism showed more advantages, for example, direct cytosolic delivery would include reduced content degradation and lowered immunogenicity of the payload and nanocarrier.35-38 In Figure 2, during the direct cytosolic delivery process, membrane fusion occurs. The lipid bilayer of nanoparticles tend to fuse with the leaflets from the cell plasma membrane bilayer, and after the formation of an aqueous bridge, the internal contents of the nanoparticles and cell plasma can mix. In many biological processes membrane fusion is also involved, e.g. transport of lipids,39 export of proteins,40 and even the entry of pathogens.41

(16)

1

Figure 2. Distinguishing features between the endocytosis pathway and direct membrane fusion.

Recently, the high efficacy of content release into the cytosol mediated by direct cytosolic delivery has been exploited by several groups. For example, Rotello et al. reported a system based on stabilized gold nanocapsules for direct delivery of an siRNA and protein.35,38 These nanoparticle-stabilized nanocapsules (NPSC) were cationic arginine-functionalized and formed a complex with siRNA. After delivery of the siRNA/NPSC complex in HEK293 cells expressing destabilized green fluorescence protein (deGFP), 90% knockdown of deGFP expression was observed. Evidence for direct cytosolic uptake came from imaging experiments using lysoTracker and from the effect of removing cholesterol from the outer membrane by nystatin, which did not affect the endocytic pathway but did inhibit uptake of siRNA complexes (Figure 3).

Figure 3. Nanoparticle-stabilized nanocapsules (NPSC) /siRNA components and schematic pre- sentation of NPSC mediated cytosolic siRNA delivery.

(17)

This thesis presents another approach for direct cytosolic delivery via membrane fusion. This approach is based on a complementary pair of coiled-coil forming peptides, K (KIAALKE)4 and E (EIAALEK)4 and is mimicking the action of the SNARE-complex. The SNARE-complex is responsible for fusion between vesicles and membranes of cells in the synapse and is also composed of coiled-coil forming peptides.

In our system, the peptides are conjugated to a cholesterol anchor via a polyethylene glycol (PEG) spacer, yielding lipopeptides CP4K4 and CP4E4. Membrane fusion between liposomes and the plasma membrane of the cells is triggered by these two lipopeptides when each embedded within the lipid bilayer of the liposomes or the plasma membrane of the cell. (Figure 4)

Figure 4. Model of direct cytosolic delivery. Cartoon showing the functionalization of a cell by CP4K4 and subsequent docking of CP4E4-decorated liposomes containing a fluorescent dye. After membrane fusion the fluorescent dye enters the cell.

Chapter 2 describes how this delivery system can be used for the delivery of fluorescent dyes and the anticancer drug doxorubicin (DOX) to HeLa cells. A wide spectrum of endocytosis inhibitors and endosome trackers are used to demonstrate that the uptake mechanism is most likely via membrane fusion.

Chapter 3 discusses how the E4/K4 coiled-coil system is adapted for in vivo delivery.

Here, K peptides are genetically expressed at the plasma membrane of HeLa cells (HeLa-K) as part of a membrane protein. In vitro HeLa-K cells but not normal HeLa cells can be targeted with CP4E4-liposomes containing a fluorescent dye. Next these HeLa-K cells are injected into zebrafish embryos and challenged with DOX loaded liposomes presenting CPE4 peptides. The specific targeting of these HeLa-K cells is demonstrated by fluorescence microscopy and by observing reduced cancer cell proliferation.

(18)

1

Chapter 4 describes the application of this coiled-coil formation to lipid-coated mesoporous silica nanoparticles (MSNs). Normally lipid-coated MSNs are taken up by endocytosis, but by employing the CPK/CPE coiled-coil system, their uptake route can be shifted to direct cytosolic delivery. This is demonstrated by the delivery and release of functional cytochrome c leading to an apoptotic response in the cell and also by testing the effect of endocytosis inhibitors.

In Chapter 5, a novel delivery system based on a pair of complementary DNA oligonucleotides is introduced. To embed these oligonucleotides into the mem- brane of liposomes four 5-(dode-1-cynyl) modified deoxyuracil nucleotides are incorporated. Compared to previous cholesterol-modifed DNA these modifications result in higher membrane fusion efficiencies,

Finally, the main results are summarized in Chapter 6, which also discusses some potential modifications to the coiled-coil mediated delivery system for future clinical applications.

(19)

References

1. Janeway, C. A., Jr.; Bottomly, K.; Horowitz, J.; Kaye, J.; Jones, B.; Tite, J., Modes of cell:cell communication in the immune system. J. Immunol. 1985, 135, 739s-742s.

2. Loewenstein, W. R., Cellular communication through membrane junctions. Special consider- ation of wound healing and cancer. Arch. Intern. Med. 1972, 129, 299-305.

3. Nel, A. E.; Madler, L.; Velegol, D.; Xia, T.; Hoek, E. M. V.; Somasundaran, P.; Klaessig, F.;

Castranova, V.; Thompson, M., Understanding biophysicochemical interactions at the nano-bio inter- face. Nat. Mater. 2009, 8, 543-557.

4. Vasir, J. K.; Labhasetwar, V., Biodegradable nanoparticles for cytosolic delivery of therapeu- tics. Adv. Drug Deliv. Rev. 2007, 59, 718-728.

5. Priya James, H.; John, R.; Alex, A.; Anoop, K. R., Smart polymers for the controlled delivery of drugs - a concise overview. Acta. Pharm. 2014, 4, 120-127.

6. Pack, D. W.; Hoffman, A. S.; Pun, S.; Stayton, P. S., Design and development of polymers for gene delivery. Nat. Rev. Drug Discov. 2005, 4, 581-593.

