Mechanistic evaluation of the transfection barriers involved in lipid-mediated gene delivery: interplay between nanostructure and composition.

Here we present a quantitative mechanism-based investigation aimed at comparing the cell uptake, intracellular trafficking, endosomal escape and final fate of lipoplexes and lipid-protamine/deoxyribonucleic acid (DNA) (LPD) nanoparticles (NPs) in living Chinese hamster ovary (CHO) cells. As a model, two lipid formulations were used for comparison. The first formulation is made of the cationic lipid 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP) and the zwitterionic lipid dioleoylphosphocholine (DOPC), while the second mixture is made of the cationic 3β-[N-(N,N-dimethylaminoethane)-carbamoyl] cholesterol (DC-Chol) and the zwitterionic helper lipid dioleoylphosphatidylethanolamine (DOPE). Our findings indicate that lipoplexes are efficiently taken up through fluid-phase macropinocytosis, while a less efficient uptake of LPD NPs occurs through a combination of both macropinocytosis and clathrin-dependent pathways. Inside the cell, both lipoplexes and LPD NPs are actively transported towards the cell nucleus, as quantitatively addressed by spatio-temporal image correlation spectroscopy (STICS). For each lipid formulation, LPD NPs escape from endosomes more efficiently than lipoplexes. When cells were treated with DOTAP-DOPC-containing systems the majority of the DNA was trapped in the lysosome compartment, suggesting that extensive lysosomal degradation was the rate-limiting factors in DOTAP-DOPC-mediated transfection. On the other side, escape from endosomes is large for DC-Chol-DOPE-containing systems most likely due to DOPE and cholesterol-like molecules, which are able to destabilize the endosomal membrane. The lipid-dependent and structure-dependent enhancement of transfection activity suggests that DNA is delivered to the nucleus synergistically: the process requires both the membrane-fusogenic activity of the nanocarrier envelope and the employment of lipid species with intrinsic endosomal rupture ability.

[1]  E. Gratton,et al.  The role of cytoskeleton networks on lipid-mediated delivery of DNA. , 2013, Therapeutic delivery.

[2]  M. Grinstaff,et al.  Macropinocytosis is the major pathway responsible for DNA transfection in CHO cells by a charge-reversal amphiphile. , 2011, Molecular pharmaceutics.

[3]  Fengjuan Wang,et al.  The biomolecular corona is retained during nanoparticle uptake and protects the cells from the damage induced by cationic nanoparticles until degraded in the lysosomes. , 2013, Nanomedicine : nanotechnology, biology, and medicine.

[4]  K. Dawson,et al.  High-speed imaging of Rab family small GTPases reveals rare events in nanoparticle trafficking in living cells. , 2012, ACS nano.

[5]  Giulio Caracciolo,et al.  Time evolution of nanoparticle-protein corona in human plasma: relevance for targeted drug delivery. , 2013, Langmuir : the ACS journal of surfaces and colloids.

[6]  L. Thoma,et al.  Core-shell-type lipid-polymer hybrid nanoparticles as a drug delivery platform. , 2013, Nanomedicine : nanotechnology, biology, and medicine.

[7]  Philip M. Kelly,et al.  Transferrin-functionalized nanoparticles lose their targeting capabilities when a biomolecule corona adsorbs on the surface. , 2013, Nature nanotechnology.

[8]  P. Felgner,et al.  Cationic liposome-mediated transfection , 1989, Nature.

[9]  S. Futaki,et al.  Nanoparticles for ex vivo siRNA delivery to dendritic cells for cancer vaccines: programmed endosomal escape and dissociation. , 2010, Journal of controlled release : official journal of the Controlled Release Society.

[10]  E Gratton,et al.  Quantitative measurement of intracellular transport of nanocarriers by spatio-temporal image correlation spectroscopy , 2013, Methods and applications in fluorescence.

[11]  M. Schliwa,et al.  Powering membrane traffic in endocytosis and recycling , 2006, Nature Reviews Molecular Cell Biology.