7. Teimouri, M.; Nia, A. H.; Abnous, K.; Eshghi, H.; Ramezani, M., Graphene oxide-cationic polymer conjugates: Synthesis and application as gene delivery vectors. Plasmid 2016, 84-85, 51-60.

8. Xu, R.; Zhang, G.; Mai, J.; Deng, X.; Segura-Ibarra, V.; Wu, S.; Shen, J.; Liu, H.; Hu, Z.;

Chen, L.; Huang, Y.; Koay, E.; Huang, Y.; Liu, J.; Ensor, J. E.; Blanco, E.; Liu, X.; Ferrari, M.; Shen, H., An injectable nanoparticle generator enhances delivery of cancer therapeutics. Nat. Biotechnol. 2016, 34, 414-418.

9. Pattni, B. S.; Chupin, V. V.; Torchilin, V. P., New developments in liposomal drug delivery.

Chem. Rev. 2015, 115, 10938-10966.

10. Chatin, B.; Mevel, M.; Devalliere, J.; Dallet, L.; Haudebourg, T.; Peuziat, P.; Colombani, T.; Berchel, M.; Lambert, O.; Edelman, A.; Pitard, B., Liposome-based Formulation for Intracellular Delivery of Functional Proteins. Mol. Ther. Nucleic Acids 2015, 4, e244.

11. Meng, H.; Wang, M.; Liu, H.; Liu, X.; Situ, A.; Wu, B.; Ji, Z.; Chang, C. H.; Nel, A. E., Cor- rection to use of a lipid-coated mesoporous silica nanoparticle platform for synergistic gemcitabine and paclitaxel delivery to human pancreatic cancer in mice. ACS Nano 2016, 10, 6416.

12. Niedermayer, S.; Weiss, V.; Herrmann, A.; Schmidt, A.; Datz, S.; Muller, K.; Wagner, E.;

Bein, T.; Brauchle, C., Multifunctional polymer-capped mesoporous silica nanoparticles for pH-re- sponsive targeted drug delivery. Nanoscale 2015, 7, 7953-7964.

13. Shan, Y.; Luo, T.; Peng, C.; Sheng, R.; Cao, A.; Cao, X.; Shen, M.; Guo, R.; Tomas, H.;

Shi, X., Gene delivery using dendrimer-entrapped gold nanoparticles as nonviral vectors. Biomaterials 2012, 33, 3025-3035.

(20)

1

14. Park, H.; Tsutsumi, H.; Mihara, H., Cell-selective intracellular drug delivery using doxoru- bicin and alpha-helical peptides conjugated to gold nanoparticles. Biomaterials 2014, 35, 3480-3487.

15. Kasyanenko, N.; Bakulev, V.; Perevyazko, I.; Nekrasova, T.; Nazarova, O.; Slita, A.; Zoloto- va, Y.; Panarin, E., Model system for multifunctional delivery nanoplatforms based on DNA-Polymer complexes containing silver nanoparticles and fluorescent dye. J. Biotechnol 2016, 236, 78-87.

16. Hasani-Sadrabadi, M. M.; Taranejoo, S.; Dashtimoghadam, E.; Bahlakeh, G.; Majedi, F. S.;

VanDersarl, J. J.; Janmaleki, M.; Sharifi, F.; Bertsch, A.; Hourigan, K.; Tayebi, L.; Renaud, P.; Jacob, K.

I., Microfluidic manipulation of core/shell nanoparticles for oral delivery of chemotherapeutics: a new treatment approach for colorectal cancer. Adv. Mater. 2016, 28, 4134-4141.

17. Koh, C. G.; Zhang, X.; Liu, S.; Golan, S.; Yu, B.; Yang, X.; Guan, J.; Jin, Y.; Talmon, Y.;

Muthusamy, N.; Chan, K. K.; Byrd, J. C.; Lee, R. J.; Marcucci, G.; Lee, L. J., Delivery of antisense oligodeoxyribonucleotide lipopolyplex nanoparticles assembled by microfluidic hydrodynamic focusing. J. Controlled Release 2010, 141, 62-69.

18. Kulhari, H.; Pooja, D.; Shrivastava, S.; Kuncha, M.; Naidu, V. G.; Bansal, V.; Sistla, R.; Ad- ams, D. J., Trastuzumab-grafted PAMAM dendrimers for the selective delivery of anticancer drugs to HER2-positive breast cancer. Sci. Rep. 2016, 6, 23179.

19. Luo, K.; Li, C.; Li, L.; She, W.; Wang, G.; Gu, Z., Arginine functionalized peptide dendrimers as potential gene delivery vehicles. Biomaterials 2012, 33, 4917-4927.

20. Petros, R. A.; DeSimone, J. M., Strategies in the design of nanoparticles for therapeutic ap- plications. Nat. Rev. Drug Discov. 2010, 9, 615-627.

21. Rejman, J.; Oberle, V.; Zuhorn, I. S.; Hoekstra, D., Size-dependent internalization of particles via the pathways of clathrin- and caveolae-mediated endocytosis. Biochem. J. 2004, 377, 159-169.

22. Foged, C.; Brodin, B.; Frokjaer, S.; Sundblad, A., Particle size and surface charge affect par- ticle uptake by human dendritic cells in an in vitro model. Int. J. Pharm. 2005, 298, 315-322.

23. Zhang, Y.; Yang, M.; Park, J. H.; Singelyn, J.; Ma, H.; Sailor, M. J.; Ruoslahti, E.; Ozkan, M.; Ozkan, C., A surface-charge study on cellular-uptake behavior of F3-peptide-conjugated iron oxide nanoparticles. Small 2009, 5, 1990-1996.

24. Mahmoud, K. A.; Mena, J. A.; Male, K. B.; Hrapovic, S.; Kamen, A.; Luong, J. H., Effect of surface charge on the cellular uptake and cytotoxicity of fluorescent labeled cellulose nanocrystals. ACS Appl. Mater. Interfaces 2010, 2, 2924-2932.

25. Vasir, J. K.; Labhasetwar, V., Quantification of the force of nanoparticle-cell membrane in- teractions and its influence on intracellular trafficking of nanoparticles. Biomaterials 2008, 29, 4244- 4252.

26. Yue, Z. G.; Wei, W.; Lv, P. P.; Yue, H.; Wang, L. Y.; Su, Z. G.; Ma, G. H., Surface charge affects cellular uptake and intracellular trafficking of chitosan-based nanoparticles. Biomacromolecules 2011, 12, 2440-2446.

(21)

27. Wilson, J. T.; Krishnamurthy, V. R.; Cui, W.; Qu, Z.; Chaikof, E. L., Noncovalent cell surface engineering with cationic graft copolymers. J. Am. Chem. Soc. 2009, 131, 18228-18229.

28. Gilleron, J.; Querbes, W.; Zeigerer, A.; Borodovsky, A.; Marsico, G.; Schubert, U.; Many- goats, K.; Seifert, S.; Andree, C.; Stoter, M.; Epstein-Barash, H.; Zhang, L.; Koteliansky, V.; Fitzgerald, K.; Fava, E.; Bickle, M.; Kalaidzidis, Y.; Akinc, A.; Maier, M.; Zerial, M., Image-based analysis of lipid nanoparticle-mediated siRNA delivery, intracellular trafficking and endosomal escape. Nat. Biotechnol.

2013, 31, 638-646.

29. Heyes, J.; Palmer, L.; Bremner, K.; MacLachlan, I., Cationic lipid saturation influences intra- cellular delivery of encapsulated nucleic acids. J. Controlled Release 2005, 107, 276-287.

30. Convertine, A. J.; Benoit, D. S.; Duvall, C. L.; Hoffman, A. S.; Stayton, P. S., Development of a novel endosomolytic diblock copolymer for siRNA delivery. J. Controlled Release 2009, 133, 221- 229.

31. Torchilin, V. P.; Rammohan, R.; Weissig, V.; Levchenko, T. S., TAT peptide on the surface of liposomes affords their efficient intracellular delivery even at low temperature and in the presence of metabolic inhibitors. Proc. Natl. Acad. Sci. U. S. A. 2001, 98, 8786-8791.

32. Brock, R., The Uptake of Arginine-Rich Cell-Penetrating Peptides: Putting the Puzzle To- gether. Bioconju. Chem. 2014, 25, 863-868.

33. Tunnemann, G.; Martin, R. M.; Haupt, S.; Patsch, C.; Edenhofer, F.; Cardoso, M. C., Car- go-dependent mode of uptake and bioavailability of TAT-containing proteins and peptides in living cells. FASAB. J. 2006, 20, 1775-1784.

34. Karagiannis, E. D.; Urbanska, A. M.; Sahay, G.; Pelet, J. M.; Jhunjhunwala, S.; Langer, R.;

Anderson, D. G., Rational design of a biomimetic cell penetrating peptide library. ACS Nano 2013, 7, 8616-8626.

35. Jiang, Y.; Tang, R.; Duncan, B.; Jiang, Z.; Yan, B.; Mout, R.; Rotello, V. M., Direct cytosolic delivery of siRNA using nanoparticle-stabilized nanocapsules. Angew. Chem. Int. Ed. 2015, 54, 506- 510.

36. Nakase, I.; Kobayashi, S.; Futaki, S., Endosome-disruptive peptides for improving cytosolic delivery of bioactive macromolecules. Biopolymers 2010, 94, 763-770.

37. Ding, Y.; Jiang, Z.; Saha, K.; Kim, C. S.; Kim, S. T.; Landis, R. F.; Rotello, V. M., Gold nan- oparticles for nucleic acid delivery. Mol. Ther. 2014, 22, 1075-1083.

38. Tang, R.; Kim, C. S.; Solfiell, D. J.; Rana, S.; Mout, R.; Velazquez-Delgado, E. M.; Chom- poosor, A.; Jeong, Y.; Yan, B.; Zhu, Z. J.; Kim, C.; Hardy, J. A.; Rotello, V. M., Direct delivery of functional proteins and enzymes to the cytosol using nanoparticle-stabilized nanocapsules. ACS Nano 2013, 7, 6667-6673.

(22)

1

39. van Meer, G.; Voelker, D. R.; Feigenson, G. W., Membrane lipids: where they are and how they behave. Nat. Rev. Mol. Cell Biol. 2008, 9, 112-124.

40. Kostakioti, M.; Newman, C. L.; Thanassi, D. G.; Stathopoulos, C., Mechanisms of protein export across the bacterial outer membrane. J. Bacteriol 2005, 187, 4306-4314.

41. Akhtar, J.; Shukla, D., Viral entry mechanisms: cellular and viral mediators of herpes simplex virus entry. FEBS. J. 2009, 276, 7228-7236.

(23)
(24)

2

Drug Delivery via Cell Membrane Fusion Using Lipopeptide Modified Liposomes

2 Chapter 2

23

Drug Delivery via Cell Membrane Fusion Using Lipopeptide

Modified Liposomes

Jian Yang, Azadeh Bahreman, Geert Daudey, Jeroen Bussmann, René C.L. Olsthoorn and Alexander Kros

Chapter 2 has been published in ACS Central Science. 2016 ACS Cent. Sci, 2016, 2, 621-630; DOI: 10.1021/acscentsci.1026b00172.