[12]  I. Zuhorn,et al.  Size-dependent internalization of particles via the pathways of clathrin- and caveolae-mediated endocytosis. , 2004, The Biochemical journal.

[13]  I. Zuhorn,et al.  Gene delivery by cationic lipids: in and out of an endosome. , 2007, Biochemical Society transactions.

[14]  G. Caracciolo,et al.  Toward the rational design of lipid gene vectors: shape coupling between lipoplex and anionic cellular lipids controls the phase evolution of lipoplexes and the efficiency of DNA release. , 2009, ACS applied materials & interfaces.

[15]  Giulio Caracciolo,et al.  The protein corona effect for targeted drug delivery , 2013 .

[16]  F. Szoka,et al.  Mechanism of oligonucleotide release from cationic liposomes. , 1996, Proceedings of the National Academy of Sciences of the United States of America.

[17]  Leaf Huang,et al.  Recent advances in nonviral vectors for gene delivery. , 2012, Accounts of chemical research.

[18]  Paul W. Wiseman,et al.  Advances in Image Correlation Spectroscopy: Measuring Number Densities, Aggregation States, and Dynamics of Fluorescently labeled Macromolecules in Cells , 2007, Cell Biochemistry and Biophysics.

[19]  S. Resina,et al.  Physico-Chemical Characteristics of Lipoplexes Influence Cell Uptake Mechanisms and Transfection Efficacy , 2009, PloS one.

[20]  A. Nogales,et al.  Effect of lipid composition on the structure and theoretical phase diagrams of DC-Chol/DOPE-DNA lipoplexes. , 2010, Biomacromolecules.

[21]  E. Gratton,et al.  Intracellular trafficking of cationic liposome-DNA complexes in living cells. , 2012, Soft matter.

[22]  R. Langer,et al.  Intracellular delivery of core-shell fluorescent silica nanoparticles. , 2008, Biomaterials.

[23]  Y. Maitani,et al.  Cationic liposome (DC-Chol/DOPE=1:2) and a modified ethanol injection method to prepare liposomes, increased gene expression. , 2007, International journal of pharmaceutics.

[24]  S. Futaki,et al.  Reprint of: Nanoparticles for ex vivo siRNA delivery to dendritic cells for cancer vaccines: Programmed endosomal escape and dissociation. , 2010, Journal of controlled release : official journal of the Controlled Release Society.

[25]  M. Yeh,et al.  Clinical development of liposome-based drugs: formulation, characterization, and therapeutic efficacy , 2011, International journal of nanomedicine.

[26]  I. Zuhorn,et al.  On the Mechanism of Cationic Amphiphile-mediated Transfection. To Fuse or not to Fuse: Is that the Question? , 2002, The Journal of Membrane Biology.

[27]  Marco P Monopoli,et al.  Biomolecular coronas provide the biological identity of nanosized materials. , 2012, Nature nanotechnology.

[28]  Angelo Bifone,et al.  Transfection efficiency boost of cholesterol-containing lipoplexes. , 2012, Biochimica et biophysica acta.

[29]  Warren C. W. Chan,et al.  Understanding and Controlling the Interaction of Nanomaterials with Proteins in a Physiological Environment , 2012 .

[30]  S. Provencher CONTIN: A general purpose constrained regularization program for inverting noisy linear algebraic and integral equations , 1984 .

[31]  C. Ladavière,et al.  An overview of lipid membrane supported by colloidal particles. , 2007, Advances in colloid and interface science.

[32]  Giulio Caracciolo,et al.  Surface adsorption of protein corona controls the cell internalization mechanism of DC-Chol-DOPE/DNA lipoplexes in serum. , 2010, Biochimica et biophysica acta.

[33]  M. Montani,et al.  Role of temperature-independent lipoplex–cell membrane interactions in the efficiency boost of multicomponent lipoplexes , 2011, Cancer Gene Therapy.

[34]  R. Macdonald,et al.  An intracellular lamellar–nonlamellar phase transition rationalizes the superior performance of some cationic lipid transfection agents , 2006, Proceedings of the National Academy of Sciences.