Supplementary Tables are available online.

(25)

ABSTRACT

Efficient delivery of drugs to living cells is still a major challenge. Currently, most methods rely on the endocytotic pathway resulting in low delivery efficiency due to limited endosomal escape and/or degradation in lysosomes. Here, we report a new method for direct drug delivery into the cytosol of live cells in vitro and vivo utilizing targeted membrane fusion between liposomes and live cells. A pair of complementary coiled-coil lipopeptides was embedded in the lipid bilayer of liposomes and cell membranes respectively, resulting in targeted membrane fusion with concomitant release of liposome encapsulated cargo including fluorescent dyes and the cytotoxic drug doxorubicin. Using a wide spectrum of endocytosis inhibitors and endosome trackers we demonstrate that the major site of cargo release is at the plasma membrane. This method thus allows for the quick and efficient delivery of drugs and is expected to have many in-vitro, ex-vivo and in-vivo applications.

(26)

2

INTRODUCTION

The plasma membrane is the protecting interface between cells and their surrounding environment. Uptake of nutrients occurs through this interface using specialized mechanisms such as endocytosis.1 Nutrients, or drugs for that matter, are frequently internalized into small transport vesicles called endosomes, which are derived from the cell membrane. For many medicines to become an active drug, they have to enter the cell’s cytosol. However, the detrimental environment inside these endosomes can result in degradation of the drug. To date, intracellular delivery of macromolecules is still a major challenge in research and therapeutic applications.2, 3 It is therefore highly desirable to develop new alternative delivery methods that circumvent the endocytosis pathway. So far, all attempts in drug delivery using particles as carriers have been unsuccessful in avoiding this pathway,4 hence current efforts to develop ways of enhancing endosomal escape.5 Cell penetrating peptides (CPP) have been studied extensively to achieve efficient uptake into the cytosol. However, the current view is that CPPs conjugated to large molecular weight cargo (e.g. liposomes) predominantly are internalized via endocytosis.6-8 Moreover, the positive charge of CPPs such as the Tat peptide9 leads to unfavorable interaction with blood components. Other transfection techniques have been devised, such as viral vectors10 and physical methods.2, 11, 12 These methods have their own limitations, including safety issues or their reliance to electrical fields or high pressure.

Fusion of lipid membranes is a vital process in biological systems, facilitating the efficient transport of molecules across membranes.13-15 In vivo membrane fusion shows a broad variety, from synaptic to viral and extracellular fusion and was found to be a highly regulated process, specific in time and place, which is achieved by a complex interplay of different functional proteins.16,17

As a bottom-up approach, several synthetic models systems have been developed to mimic membrane fusion events, but in general these simple systems do not always recapitulate the basic characteristics of native membrane fusion.18-22 Furthermore, all these approaches were limited to liposome-liposome fusion studies and have not shown to induce fusion events in live cells, thereby limiting their use for future drug delivery purposes.

Inspired by the SNARE protein complex, our laboratory has developed a fully artificial membrane fusion system composed of a complementary pair of lipidated coiled coil peptides enabling targeted liposome-liposome fusion.23 This model system possesses all the key characteristics of targeted membrane fusion similar to SNARE mediated fusion including lipid and content mixing in the absence of leakage (Figure 1A-B).24, 25 In our membrane fusion system, coiled-coil forming peptides

”E3” [(EIAALEK)3] and “K3” [(KIAALKE)3] were conjugated to a cholesterol moiety via a polyethylene glycol (PEG) spacer, yielding lipopeptides CPE3 and CPK3. The

(27)

cholesterol moiety allows for the immediate insertion of the lipidated peptides into any phospholipid membrane. We demonstrated that plain membranes could become fusogenic by the spontaneous insertion of CPE3 and CPK3 in the bilayer. A follow-up study showed that CPK3 modified cells and zebrafish embryos could be specifically labeled with the complementary fluorescently-labeled E3 peptide,23 revealing that E3/K3 coiled-coil formation is also functional in an in vivo environment thereby paving the way for targeted delivery using peptide modified liposomes.

Here, we report a new drug delivery method based on targeted membrane fusion between liposomes and live cells. We demonstrate that a wide range of cell lines can be specifically modified with lipopeptide CPK4 and upon addition of CPE4 decorated liposomes membrane fusion occurs with concomitant efficient cytosolic delivery of a variety of compounds like fluorescent dyes propidium iodide (PI), TOPRO3, and the cytotoxic drug doxorubicin (DOX). The mechanism of content uptake was studied using endocytosis inhibitors and endosome trackers in order to prove that the major site of cargo release into the cells is indeed at the plasma membrane due to liposome-cell fusion. Additionally, we show cytosolic dye (and drug) delivery in vivo using zebrafish embryos. Our method thus allows for quick and efficient delivery of drugs and bio(macromolecules) without cell damage and is expected to have many applications in vitro, ex vivo and in vivo.

Results and Discussion

Coiled-Coil Formation between CPE4 and CPK4

Previously, we reported docking of liposomes at cell membranes using peptides CPE3 and CPK3, but membrane fusion was not observed.23 In the present study we increased the number of heptad repeats in CPE and CPK to four thereby enhancing coiled-coil stability,26 expecting that this would favor liposome-cell fusion. Figure 1C shows that the cholesterol- and PEG-modified E4 and K4 – hereafter called lipopeptides CPE4 and CPK4 - when attached to liposomes, are capable of coiled-coil formation as evident from circular dichroism (CD) spectroscopy, in agreement with previous experiments using CPE3 and CPK3. Next, lipid mixing experiments were performed to investigate the fusogenicity of the CPE4/CPK4 pair in a liposome-liposome assay. In these experiments a fluorescence resonance energy transfer (FRET)-pair consisting of nitrobenzoxadiazole (NBD) and lissamine rhodamine (LR) fluorophore labeled lipids was incorporated into the membrane of CPK-decorated liposomes.21 Upon lipid

(28)

2

mixing of the latter liposomes with CPE4-liposomes the distance between NBD and LR increased, resulting in increased NBD-fluorescence as shown in Figure 1D. Content mixing was quantified by incorporating a sulforhodamine B at a self-quenching concentration of 20 mM into CPE-decorated liposomes and mixing these with CPK-liposomes as described.23 The increase in sulforhodamine B fluorescence over time indicated that full fusion took place between CPE4 and CPK4-liposomes (Figure 1D). Control experiments verified that the increase in sulforhodamine B fluorescence was not caused by leakage during fusion (Figure S1).

Coiled-Coil Formation Triggers Liposome-Cell Fusion

Next we investigated whether CPE4 and CPK4 could also mediate membrane fusion between liposomes and living cells. To this end, HeLa cells were pre-incu- bated with a micellar solution of CPK4 for 0.5-2 h before CPE4-decorated liposomes (lipid composition DOPC:DOPE:CH, 50:25:25 mol%) containing the nucleic acid stain propidium iodide (PI) or TOPRO3 in their aqueous interior were added as schematically shown in Figure 1E. In order to localize the lipid bilayer, these liposomes also contained 1 mol% of green-fluorescent NBD-DOPE lipids. As expected, confocal microscopy showed that cell membranes became labeled with the green NBD-dye on their outside in line with previous studies23. Strikingly, the red dye was observed in the cytosol and nucleus, indicating that membrane fusion and content release had occurred (Figure 2A & Figure S2A for TOPRO3). Control experiments in which one of the two lipopeptides was omitted showed neither uptake of PI or TOPRO3 nor NBD-labeling of the cell plasma membrane (Figure 2B, 2C, 2E, and Figure S2). We note that when CPK-treated cells were incubated with empty CPE4-decorated liposomes in the presence of free dye only a weak fluorescent signal was observed inside cells (Figure 2C & Figure S2C). This control experiment rules out the possibility that residual non-encapsulated dye in our liposome preparation entered the cell by transient membrane destabilization during fusion events. Finally cell incubation with free dyes also did not show any signal of the dye inside the cells (Figure 2F &Figure S2F). Similar to CPE4 decorated liposomes, we also used CPK4 decorated liposomes containing PI and incubated these with CPE4 pre-treated HeLa cells. However, the delivery of PI was less efficient. A reason might be the asymmetric nature of the fusion system. It was recently shown that peptide E does not interact with a membrane. In contrast, peptide K does interact with the membrane in a so-called snorkeling mode, and this peptide-mem- brane interaction is in equilibrium with either peptide K homocoiling or E/K coiled coil formation.27, 28 These studies suggest that peptide K-membrane interactions result in increased membrane curvature supporting membrane fusion. A cell membrane is more complex in composition and therefore less susceptible to undergo fusion as compared to the fusogenic liposomes (DOPC:DOPE:CH 2:1:1) used in this study.

(29)

Figure 1. Schematic representation of (A) coiled-coil structure between peptides E and K (adapted from PDB 1UOI), (B) Targeted liposome fusion mediated by coiled-coil formation between CPE4 modified liposomes and CPK4 modified liposomes, (C) CD spectra of CPE4 modified liposomes, and CPK4 modified liposomes and a equimolar mixture thereof. The total lipid concentrations were 0.5 mM with 1mol% of ipidated peptide in PBS. (D) lipid mixing and content mixing between CPE4-liposomes and CPK4-lipos- omes. Fluorescence traces showing lipid mixing between E and K decorated liposomes, as measured through an increase in NBD fluorescence. Total lipid concentrations were 0.1 mM with 1mol% of lipidi- dated peptide, in PBS; Fluorescence graphs indicating content mixing between sulphorhodamine loaded (20 mM), CPE4 decorated liposomes and non-fluorescent, CPK4 decorated liposomes. Total lipid con- centrations were 0.25 mM with 1 mol% lipidated peptide in PBS. (E) Scheme of fusion between cell and liposomes.

Our current thought is that peptide K needs to be on the cell membrane prior to a fusion event in order to activate the complex cell membrane by inducing membrane curvature.29 However, more studies are required to support this hypothesis. To ex- clude the possibility that peptidemediated liposomal dye delivery was a peculiarity of HeLa cells, the membrane fusion experiments were repeated with Chinese hamster ovary (CHO) and mouse fibroblast (NIH/3T3) cell lines. Again the appearance of TOPRO3 and PI was observed inside cells suggesting that the peptide-mediated delivery of the dye is cell type independent (Figure S3 & S4). Importantly, we found that uveal melanoma cells (Mel270), which are generally hard to transfect,30, 31 could also be modified with TOPRO3 using this method (Figure S3D).

(30)

2

Figure 2: Delivery of PI by peptidated-liposomes is dependent on coiled-coil formation between CPK and CPE. Confocal microscopy images of Hela cells. Cells were pre-incubated with CPK (A, B, C) or medium (D, E, F) for 2 hours, followed by treatment with: CPE-decorated liposomes containing PI (a,e), liposomes containing PI (B,D), CPE-decorated liposomes plus free PI (C), or free PI(F). Green: NBD, Red: PI. Scale bar is 25 µm. Overlay is red and green channel plus bright field image.