[35]  G. Caracciolo,et al.  Tailoring lipoplex composition to the lipid composition of plasma membrane: a Trojan horse for cell entry? , 2010, Langmuir : the ACS journal of surfaces and colloids.

[36]  Giulio Caracciolo,et al.  Interaction of lipoplexes with anionic lipids resulting in DNA release is a two-stage process. , 2007, Langmuir : the ACS journal of surfaces and colloids.

[37]  Daniel A. Balazs,et al.  Liposomes for Use in Gene Delivery , 2010, Journal of drug delivery.

[38]  J. Betker,et al.  Cholesterol domains enhance transfection. , 2013, Therapeutic delivery.

[39]  Liangfang Zhang,et al.  LIPID–POLYMER HYBRID NANOPARTICLES: SYNTHESIS, CHARACTERIZATION AND APPLICATIONS , 2010 .

[40]  Iseult Lynch,et al.  Designing the nanoparticle-biomolecule interface for "targeting and therapeutic delivery". , 2012, Journal of controlled release : official journal of the Controlled Release Society.

[41]  Kenneth A Dawson,et al.  Nanoparticle adhesion to the cell membrane and its effect on nanoparticle uptake efficiency. , 2013, Journal of the American Chemical Society.

[42]  D. Hoekstra,et al.  Molecular Shape of the Cationic Lipid Controls the Structure of Cationic Lipid/Dioleylphosphatidylethanolamine-DNA Complexes and the Efficiency of Gene Delivery* , 2001, The Journal of Biological Chemistry.

[43]  T. Anchordoquy,et al.  The effect of cholesterol domains on PEGylated liposomal gene delivery in vitro. , 2011, Therapeutic delivery.

[44]  Kenneth A. Dawson,et al.  Role of cell cycle on the cellular uptake and dilution of nanoparticles in a cell population. , 2011, Nature nanotechnology.

[45]  Radostin Danev,et al.  Multi-layered nanoparticles for penetrating the endosome and nuclear membrane via a step-wise membrane fusion process. , 2009, Biomaterials.

[46]  Enrico Gratton,et al.  Factors determining the superior performance of lipid/DNA/protammine nanoparticles over lipoplexes. , 2011, Journal of medicinal chemistry.

[47]  Hiroyuki Mizuguchi,et al.  Quantitative and mechanism-based investigation of post-nuclear delivery events between adenovirus and lipoplex , 2007, Nucleic acids research.

[48]  H Akita,et al.  Quantitative three-dimensional analysis of the intracellular trafficking of plasmid DNA transfected by a nonviral gene delivery system using confocal laser scanning microscopy. , 2004, Molecular therapy : the journal of the American Society of Gene Therapy.

[49]  Santiago Costantino,et al.  Spatiotemporal image correlation spectroscopy (STICS) theory, verification, and application to protein velocity mapping in living CHO cells. , 2005, Biophysical journal.

[50]  Angelo Bifone,et al.  Cholesterol-dependent macropinocytosis and endosomal escape control the transfection efficiency of lipoplexes in CHO living cells. , 2012, Molecular pharmaceutics.

[51]  G. Caracciolo,et al.  Cationic liposome/DNA complexes: from structure to interactions with cellular membranes , 2012, European Biophysics Journal.

[52]  R. Spontak,et al.  Autophobicity-driven surface segregation and patterning of core-shell microgel nanoparticles. , 2008, Nano letters.

[53]  T. Anchordoquy,et al.  Effect of cholesterol nanodomains on the targeting of lipid-based gene delivery in cultured cells. , 2010, Molecular pharmaceutics.

[54]  I. Zuhorn,et al.  Mechanism of polyplex- and lipoplex-mediated delivery of nucleic acids: real-time visualization of transient membrane destabilization without endosomal lysis. , 2013, ACS nano.

[55]  M. Conese,et al.  Role of clathrin- and caveolae-mediated endocytosis in gene transfer mediated by lipo- and polyplexes. , 2005, Molecular therapy : the journal of the American Society of Gene Therapy.