To address the potential toxicity of CPK4, CPE4, and liposomes towards CHO, NIH/3T3 and HeLa cells cell viability assays were carried out. These assays indicated that lipopeptides CPE4 and CPK4 and liposomes, with or without CPE4, at the concentrations used throughout this study are well tolerated by all cell lines (Fig- ure S5A). Higher concentrations of these lipopeptides, even up to 100 µM, did not significantly reduce cell viability when exposed for 2 hours but only did so after 24 hours of exposure (Figure S5B & S5C).Altogether, these results show that coiled-coil formation between CPK4 and CPE4 is critical for fusion and release of the dyes, and that these compounds are not toxic for living cells at the concentrations used allowing to investigate potential applications and their uptake mechanism.

Delivery of Doxorubicin

Doxorubicin (DOX) is one of the mostly used drugs for cancer treatments in the clinic today but as a free drug has serious cardiotoxicity. DOX is a cell permeable drug whose fluorescence is strongly enhanced upon binding to nucleic acids.

Intercalation into DNA ultimately results in apoptosis.32 To test delivery of liposo-

(31)

mal DOX, HeLa cells were pre-incubated with CPK4 and subsequently exposed to CPE4-decorated liposomes containing 5 µM DOX for 15 min. As can be seen in Figure 3A and 3B this resulted in strong nuclear (and cytosolic) fluorescence. Control experiments showed that DOX delivery is highly dependent on the presence of CPE4 and CPK4 (Figure S6). To investigate cytotoxicity of liposomal delivered DOX, HeLa cells pre-incubated with CPK were exposed with increasing concentrations of DOX-loaded liposomes for 12 h. Cell viability was measured 24 h later. Figure 3C shows cell viability as a function of liposomal and free DOX. As expected, very low concentrations of free DOX (< 1 µM) did not affect the viability of HeLa cells as passive crossing into cells is not efficient at this concentration. Importantly, in current treatments in the clinic the DOX concentration is up to 9 µM in the serum of patients.

In contrast, liposomes loaded with 1 µM DOX did showed a significant effect as the DOX uptake is significantly enhanced. Liposomally delivered DOX reduced cell viability at DOX concentrations as low as 0.1 nM with an IC50 of ~0.01 µM, while free DOX did not affect cell viability at concentrations up to 1 µM (IC50 ~5 µM).

Control experiments in which either CPK4 or CPE4 was omitted showed 100-fold or higher IC50 values (Figure S7). Thus, our peptide-mediated delivery of DOX can potentially reduce the dose of DOX needed for anticancer treatments thereby lowering the cardiotoxicity of DOX.33 The presented fusion mediated delivery approach is also promising for the delivery of other drugs or biomolecules like DNA or siRNA.

0.01 0.1 1 10 100

0 20 40 60 80 100

120 CPK + CPE-lipo-DOX

Free DOX

Cell Viability (%)

DOX (µM)

A) B) C)

Figure 3. Delivery of DOX into HeLa cells. (A) CPE4/CPK4 mediated delivery of DOX into HeLa cells.

Cells were treated with CPK4 for 1 h followed by incubation with 0.25 mM CPE4-liposomes containing with DOX for 15 min. Images were taken after washing. A) bright field. (B) fluorescence channel. The inset shows a magnified overlay image, revealing the presence of DOX in the nucleus. The concentration of DOX loaded into liposomes is 5 µM. Scale bar represents 25 µm. (C) Cytotoxicity of CPE4/CPK4 delivered DOX and free DOX. HeLa cells were treated with CPK4 for 1 h and series of concentrations of CPE4 decorated liposomes containing DOX (blue line), or the same concentrations of free DOX (red line) for 12 h. After washing and incubation with medium for 24 h, cell viability was measured by a WST-1 assay.

(32)

2

Liposomes and Content Only Partially Co-localize with Endosomes

Endocytosis is the most common pathway for the uptake of small particles including liposomes by cells.4 To investigate whether endocytosis played a role in the liposomal delivery, the endosome tracker pHrodo, a fluorescently labeled dextran, was used in combination with TOPRO3 loaded liposomes. TOPRO3 was chosen as encapsulated dye for this experiment instead of PI because its emission (Ex/Em 642/661 nm) is expected not to interfere with emission of pHrodo (Ex/Em 560/585 nm) making investigation of colocalization of dyes easier. pHrodo and CPE-

4decorated liposomes containing 1 mol% NBD-DOPE and TOPRO3 were simul- taneously added to CPK4-modified HeLa cells. Confocal microscopy showed the presence of TOPRO3 in the cytosol and to a lesser extent in the nucleus (Figure 4B) while pHrodo was mainly observed as individual dots in the cytosol in agree- ment with its endosomal uptake (Figure 4C). Overlaying the fluorescent images of TOPRO3 and pHrodo revealed some overlap between TOPRO3 and endo- somes (Figure 4E, pink dots) but the majority of TOPRO3 signal remains unmixed.

Again, the signal from NBD-DOPE (Figure 4A, white dots) remained at the plasma membrane, although some overlap with pHrodo was observed at the plasma membrane (Figure 4F).

Figure 4. Visualization of endosomes using an endosome tracker. CHO cells were treated with CPK4

for 2 h, followed by co-incubation with pHrodo red dextran and CPE4-decorated liposomes (0.25 mM total lipid concentration and 1 mol% CPE4 ) loaded with TOPRO 3. (A) White channel showing DOPE-NBD liposomes. (B) Red channel (TOPRO3). (C) Blue channel (pHrodo). (D) Overlay of panels A and B. (E) Overlay of panels B and C. (F) Overlay of panels A and C. Scale bar is 25 μm.

(33)

This could be the result of both liposomes and endosome tracker binding at a common spot at the plasma membrane or could mean that some liposomes are initially taken up by endocytosis but then rapidly fuse with the endosomal mem- brane.These results suggest that the endosomal uptake pathway only plays a mi- nor role in CPE4-CPK4 mediated liposomal uptake and that liposome-cell membrane fusion is the main route for cargo delivery. This is also illustrated by performing the same experiment at 4ºC, conditions under which active uptake by endocytosis is inhibited. Imaging of cells over a period of three hours showed the increasing uptake of TOPRO3 (Figure 5A, upper panels). In contrast only a faint signal of endosome tracker pHrodo was observed after three hours indicating that endocytosis was severely limited at 4ºC (Figure 5A, lower panels). Quantification of the fluorescence intensity using software (Image-J) showed that after 3 h the uptake of TOPRO3 reached ~80% of the level obtained after 30 min at 37ºC (Figure 5B). The slower uptake is presumably caused by the reduced rate of liposome-cell fusion events at 4ºC. This is supported by the observation that liposome-liposome lipid mixing induced by CPE4/CPK4 is also significantly slower at 4ºC than at room temperature (Figure S8).

Endocytosis and Macropinocytosis Inhibitors Marginally Affect Delivery

As independent support for our conclusion that fusion at the plasma membrane is the major pathway for our liposome-based delivery system, several well characterized inhibitors of endocytotic pathways were tested using flow cytometry measurements and confocal microscopy imaging. Wortmannin blocks PI3-kinase and inhibits macropinocytosis,34-37 Chlorpromazine interferes with clathrin-dependent endocytosis,38-40Genistein inhibits tyrosine-phosphorylation of Cav 1 and caveo- la-dependent endocytosis.41-43 In addition, nocodazole, an inhibitor of microtubule formation, was used to investigate whether intracellular trafficking and internaliza- tion mechanisms are involved.36, 37, 40, 44, 45 Moreover, endocytosis of nanoparticles is an energy-dependent mechanism. Sodium azide was therefore used to deplete the energy needs for endocytosis and restrict metabolic activity.46, 47

HeLa cells were first incubated for 1 h with each inhibitor at concentrations that have been reported by others to show optimal activity. After removal of the inhibi- tors, cells were treated with CPK4 and subsequently with CPE4-decorated liposomes containing PI dye in the presence of freshly added inhibitors. FACS analysis showed that genistein and nocodazole had no adverse effect on the delivery of PI (Figure 5C), whereas in the presence of wortmannin, chlorpromazine and sodium azide PI uptake was reduced less than 20%. These results argue against a major role of

(34)

2

endocytosis or pinocytosis in uptake of liposomal cargo and support that the dominant pathway for delivery is indeed targeted membrane fusion between liposomes with the plasma membrane of live cells.

Figure 5. Investigation into the uptake mechanism. (A) Effect of low temperature incubation of HeLa cells on liposomal delivery of TOPRO3 and endosomal uptake of pHrodo. Cells were pre-incubated on ice with 5 μM CPK (2h), followed by 15 min incubation with 0.25 mM CPE-decorated liposomes contain- ing TOPRO3. After three washes confocal images were taken immediately (0 min) and after 60 min, 120 min and 180 min. Top row: TOPRO3 (red), bottom row: pHrodo (blue). (B) Graphical representation of the percentage of TOPRO dye uptake by HeLa cells on ice. Fluorescence intensities were calculated by Image J and plotted as a percentage relative to the fluorescence of TOPRO3 delivery at 37˚C (100%).

Scale bar is 25 μm. (C) Effect of endocytosis and macropinocytosis inhibitors on delivery of PI by liposomes to HeLa cells. Cells were incubated with medium (Ctrl+), or medium containing 0.25 µM wortmannin (Wor), 40 µM chlorpromazine (Chl), 200 µM genistein (Gen), 40 µM nocodazole (Noc) for 1 hour, 0.01% w/v sodium azide (NaN3), followed by 2 hour incubation with 5 µM CPK in presence of inhibitors, and then treated for 15 min with CPE-liposomes containing PI. Final concentration of lipids (liposomes) was 0.25 mM. Cellular uptake was measured by flow cytometry. Positive control (100%):

fluorescence of PI dye in the absence of inhibitors.

Intracellular Delivery in Vivo

As a first step towards clinical application, we used zebrafish embryos to evaluate direct cytoplasmic delivery in vivo. We previously established coiled-coil mediated docking of liposomes onto the zebrafish embryonic skin.21 During embryonic stages, the zebrafish skin is composed of a layer of ridged, mucus-covered enveloping layer (EVL) cells. Through interspersed gaps in the EVL layer, cells within the underlying epidermal basal layer (EBL), including mucus-secreting cells and ionocytes, are exposed to the external environment45. To test for in vivo de- livery to skin epithelial cells, we exposed 48h-old zebrafish embryos to CPK in embryo medium for 30 minutes. After washing, embryos were exposed to NBD-la- beled, CPE4-decorated liposomes containing DOX for 30 minutes. Consistent with previous results,21 we observed widespread liposome docking after 30 min of incubation, as evidenced by NBD and DOX co-labeling. Importantly, we identified nuclear DOX labeling within a subset of skin epithelial cells (Figure 6) consistent with delivery into EBL-layer, but not EVL layer cells, which appeared to be inaccessible due to mucus covering or membrane ridging. Control

(35)

experiments established that cytoplasmic delivery was specific to coiled-coil interaction (Figure S9).

Figure 6. In vivo delivery of DOX using CPK4 and CPE4. 2 dpf zebrafish were treated with CPK4 for 30 minutes, followed by 30 minutes incubation with CPE4-decorated liposomes (0.25 mM total lipid con- centration and 1 mol% CPE4) loaded with DOX. (A,B) Whole-embryo imaging showing widespread DOX delivery in living zebrafish embryos (control experiments in Supplementary Figure S9). (C-E) Single ze- brafish skin epithelial cell (from the indicated site of the embryo in (A,B) displaying membrane associated DOPE-NBD labeling (NBD) and predominantly nuclear DOX labeling.

We further confirmed intracellular delivery using liposomes loaded with PI, which becomes highly fluorescent only after interaction with cellular DNA or RNA (Figure S10 & S11). Together, these results indicate the potential application of coiled-coiled induced membrane fusion for direct cellular drug delivery in vivo.

CONCLUSION

Numerous methods exist to deliver drugs and (bio)macromolecules to living cells. Depending on the nature of these molecules they can be deliv- ered into cells via electroporation, micro-injection, calcium phosphate co-pre- cipitation, nanoparticles, or viral particles. However many of these methods are either not suitable for in vitro use or cannot be safely applied in in vivo applica- tions, or are inefficient due to endosomal entrapment and degradation. The mem- brane fusion system described here involves the targeted fusion of liposomes with the plasma membrane of live cells. As a result, endosomal pathways are al- most completely circumvented and therefore this efficient drug delivery meth- od is suited for labile (bio)molecules. In addition, the lipopeptides and modified liposomes have a low toxicity at the used concentration - in contrast to CPP-based delivery approaches or PEG-induced liposome fusion48. We anticipate that this membrane fusion strategy will spark new in vitro, ex vivo in the field of chemical biology and possibly in the long-term in vivo applications enabling new basic and applied research studies for gene therapy. Moreover any compound that can be encapsulated in liposomes like hydrophilic low molecular weight drugs49 or DNA/

siRNA50, 51 could be considered as well as many hydrophilic drugs are unable to enter

(36)

2

cells effectively and are known to be degraded in a lysosomal environment thereby lowering their therapeutic efficacy.52

Here, fusion mediated delivery could result in less degradation of sensitive mole- cules and might therefore find use as a new transfection agent in in vitro cell studies.

Also lipid bilayer-coated nanoparticles53-57 might be delivered more efficiently when coiled coil mediated membrane fusion is applied thereby increasing the scope of molecules and nanoparticles/nanomdeicines that can be delivered into cells. Future in vivo application of this technique requires cells to be pre-modified with one of the two peptides and is currently not cell-type specific due to the cholesterol-anchor, several applications are still conceivable. These include topical administration of drugs to treat e.g. pulmonary disease or combat respiratory infections like influenza. On the other hand delivery of liposomally encapsulated mRNA or DNA coding for the tumor suppressor p53 will only affect tumor cells and leave healthy cells unharmed.58 Similarly, liposomal delivery of miRNA or siRNA to upregulate tumor suppressors or downregulate oncogenes could selectively kill only tumor cells.59

Finally, a certain degree of selectivity can be achieved using a light-induced membrane fusion system that was recently developed in our laboratory. This system makes use of photoinduced deshielding of a PEGylated CPE and thus allows potentially for spatiotemporal control of liposomal drug delivery in vivo.60

EXPERIMENTAL PROCEDURES Materials and Methods

Fmoc-protected amino acids were purchased from Novabiochem and Biosolve Sie- ber Amide resin was purchased from Chem-Impex International and Agilent Tech- nologies. DOPE, DOPC, DOPE-NBD, and DOPE-LR were purchased from Avanti Polar Lipids. cholesterol, propidium iodide (#BCBM1455V) and sulphorhodamine were obtained from Sigma-Aldrich. Topro3-Iodide (#1301286), pHrodo™ Red dex- tran 10,000MW were purchased from Life Technologies. 8 wells slide Lab-tek was purchased from Thermo Scientific, USA. DMEM medium was obtained from Gibco, life technologies. N3-PEG4-COOH61 and 3-Azido-5-cholestene62 were synthesized following literature procedures.

Referenties

GERELATEERDE DOCUMENTEN

Coiled-coil lipopeptides, CPK 4 /CPE 4 lead to the membrane fusion between liposomes and cell membranes, resulting in direct cytosolic delivery of encapsulated drugs.. During

The module isomorphism problem can be formulated as follows: design a deterministic algorithm that, given a ring R and two left R-modules M and N , decides in polynomial time

The handle http://hdl.handle.net/1887/40676 holds various files of this Leiden University dissertation.. Algorithms for finite rings |

Professeur Universiteit Leiden Directeur BELABAS, Karim Professeur Universit´ e de Bordeaux Directeur KRICK, Teresa Professeur Universidad de Buenos Aires Rapporteur TAELMAN,

We are interested in deterministic polynomial-time algorithms that produce ap- proximations of the Jacobson radical of a finite ring and have the additional property that, when run

The handle http://hdl.handle.net/1887/40676 holds various files of this Leiden University

Analyses of strategy use (Fagginger Auer et al., 2013; Hickendorff et al., 2009) showed that from 1997 to 2004, the use of digit-based algorithms for multidigit multiplication

A total of 39 questions were selected from this question- naire (see the Appendix) that were either relevant to the mathematics lessons in general (teacher characteristics,